US20210403444A1 - Heterocyclic derivatives for the treatment of rsv - Google Patents

Heterocyclic derivatives for the treatment of rsv Download PDF

Info

Publication number
US20210403444A1
US20210403444A1 US17/370,586 US202117370586A US2021403444A1 US 20210403444 A1 US20210403444 A1 US 20210403444A1 US 202117370586 A US202117370586 A US 202117370586A US 2021403444 A1 US2021403444 A1 US 2021403444A1
Authority
US
United States
Prior art keywords
avg
alkyl
together form
independently selected
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/370,586
Inventor
Richard K. Plemper
Eddy Lee
John Vernachio
Elyse Bourque
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Georgia State University Research Foundation Inc
Original Assignee
Georgia State University Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Georgia State University Research Foundation Inc filed Critical Georgia State University Research Foundation Inc
Priority to US17/370,586 priority Critical patent/US20210403444A1/en
Publication of US20210403444A1 publication Critical patent/US20210403444A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/101,4-Thiazines; Hydrogenated 1,4-thiazines
    • C07D279/141,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems
    • C07D279/161,4-Thiazines; Hydrogenated 1,4-thiazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus

Definitions

  • the invention relates to the use of small molecule therapeutics for the treatment of respiratory syncytial virus (RSV) and related members of the pneumovirus and paramyxovirus family such as human metapneumovirus, mumps virus, human parainfluenzaviruses, and Nipah and hendra virus.
  • RSV respiratory syncytial virus
  • related members of the pneumovirus and paramyxovirus family such as human metapneumovirus, mumps virus, human parainfluenzaviruses, and Nipah and hendra virus.
  • Respiratory syncytial virus is a member of the paramyxovirus family, which consists of mostly highly contagious nonsegmented, negative polarity RNA viruses that spread through the respiratory route.
  • RSV is a member of the order Mononegavirales, which consists of the non-segmented negative strand RNA viruses in the Families Paramyxoviridae, Pneumoviridae; Bunyaviridae, Rhabdoviridae and Filoviridae.
  • RSV of humans (often also termed RSV or HRSV) is a member of the Pneumoviridae.
  • RSV is classified into two subgroups, A and B (Mufson, M. et al., J. Gen. Virol. 66:2111-2124).
  • Other members of the Pneumovirus family include viruses such as bovine RSV (BRSV), ovine RSV (ORSV), pneumonia virus of mice (PVM), and the human metapneumoviruses amongst others.
  • family characteristics include a lipid envelope containing one or more glycoprotein species considered to be associated with attachment and entry of the host cell. Entry is considered to require a process by which the viral envelope fuses with the membrane of the host cell. Fusion of infected cells with, for example, their neighbors, can also result in the formation of fused multinucleate cells known as syncytia in some cases. The fusion process is believed to be glycoprotein mediated and is a feature shared with diverse enveloped viruses in other taxonomic groups. In the case of the pneumo- and paramyxoviruses, virions characteristically express a fusion glycoprotein (F), which mediates membrane fusion.
  • F fusion glycoprotein
  • Respiratory syncytial virus is the leading cause of acute upper and lower respiratory tract infections (LRTI) in adults, young children and infants. Although at risk populations include the hospitalized, elderly and high-risk adults, RSV is primarily considered to be a pediatric disease due to the prevalence and severity of unfavorable outcomes in infants. Acute LRTI infections are a leading cause of global childhood mortality and morbidity. Serological evidence indicates that in the western world approximately 95% of all children have been infected with RSV by the age of two and 100% of children have been exposed by the time they reach adulthood.
  • RSV disease is thus the leading cause of virus infection-induced death among children less than 1 year of age and can be life-threatening to the elderly and the immunocompromised. Reinfection with RSV can occur throughout life, but infants born prematurely, or with bronchopulmonary dysplasia or a congenital heart defect, are at highest risk of developing severe disease.
  • initial RSV infection of airway epithelia cells is followed by rapid spread from the nasopharynx to the lower airways that can affect respiratory function through excessive mucus, necrotic epithelial debris, and inflammatory cells obstructing the airways.
  • RSV is a seasonal infectious disease that generally runs from November to March/April in the Northern Hemisphere. In more tropical climates, the annual epidemics are more variable, often coinciding with the wet season. In most cases the RSV infections will only cause minor upper respiratory illness with symptoms resembling that of the common cold. However, severe infection with the virus may result in bronchiolitis or pneumonia, which may result in hospitalization or death. Further, since the immune response to RSV infection is not protective, RSV infections reoccur throughout adulthood. Annual re-infection rates in adults of 3-6% have been observed.
  • RSV infections place a significant burden on the healthcare system. This is particularly so in the case of infants such as, for example, immunodeficient infants, which on average spend twice as long in hospital as other patients with an RSV infection (7-8 days compared to 3-4 days). Hospitalization of infants with acute RSV-related bronchiolitis or RSV-related pneumonia involves supportive care management with oxygen therapy, fluids to prevent dehydration, nasal suctioning and respiratory support. There is also an economic impact associated with parents taking time away from work to care for their child.
  • RSV vaccine attempts to develop an effective RSV vaccine have been fruitless thus far, because the virus is poorly immunogenic overall and neutralizing antibody titers wane quickly after infection.
  • ribavirin has been approved for RSV treatment, it has not been widely adopted in clinical use due to efficacy and toxicity issues.
  • the humanized neutralizing antibody palivizumab is used for immunoprophylaxis of high-risk pediatric patients, but high costs prohibit broad-scale implementation.
  • Small-molecule drug-like therapeutics have high promise to provide a novel avenue towards RSV disease management and prevention. It is therefore an object of the present invention to provide new small-molecule therapeutics classes for the treatment of human patients and other hosts infected with RSV.
  • RSV can be inhibited, and RSV infection can be treated or prevented by administering to a patient in need thereof a composition containing an anti-RSV compound of Formula 1:
  • FIG. 1 includes a depiction of the virus yield of an RSV inhibitor in accordance with the invention [RSVP-172944] as a function of concentration.
  • FIG. 2 includes a depiction of the RdRp activity of an RSV inhibitor in accordance with the invention [RSVP-172944] as a function of concentration.
  • FIG. 3 includes a depiction of the activity of an RSV inhibitor in accordance with the invention [RSVP-172944] in tabular format.
  • the word “comprise” and variations of the word, such as “comprising” and “comprises,” means “including but not limited to,” and is not intended to exclude, for example, other additives, components, integers or steps.
  • “Exemplary” means “an example of” and is not intended to convey an indication of a preferred or ideal embodiment. “Such as” is not used in a restrictive sense, but for explanatory purposes.
  • a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer, diastereomer, and meso compound, and a mixture of isomers, such as a racemic or scalemic mixture.
  • alkyl as used herein is a branched or unbranched hydrocarbon group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, and the like.
  • the alkyl group can also be substituted or unsubstituted. Unless stated otherwise, the term “alkyl” contemplates both substituted and unsubstituted alkyl groups.
  • the alkyl group can be substituted with one or more groups including, but not limited to, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein.
  • An alkyl group which contains no double or triple carbon-carbon bonds is designated a saturated alkyl group, whereas an alkyl group having one or more such bonds is designated an unsaturated alkyl group.
  • Unsaturated alkyl groups having a double bond can be designated alkenyl groups, and unsaturated alkyl groups having a triple bond can be designated alkynyl groups. Unless specified to the contrary, the term alkyl embraces both saturated and unsaturated groups.
  • cycloalkyl as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms.
  • examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.
  • heterocycloalkyl is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, selenium or phosphorus.
  • the cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted.
  • cycloalkyl and heterocycloalkyl contemplate both substituted and unsubstituted cyloalkyl and heterocycloalkyl groups.
  • the cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein.
  • a cycloalkyl group which contains no double or triple carbon-carbon bonds is designated a saturated cycloalkyl group, whereas an cycloalkyl group having one or more such bonds (yet is still not aromatic) is designated an unsaturated cycloalkyl group.
  • alkyl embraces both saturated and unsaturated groups.
  • aryl as used herein is an aromatic ring composed of carbon atoms. Examples of aryl groups include, but are not limited to, phenyl and naphthyl, etc.
  • heteroaryl is an aryl group as defined above where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, selenium or phosphorus.
  • the aryl group and heteroaryl group can be substituted or unsubstituted. Unless stated otherwise, the terms “aryl” and “heteroaryl” contemplate both substituted and unsubstituted aryl and heteroaryl groups.
  • the aryl group and heteroaryl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein.
  • heteroaryl and heterocyclyl rings include: benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyL cirrnolinyl, decahydroquinolinyl, 2H,6H ⁇ 1,5,2-dithiazinyl, dihydrofuro[2,3 b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, IH-indazolyl, indolenyl, indolinyl, indolizinyl,
  • alkoxy has the aforementioned meanings for alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, further providing said group is connected via an oxygen atom.
  • the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described below.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms, such as nitrogen can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms.
  • substitution or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • a substituent that is said to be “substituted” is meant that the substituent is substituted with one or more of the following: alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein.
  • groups that are said to be substituted are substituted with a protic group, which is a group that can be protonated or deprotonated, depending on the pH.
  • patient refers to any mammalian organism, including but not limited to, humans.
  • salts are salts that retain the desired biological activity of the parent compound and do not impart undesirable toxicological effects.
  • examples of such salts are acid addition salts formed with inorganic acids, for example, hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids and the like; salts formed with organic acids such as acetic, oxalic, tartaric, succinic, maleic, fumaric, gluconic, citric, malic, methanesulfonic, p-toluenesulfonic, napthalenesulfonic, and polygalacturonic acids, and the like; salts formed from elemental anions such as chloride, bromide, and iodide; salts formed from metal hydroxides, for example, sodium hydroxide, potassium hydroxide, calcium hydroxide, lithium hydroxide, and magnesium hydroxide; salts formed from metal carbonates, for example, sodium carbonate, potassium carbonate, calcium carbonate, and magnesium carbonate; salts formed from metal bi
  • Pharmaceutically acceptable and non-pharmaceutically acceptable salts may be prepared using procedures well known in the art, for example, by reacting a sufficiently basic compound such as an amine with a suitable acid comprising a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid comprising a physiologically acceptable anion.
  • Alkali metal for example, sodium, potassium, or lithium
  • alkaline earth metal for example, calcium
  • RSV inhibiting compound has the structure of Formula I:
  • olefin includes unsubstituted methylene (e.g., ⁇ CH 2 ), as well as substituted groups including the functional groups falling with the definitions of R 3 , R 4 , R 5 , or R 6 .
  • imine includes the primary imine (e.g., ⁇ NH) as well as substituted imines including the functional groups falling with the definitions of R 7 .
  • RSV inhibiting compound can have the structure of Formula 1a
  • R 1 -R 6 , y, and z have the meanings given for the compound of Formula 1.
  • the R 3 and R 4 together form a ring, preferably an aryl or heteroaryl ring. In some instance, the R 3 and R 4 can together form a phenyl ring, e.g., the RSV inhibiting compound can have the structure of Formula 1 b or 1 b-i:
  • R 1 , R 2 , X, y and z are as defined above, a is selected from 0, 1, 2, 3 and 4, and R 8 includes —R a , —OR a , —N(R a ) 2 , —SR a , —SO 2 R a , —SO 2 N(R a ) 2 ; —C(O)R a , OC(O)R a , —COOR a , —C(O)N(R a ) 2 , —OC(O)N(R a ) 2 , —N(R a )C(O), —N(R a )C(O)N(R a ) 2 , —F, —Cl, —Br, —I, —CN, —NO 2 .
  • each R 8 group is selected independently from the list provided above. In some embodiments, it is preferred that a is 0.
  • z is 0 and y is 1:
  • R 1 -R 8 , X, and a have the meanings given above.
  • R 1 is defined as N(R a ) 2 .
  • N(R a ) 2 is a group selected from:
  • R 9 is independently selected from —R a , —OR a , —N(R a ) 2 , —SR a , —SO 2 R a , —SO 2 N(R a ) 2 , —C(O)R a , OC(O)R a , —COOR a , —C(O)N(R a ) 2 , —OC(O)N(R a ) 2 , —N(R a )C(O), —N(R a )C(O)N(R a ) 2 , —F, —Cl, —Br, —I, —CN, —NO 2 , wherein R a has the meanings given above, and any two or more R 9 groups may together form a ring, and b is selected from 0, 1, 2, 3 and 4.
  • R 1 is a group of the formula:
  • R 2 is defined as C( ⁇ O)N(R a ) 2 .
  • R 2 can be C( ⁇ O)NHR a , wherein the R a group present in R 2 is C 3-8 cycloalkyl, C 2-8 heterocyclyl, C 6-12 aryl, or C 3-12 heteroaryl.
  • Preferred groups include C 5-6 cycloalkyl and C 6 aryl.
  • AVG-044 (172944) C AVG-045 C AVG-046 C AVG-047 C AVG-048 C AVG-049 C AVG-050 C AVG-051 C AVG-052 C AVG-053 C AVG-054 C AVG-055 C AVG-056 C AVG-057 C AVG-058 C AVG-059 C AVG-060 C AVG-061 C AVG-062 C AVG-063 C AVG-064 C AVG-083 C AVG-084 C AVG-085 C AVG-086 C AVG-087 (purity ⁇ 90%) C AVG-111 C AVG-112 C AVG-113 C AVG-114 C AVG-137 C AVG-138 C AVG-139 C AVG-140 C AVG-141 C AVG-142 C AVG-143 C AVG-144 C AVG-145 C AVG-146 C AVG-147 C AVG-148 C AVG-149
  • RSV antiviral agents are useful in the treatment of RSV infections. Accordingly, these compounds of the invention are useful in the treatment of RSV disease, such as bronchiolitis or pneumonia, or in reducing exacerbation of underlying or pre-existing respiratory diseases or conditions wherein RSV infection is a cause of said exacerbation.
  • RSV disease such as bronchiolitis or pneumonia
  • underlying or pre-existing respiratory diseases or conditions may include asthma, chronic obstructive pulmonary disease (COPD) and immunosuppression such as immunosuppression experienced by bone marrow transplant recipients.
  • COPD chronic obstructive pulmonary disease
  • immunosuppression such as immunosuppression experienced by bone marrow transplant recipients.
  • the compounds above may also be combined with one or more other RSV antiviral agents.
  • the compounds of the invention may be formulated as pharmaceutical compositions and administered to a human patient as set forth in more detail below.
  • the compounds can be delivered in a number of suitable ways including orally, intravenously, topically, parentally, subcutaneously, intradermally, or by inhalation.
  • Exemplary routes of administration include buccal, oral, intravenous, intramuscular, topical, subcutaneous, rectal, vaginal, parenteral, pulmonary, intranasal, ophthalmic, and the like, as set forth in more detail below.
  • Useful dosages of the compounds of the invention for inclusion in the pharmaceutical compositions of the invention can be determined by comparing in vitro activity and in vivo activity of the compounds in appropriate animal models.
  • concentration of the compound(s) of the invention in a liquid composition will range from about 0.1% to about 95% by weight, preferably from about 0.5% to about 25% by weight.
  • concentration in a semi-solid or solid composition will range from about 0.1% to 100% by weight, preferably about 0.5% to about 5% by weight.
  • Single doses for intravenous injection, subcutaneous, intramuscular or topical administration, infusion, ingestion or suppository will generally be from about 0.001 to about 5000 mg, and be administered from about 1 to about 3 times daily, to yield levels of about 0.01 to about 500 mg/kg, for adults.
  • the compounds can be co-administered with one or more other agents for the treatment or prevention of RSV infection.
  • the other agents can be formulated separately, and administered either at the same or different time as the compounds of the instant invention.
  • the other agents can be co-formulated with the compounds of the instant invention to give a combination dosage form.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable vehicle, excipient or carrier, and the form of this composition can be suitable for a number of different modes of administration to a patient as set forth below.
  • the pharmaceutical composition may further comprise or be administered in combination with one or more other RSV antiviral agents such as Virazole®, BMS-4337715, TMC3531216, MDT-637 (formerly VP-14637), GS-5806, RSV604, ALNRSV01, AL-8176 (or ALS-8176) and/or other agents that may be developed as inhibitors of viral entry, assembly, replication, egress or host-virus interactions
  • RSV antiviral agents such as Virazole®, BMS-4337715, TMC3531216, MDT-637 (formerly VP-14637), GS-5806, RSV604, ALNRSV01, AL-8176 (or ALS-8176) and/or other agents that may be developed as inhibitors of viral entry, assembly, replication, egress or host-virus interactions
  • composition is intended to include the formulation of an active ingredient with conventional vehicles, carriers and excipients, and also with encapsulating materials as the carrier, to give a capsule in which the active ingredient (with or without other carriers) is surrounded by the encapsulation carrier.
  • Any carrier must be “pharmaceutically acceptable” meaning that it is compatible with the other ingredients of the composition and is not deleterious to a subject.
  • compositions of the present invention may contain other therapeutic agents as described above, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavours and the like) according to techniques such as those well known in the art of pharmaceutical formulation (see, for example, Remington: The Science and Practice of Pharmacy, 21st Ed., 2005, Lippincott Williams & Wilkins).
  • the pharmaceutical composition includes those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
  • the compounds of the invention may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispensable granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavouring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • Suitable vehicles, carriers or excipients include magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter and the like.
  • the term “preparation” is intended to include the formulation of the active compound with an encapsulating material as the carrier by providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • Sterile liquid form compositions include sterile solutions, suspensions, emulsions, syrups and elixirs.
  • the active ingredient can be dissolved or suspended in a pharmaceutically acceptable carrier, such as sterile water, sterile organic solvent or a mixture of both.
  • compositions according to the present invention may thus be formulated for parenteral administration (for example, by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilising and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, for example, sterile, pyrogen-free water, before use.
  • compositions suitable for injectable use include sterile injectable solutions or dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions. They should be stable under the conditions of manufacture and storage and may be preserved against oxidation and the contaminating action of microorganisms such as bacteria or fungi.
  • the solvent or dispersion medium for the injectable solution or dispersion may contain any of the conventional solvent or carrier systems for the compounds, and may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils.
  • compositions suitable for injectable use may be delivered by any appropriate route including intravenous, intramuscular, intracerebral, intrathecal, epidural injection or infusion.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients such as these enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilised active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • preferred methods of preparation are vacuum drying or freeze-drying of a previously sterile-filtered solution of the active ingredient plus any additional desired ingredients.
  • the active ingredients When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers and the like.
  • the amount of active compound in therapeutically useful compositions should be sufficient that a suitable dosage will be obtained.
  • the tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such a sucrose, lactose or saccharin; or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring.
  • a binder such as gum, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin
  • a flavouring agent such as peppermint, oil of wintergreen, or cherry
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound(s) may be incorporated into sustained-release preparations and formulations, including those that allow specific delivery of the active peptide to specific regions of the gut.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavours, stabilising and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • solid form preparations that are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavours, stabilisers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilising agents and the like.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the formulations may be provided in single or multidose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension.
  • the compounds according to the invention may be encapsulated with cyclodextrins, or formulated with other agents expected to enhance delivery and retention in the nasal mucosa.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurised pack with a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, a hydrofluorocarbon (HFC) for example hydrofluoroalkanes (HFA), carbon dioxide, or other suitable gas.
  • a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane
  • HFC hydrofluorocarbon
  • HFA hydrofluoroalkanes
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • the active ingredients may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of, for example gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the compound In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 5 to 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • formulations adapted to give sustained release of the active ingredient may be employed.
  • the pharmaceutical preparations are preferably in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of viral infection in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail.
  • the invention also includes the compounds in the absence of carrier where the compounds are in unit dosage form.
  • Liquids or powders for intranasal administration, tablets or capsules for oral administration and liquids for intravenous administration are the preferred compositions.
  • the compounds as set forth above can be useful in a method of inhibiting RSV or in treating or preventing an RSV infection of other infections caused by related members of the paramyxovirus family such as mumps virus, human parainfluenzaviruses, and Nipah and hendra virus.
  • the reference to RSV as used hereinbelow also include these related members or the paramyxovirus family compounds can also be used to treat an RSV disease or reduce exacerbation of an underlying or pre-existing respiratory disease wherein RSV infection is a cause of said exacerbation.
  • the RSV disease may include bronchiolitis or pneumonia.
  • the underlying or pre-existing respiratory diseases or conditions may include asthma, chronic obstructive pulmonary disease (COPD) and immunosuppression such as immunosuppression experienced by bone marrow transplant recipients.
  • COPD chronic obstructive pulmonary disease
  • Treatment may be therapeutic treatment or prophylactic treatment or prevention.
  • the term “treating” means affecting a subject, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes: (a) inhibiting the viral infection or RSV disease, such as by arresting its development or further development; (b) relieving or ameliorating the effects of the viral infection or RSV disease, such as by causing regression of the effects of the viral infection or RSV disease; (c) reducing the incidence of the viral infection or RSV disease or (d) preventing the viral infection or RSV disease from occurring in a subject, tissue or cell predisposed to the viral infection or RSV disease or at risk thereof, but has not yet been diagnosed with a protective pharmacological and/or physiological effect so that the viral infection or RSV disease does not develop or occur in the subject, tissue or cell.
  • subject refers to any animal, in particular mammals such as humans, having a disease which requires treatment with the compound of formula (I).
  • Particularly preferred treatment groups include at risk populations such as hospitalised subjects, the elderly, high-risk adults and infants.
  • an effective amount of the above compounds, or pharmaceutical compositions thereof, is administered to a patient or subject in need thereof.
  • administering should be understood to mean providing a compound or pharmaceutical composition of the invention to a subject suffering from or at risk of the disease or condition to be treated or prevented.
  • RSV infections and diseases more particularly human and animal RSV infections or diseases
  • the invention may also be useful in the treatment of other viruses of the sub-family Pneumovirinae, more particularly, the genera Pneumovirus and Metapneumovirus.
  • terapéuticaally effective amount refers to the amount of the compound of formula (I) that will elicit the biological or medical response of a subject, tissue or cell that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • effective amount is generally considered that amount that will be effective to treat the condition sought to be treated, or to inhibit RSV, and this effective amount is variable based on a variety of factors including age, size and condition of the patient being treated. Accordingly, one skilled in the art would be readily able to determine the specific effective amount for each patient being treated for RSV, an RSV-related condition, or to inhibit RSV in a given case.
  • an appropriate dosage level will generally be about 0.01 to about 500 mg per kg subject body weight per day which can be administered in single or multiple doses.
  • the dosage may be selected, for example, to any dose within any of these ranges, for therapeutic efficacy and/or symptomatic adjustment of the dosage to the subject to be treated.
  • the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the subject undergoing therapy.
  • the compounds of the invention may generally be prepared by the following methods. Unless otherwise stated, the groups of each of the compounds are as previously defined.
  • DIPEA diisopropylethylamine
  • HATU O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate
  • LiHMDS Lithium bis(trimethylsilyl)amide
  • Wavelength 214 nm and 254 nm.
  • Human carcinoma HEp-2, ATCC CCL-23
  • human lung carcinoma A549, ATCC CCL-185
  • human bronchial epithelial BEAS-2B, ATCC CRL-9609
  • human embryonic kidney 293T, ATCC CRL-3216
  • Madin Darby canine kidney MDCK, ATCC CCL-344 cells were maintained at 37° C. and 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) supplemented with 7.5% fetal bovine serum.
  • DMEM Dulbecco's modified Eagle's medium
  • Lipofectamine 2000 or GeneJuice were used for all transient transfection reactions.
  • recRSV stocks were grown on HEp-2 cells inoculated at a multiplicity of infection (MOI) of 0.01 pfu/cell. Infected cells were kept for 16 hours at 37° C., followed by incubation at 32° C. for five to seven days. Cell-associated progeny virus was released through one freeze/thaw cycle and titers determined by TCID 50 titration on HEp-2 cells. Two alternative strategies were explored to remove contaminating luciferase proteins from virus stocks.
  • MOI multiplicity of infection
  • Progeny virions in culture supernatants (IAV stocks) or released through one freeze/thaw cycle from infected cells (RSV stocks) were cleared (4,000 ⁇ g for 20 minutes at 4° C.), then pelleted (60,000 ⁇ g for 30 minutes at 4° C.). Pelleted material was resuspended in TNE buffer (50 mM Tris/Cl pH 7.2, 10 mM EDTA) and purified through a 20/60% one-step sucrose gradient in TNE buffer (100,000 ⁇ g for 90 minutes at 4° C.). Virions were harvested from the gradient intersection.
  • BEAS-2B cells (8 ⁇ 10 3 /well) were injected in 30 ⁇ l/well into barcoded white wall/clear bottom 384-well plates using a MultiFlo automated dispenser (BioTek) equipped with dual 10- ⁇ l peristaltic pump manifolds, collected (150 ⁇ g for 90 seconds at 25° C.), and incubated for five hours at 37° C. and 5% CO 2 .
  • BioTek MultiFlo automated dispenser
  • Compound was added to a final concentration of 5 ⁇ M (20 nl/well) using a high-density pin tool (V&P Scientific) attached to the pipetting head of Hamilton Nimbus liquid handler, followed by infection in 10 ⁇ l/well using the MultiFlo dispenser unit, spin collection (150 ⁇ g for 90 seconds at 25° C.), and incubation for 40 hours at 37° C. and 5% CO 2 .
  • Final vehicle (DMSO) concentration was 0.05%. Barcodes of source and assay plates were automatically detected and recorded by the Nimbus unit at the time of stamping.
  • RSV minigenome reporter plasmid (pHH-RSV-repl-firefly) was constructed under the control of the constitutive RNA pol I promoter by generating a firefly luciferase open reading frame flanked by the 5′- and 3′-non-coding regions and transfer into a pHH plasmid backbone harboring an RNA pol I promoter.
  • Huh-7 cells were co-transfected with this plasmid and expression plasmids pRSV-L, pRSV-M2-1, pRSV-N and pRSV-P, which encode the RSV polymerase protein subunits.
  • Test compounds were added in serial dilutions, luciferase reporter activities determined 40 hours post-transfection, and EC 50 concentrations calculated if possible.
  • Hep2 cells were seeded in a 12-well plate format and exposed to serial dilutions of compound (3-fold, 20 ⁇ M highest), followed by infection with recombinant RSV-A2 harboring the fusion protein of the L19F isolate at a multiplicity of infection (MOI) of 0.1.
  • MOI multiplicity of infection
  • Cell-associated progeny virions were harvested after 48-hour incubation at 37° C. and subjected to TCID 50 titration on Hep2 cells.
  • Viral titers were calculated based on the Spearman Karber method and inhibitory concentrations based on virus yields determined through four-parameter variable slope regression modeling.
  • compositions and methods of the appended claims are not limited in scope by the specific compositions and methods described herein, which are intended as illustrations of a few aspects of the claims and any compositions and methods that are functionally equivalent are intended to fall within the scope of the claims.
  • Various modifications of the compositions and methods in addition to those shown and described herein are intended to fall within the scope of the appended claims.
  • AVG-049 430.3 2.89, 2.91 10.28, 10.36 C (CDCl 3 ) ⁇ 8.04-7.96 (m, 1H), 7.37-7.30 (m, 1H), 7.29-7.25 (m, 1H), 7.08- 7.05 (m, 1H), 6.98-6.96 (m, 1H), 5.41-5.36 (m, 1H), 4.43-4.41 (m, 1H), 4.06-3.73 (m, 2H), 3.61- 3.44 (m, 2H), 3.42-3.18 (m, 2H), 1.89-1.75 (m, 1H), 1.69-1.64 (m, 4H), 1.54-1.50 (m,4H), 1.48- 1.32 (m, 3H), 1.32-1.22 (m, 2H), 1.26-1.14 (m, 1H), 1.09-0.88 (m, 1H), 0.88-0.84 (m, 1H), 0.83- 0.78 (m, 2H).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Disclosed herein are compounds and compositions for treating or inhibiting RSV and related members of the pneumovirus and paramyxovirus families such as human metapneumovirus, mumps virus, human parainfluenzaviruses, and Nipah and hendra virus, and methods of treatment or prevention thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional application of U.S. Ser. No. 16/099,957 having a filing date of Nov. 8, 2018, which was a 371 application of International application PCT/US2017/031945 filed on May 10, 2017, which claimed the benefit of U.S. Provisional Application Ser. No. 62/333,996, filed May 10, 2016, the disclosures of all said applications being incorporated herein in their entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
  • This invention was made with government support under grants HD079327 and AI071002 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.
  • FIELD OF THE INVENTION
  • The invention relates to the use of small molecule therapeutics for the treatment of respiratory syncytial virus (RSV) and related members of the pneumovirus and paramyxovirus family such as human metapneumovirus, mumps virus, human parainfluenzaviruses, and Nipah and hendra virus.
  • BACKGROUND
  • Respiratory syncytial virus (RSV) is a member of the paramyxovirus family, which consists of mostly highly contagious nonsegmented, negative polarity RNA viruses that spread through the respiratory route. Specifically, RSV is a member of the order Mononegavirales, which consists of the non-segmented negative strand RNA viruses in the Families Paramyxoviridae, Pneumoviridae; Bunyaviridae, Rhabdoviridae and Filoviridae. RSV of humans (often also termed RSV or HRSV) is a member of the Pneumoviridae. Based on genetic and antigenic variations in the structural proteins, RSV is classified into two subgroups, A and B (Mufson, M. et al., J. Gen. Virol. 66:2111-2124). Other members of the Pneumovirus family include viruses such as bovine RSV (BRSV), ovine RSV (ORSV), pneumonia virus of mice (PVM), and the human metapneumoviruses amongst others.
  • In addition to the genome features described above, family characteristics include a lipid envelope containing one or more glycoprotein species considered to be associated with attachment and entry of the host cell. Entry is considered to require a process by which the viral envelope fuses with the membrane of the host cell. Fusion of infected cells with, for example, their neighbors, can also result in the formation of fused multinucleate cells known as syncytia in some cases. The fusion process is believed to be glycoprotein mediated and is a feature shared with diverse enveloped viruses in other taxonomic groups. In the case of the pneumo- and paramyxoviruses, virions characteristically express a fusion glycoprotein (F), which mediates membrane fusion.
  • Respiratory syncytial virus (RSV) is the leading cause of acute upper and lower respiratory tract infections (LRTI) in adults, young children and infants. Although at risk populations include the hospitalized, elderly and high-risk adults, RSV is primarily considered to be a pediatric disease due to the prevalence and severity of unfavorable outcomes in infants. Acute LRTI infections are a leading cause of global childhood mortality and morbidity. Serological evidence indicates that in the western world approximately 95% of all children have been infected with RSV by the age of two and 100% of children have been exposed by the time they reach adulthood.
  • RSV disease is thus the leading cause of virus infection-induced death among children less than 1 year of age and can be life-threatening to the elderly and the immunocompromised. Reinfection with RSV can occur throughout life, but infants born prematurely, or with bronchopulmonary dysplasia or a congenital heart defect, are at highest risk of developing severe disease. In a typical case, initial RSV infection of airway epithelia cells is followed by rapid spread from the nasopharynx to the lower airways that can affect respiratory function through excessive mucus, necrotic epithelial debris, and inflammatory cells obstructing the airways.
  • RSV is a seasonal infectious disease that generally runs from November to March/April in the Northern Hemisphere. In more tropical climates, the annual epidemics are more variable, often coinciding with the wet season. In most cases the RSV infections will only cause minor upper respiratory illness with symptoms resembling that of the common cold. However, severe infection with the virus may result in bronchiolitis or pneumonia, which may result in hospitalization or death. Further, since the immune response to RSV infection is not protective, RSV infections reoccur throughout adulthood. Annual re-infection rates in adults of 3-6% have been observed.
  • RSV infections place a significant burden on the healthcare system. This is particularly so in the case of infants such as, for example, immunodeficient infants, which on average spend twice as long in hospital as other patients with an RSV infection (7-8 days compared to 3-4 days). Hospitalization of infants with acute RSV-related bronchiolitis or RSV-related pneumonia involves supportive care management with oxygen therapy, fluids to prevent dehydration, nasal suctioning and respiratory support. There is also an economic impact associated with parents taking time away from work to care for their child.
  • Attempts to develop an effective RSV vaccine have been fruitless thus far, because the virus is poorly immunogenic overall and neutralizing antibody titers wane quickly after infection. Although ribavirin has been approved for RSV treatment, it has not been widely adopted in clinical use due to efficacy and toxicity issues. The humanized neutralizing antibody palivizumab is used for immunoprophylaxis of high-risk pediatric patients, but high costs prohibit broad-scale implementation.
  • Accordingly, there remains an urgent and unmet need for new compounds that are useful in the treatment and prevention of RSV infections. Small-molecule drug-like therapeutics have high promise to provide a novel avenue towards RSV disease management and prevention. It is therefore an object of the present invention to provide new small-molecule therapeutics classes for the treatment of human patients and other hosts infected with RSV.
  • SUMMARY
  • Disclosed herein are compounds, compositions, and methods of inhibiting RSV, or treating or preventing RSV infection in a patient in need thereof. RSV can be inhibited, and RSV infection can be treated or prevented by administering to a patient in need thereof a composition containing an anti-RSV compound of Formula 1:
  • Figure US20210403444A1-20211230-C00001
  • or a pharmaceutically acceptable salt thereof, wherein
    • X is selected from O, S, or NR7;
    • z and y are independently selected from an integer from 0-6;
    • R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, wherein Ra is in each case independently selected hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl; and when N(Ra)2 is present,
    • R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein Ra is independently selected from the definitions given above,
    • R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
      wherein either of R3 and R4, or R5 and R6 may together form a double bond
      wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin;
      wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring.
  • The details of one or more embodiments are set forth in the descriptions below. Other features, objects, and advantages will be apparent from the description and from the claims.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 includes a depiction of the virus yield of an RSV inhibitor in accordance with the invention [RSVP-172944] as a function of concentration.
  • FIG. 2 includes a depiction of the RdRp activity of an RSV inhibitor in accordance with the invention [RSVP-172944] as a function of concentration.
  • FIG. 3 includes a depiction of the activity of an RSV inhibitor in accordance with the invention [RSVP-172944] in tabular format.
  • DETAILED DESCRIPTION
  • Before the present compounds, compositions, methods and systems are disclosed and described, it is to be understood that the methods and systems are not limited to specific synthetic methods, specific compounds, specific components, or to particular compositions. 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.
  • As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Ranges may be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another embodiment includes, from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint.
  • “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • Throughout the description and claims of this specification, the word “comprise” and variations of the word, such as “comprising” and “comprises,” means “including but not limited to,” and is not intended to exclude, for example, other additives, components, integers or steps. “Exemplary” means “an example of” and is not intended to convey an indication of a preferred or ideal embodiment. “Such as” is not used in a restrictive sense, but for explanatory purposes.
  • Disclosed are components that can be used to perform the disclosed methods and systems. These and other components are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these components are disclosed that while specific reference of each various individual and collective combinations and permutation of these may not be explicitly disclosed, each is specifically contemplated and described herein, for all methods and systems. This applies to all aspects of this application including, but not limited to, steps in disclosed methods. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods.
  • Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer, diastereomer, and meso compound, and a mixture of isomers, such as a racemic or scalemic mixture.
  • The term “alkyl” as used herein is a branched or unbranched hydrocarbon group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, and the like. The alkyl group can also be substituted or unsubstituted. Unless stated otherwise, the term “alkyl” contemplates both substituted and unsubstituted alkyl groups. The alkyl group can be substituted with one or more groups including, but not limited to, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein. An alkyl group which contains no double or triple carbon-carbon bonds is designated a saturated alkyl group, whereas an alkyl group having one or more such bonds is designated an unsaturated alkyl group.
  • Unsaturated alkyl groups having a double bond can be designated alkenyl groups, and unsaturated alkyl groups having a triple bond can be designated alkynyl groups. Unless specified to the contrary, the term alkyl embraces both saturated and unsaturated groups.
  • The term “cycloalkyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc. The term “heterocycloalkyl” is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, selenium or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. Unless stated otherwise, the terms “cycloalkyl” and “heterocycloalkyl” contemplate both substituted and unsubstituted cyloalkyl and heterocycloalkyl groups. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein. A cycloalkyl group which contains no double or triple carbon-carbon bonds is designated a saturated cycloalkyl group, whereas an cycloalkyl group having one or more such bonds (yet is still not aromatic) is designated an unsaturated cycloalkyl group. Unless specified to the contrary, the term alkyl embraces both saturated and unsaturated groups.
  • The term “aryl” as used herein is an aromatic ring composed of carbon atoms. Examples of aryl groups include, but are not limited to, phenyl and naphthyl, etc. The term “heteroaryl” is an aryl group as defined above where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, selenium or phosphorus. The aryl group and heteroaryl group can be substituted or unsubstituted. Unless stated otherwise, the terms “aryl” and “heteroaryl” contemplate both substituted and unsubstituted aryl and heteroaryl groups. The aryl group and heteroaryl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein.
  • Exemplary heteroaryl and heterocyclyl rings include: benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, chromenyL cirrnolinyl, decahydroquinolinyl, 2H,6H˜1,5,2-dithiazinyl, dihydrofuro[2,3 b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, IH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, and xanthenyl.
  • The terms “alkoxy,” “cycloalkoxy,” “heterocycloalkoxy,” “cycloalkoxy,” “aryloxy,” and “heteroaryloxy” have the aforementioned meanings for alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, further providing said group is connected via an oxygen atom.
  • As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms “substitution” or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. Unless specifically stated, a substituent that is said to be “substituted” is meant that the substituent is substituted with one or more of the following: alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, or thiol as described herein. In a specific example, groups that are said to be substituted are substituted with a protic group, which is a group that can be protonated or deprotonated, depending on the pH.
  • Unless specified otherwise, the term “patient” refers to any mammalian organism, including but not limited to, humans.
  • Pharmaceutically acceptable salts are salts that retain the desired biological activity of the parent compound and do not impart undesirable toxicological effects. Examples of such salts are acid addition salts formed with inorganic acids, for example, hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids and the like; salts formed with organic acids such as acetic, oxalic, tartaric, succinic, maleic, fumaric, gluconic, citric, malic, methanesulfonic, p-toluenesulfonic, napthalenesulfonic, and polygalacturonic acids, and the like; salts formed from elemental anions such as chloride, bromide, and iodide; salts formed from metal hydroxides, for example, sodium hydroxide, potassium hydroxide, calcium hydroxide, lithium hydroxide, and magnesium hydroxide; salts formed from metal carbonates, for example, sodium carbonate, potassium carbonate, calcium carbonate, and magnesium carbonate; salts formed from metal bicarbonates, for example, sodium bicarbonate and potassium bicarbonate; salts formed from metal sulfates, for example, sodium sulfate and potassium sulfate; and salts formed from metal nitrates, for example, sodium nitrate and potassium nitrate. Pharmaceutically acceptable and non-pharmaceutically acceptable salts may be prepared using procedures well known in the art, for example, by reacting a sufficiently basic compound such as an amine with a suitable acid comprising a physiologically acceptable anion. Alkali metal (for example, sodium, potassium, or lithium) or alkaline earth metal (for example, calcium) salts of carboxylic acids can also be made.
  • Disclosed herein are compounds, compositions and methods of inhibiting RSV or treating or preventing RSV infection in a patient in need thereof by administering to the patient an effective amount of at least one RSV inhibiting compound. In certain embodiments, the RSV inhibiting compound has the structure of Formula I:
  • Figure US20210403444A1-20211230-C00002
  • or a pharmaceutically acceptable salt thereof, wherein
    • X is selected from O, S, or NR7;
    • z and y are independently selected from an integer from 0-6;
    • R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, C(═O)Ra, C(═O)ORa, C(═O)N(Ra)2, C(═O)SRa,
    • wherein Ra is in each case independently selected hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl; and when N(Ra)2 is present, said two Ra groups may together form a ring;
    • R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein Ra is independently selected from the definitions given above,
    • R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
      wherein either of R3 and R4, or R5 and R6 may together form a double bond
      wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin;
      wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring.
  • As used herein, the term olefin includes unsubstituted methylene (e.g., ═CH2), as well as substituted groups including the functional groups falling with the definitions of R3, R4, R5, or R6. The term imine includes the primary imine (e.g., ═NH) as well as substituted imines including the functional groups falling with the definitions of R7.
  • In certain embodiments, X is S. The RSV inhibiting compound can have the structure of Formula 1a
  • Figure US20210403444A1-20211230-C00003
  • wherein R1-R6, y, and z have the meanings given for the compound of Formula 1.
  • In some cases, the R3 and R4 together form a ring, preferably an aryl or heteroaryl ring. In some instance, the R3 and R4 can together form a phenyl ring, e.g., the RSV inhibiting compound can have the structure of Formula 1 b or 1 b-i:
  • Figure US20210403444A1-20211230-C00004
  • wherein R1, R2, X, y and z are as defined above, a is selected from 0, 1, 2, 3 and 4, and R8 includes —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2. When multiple R8 groups are present, each R8 group is selected independently from the list provided above. In some embodiments, it is preferred that a is 0.
  • In certain embodiments, it is preferred that z is 0 and y is 1:
  • Figure US20210403444A1-20211230-C00005
  • wherein R1-R8, X, and a have the meanings given above.
  • In certain preferred embodiments of the above compounds (e.g., Formula 1, Formula 1a, Formula 1b, Formula 1b-I, Formula 1-xy, Formula 1a-xy, Formula 1b-xy, and Formula 1 b-i-xy), R1 is defined as N(Ra)2. In especially preferred embodiments, N(Ra)2 is a group selected from:
  • Figure US20210403444A1-20211230-C00006
  • Wherein R7 is as defined above, R9 is independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2, —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2, wherein Ra has the meanings given above, and any two or more R9 groups may together form a ring, and b is selected from 0, 1, 2, 3 and 4. In certain preferred embodiments, R1 is a group of the formula:
  • Figure US20210403444A1-20211230-C00007
  • In certain preferred embodiments of the above compounds (e.g., Formula 1, Formula 1a, Formula 1b, Formula 1b-I, Formula 1-xy, Formula 1a-xy, Formula 1b-xy, and Formula 1 b-i-xy), R2 is defined as C(═O)N(Ra)2. For instance, R2 can be C(═O)NHRa, wherein the Ra group present in R2 is C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, or C3-12 heteroaryl. Preferred groups include C5-6 cycloalkyl and C6 aryl.
  • The following compounds in accordance with the present invention and also known as the “C” series are shown in Table 1 below:
  • TABLE 1
    Compounds of the Invention
    AVG-044 (172944) C
    Figure US20210403444A1-20211230-C00008
    AVG-045 C
    Figure US20210403444A1-20211230-C00009
    AVG-046 C
    Figure US20210403444A1-20211230-C00010
    AVG-047 C
    Figure US20210403444A1-20211230-C00011
    AVG-048 C
    Figure US20210403444A1-20211230-C00012
    AVG-049 C
    Figure US20210403444A1-20211230-C00013
    AVG-050 C
    Figure US20210403444A1-20211230-C00014
    AVG-051 C
    Figure US20210403444A1-20211230-C00015
    AVG-052 C
    Figure US20210403444A1-20211230-C00016
    AVG-053 C
    Figure US20210403444A1-20211230-C00017
    AVG-054 C
    Figure US20210403444A1-20211230-C00018
    AVG-055 C
    Figure US20210403444A1-20211230-C00019
    AVG-056 C
    Figure US20210403444A1-20211230-C00020
    AVG-057 C
    Figure US20210403444A1-20211230-C00021
    AVG-058 C
    Figure US20210403444A1-20211230-C00022
    AVG-059 C
    Figure US20210403444A1-20211230-C00023
    AVG-060 C
    Figure US20210403444A1-20211230-C00024
    AVG-061 C
    Figure US20210403444A1-20211230-C00025
    AVG-062 C
    Figure US20210403444A1-20211230-C00026
    AVG-063 C
    Figure US20210403444A1-20211230-C00027
    AVG-064 C
    Figure US20210403444A1-20211230-C00028
    AVG-083 C
    Figure US20210403444A1-20211230-C00029
    AVG-084 C
    Figure US20210403444A1-20211230-C00030
    AVG-085 C
    Figure US20210403444A1-20211230-C00031
    AVG-086 C
    Figure US20210403444A1-20211230-C00032
    AVG-087 (purity <90%) C
    Figure US20210403444A1-20211230-C00033
    AVG-111 C
    Figure US20210403444A1-20211230-C00034
    AVG-112 C
    Figure US20210403444A1-20211230-C00035
    AVG-113 C
    Figure US20210403444A1-20211230-C00036
    AVG-114 C
    Figure US20210403444A1-20211230-C00037
    AVG-137 C
    Figure US20210403444A1-20211230-C00038
    AVG-138 C
    Figure US20210403444A1-20211230-C00039
    AVG-139 C
    Figure US20210403444A1-20211230-C00040
    AVG-140 C
    Figure US20210403444A1-20211230-C00041
    AVG-141 C
    Figure US20210403444A1-20211230-C00042
    AVG-142 C
    Figure US20210403444A1-20211230-C00043
    AVG-143 C
    Figure US20210403444A1-20211230-C00044
    AVG-144 C
    Figure US20210403444A1-20211230-C00045
    AVG-145 C
    Figure US20210403444A1-20211230-C00046
    AVG-146 C
    Figure US20210403444A1-20211230-C00047
    AVG-147 C
    Figure US20210403444A1-20211230-C00048
    AVG-148 C
    Figure US20210403444A1-20211230-C00049
    AVG-149 C
    Figure US20210403444A1-20211230-C00050
    AVG-150 C
    Figure US20210403444A1-20211230-C00051
    AVG-151 C
    Figure US20210403444A1-20211230-C00052
    AVG-152 C
    Figure US20210403444A1-20211230-C00053
    AVG-153 C
    Figure US20210403444A1-20211230-C00054
    AVG-154 C
    Figure US20210403444A1-20211230-C00055
    AVG-155 C
    Figure US20210403444A1-20211230-C00056
    AVG-156 C
    Figure US20210403444A1-20211230-C00057
    AVG-182 C
    Figure US20210403444A1-20211230-C00058
    AVG-215 C
    Figure US20210403444A1-20211230-C00059
    AVG-216 C
    Figure US20210403444A1-20211230-C00060
    AVG-217 C
    Figure US20210403444A1-20211230-C00061
    AVG-218 C
    Figure US20210403444A1-20211230-C00062
    AVG-219 C
    Figure US20210403444A1-20211230-C00063
    AVG-220 C
    Figure US20210403444A1-20211230-C00064
    AVG-221 C
    Figure US20210403444A1-20211230-C00065
    AVG-222 C
    Figure US20210403444A1-20211230-C00066
    AVG-223 C
    Figure US20210403444A1-20211230-C00067
    AVG-224 C
    Figure US20210403444A1-20211230-C00068
    AVG-235 C
    Figure US20210403444A1-20211230-C00069
    AVG-236 C
    Figure US20210403444A1-20211230-C00070
    AVG-237 C
    Figure US20210403444A1-20211230-C00071
    AVG-238 C
    Figure US20210403444A1-20211230-C00072
  • The compounds defined in the above aspects are RSV antiviral agents and are useful in the treatment of RSV infections. Accordingly, these compounds of the invention are useful in the treatment of RSV disease, such as bronchiolitis or pneumonia, or in reducing exacerbation of underlying or pre-existing respiratory diseases or conditions wherein RSV infection is a cause of said exacerbation. The underlying or pre-existing respiratory diseases or conditions may include asthma, chronic obstructive pulmonary disease (COPD) and immunosuppression such as immunosuppression experienced by bone marrow transplant recipients. The compounds above may also be combined with one or more other RSV antiviral agents.
  • The compounds of the invention may be formulated as pharmaceutical compositions and administered to a human patient as set forth in more detail below. The compounds can be delivered in a number of suitable ways including orally, intravenously, topically, parentally, subcutaneously, intradermally, or by inhalation. Exemplary routes of administration include buccal, oral, intravenous, intramuscular, topical, subcutaneous, rectal, vaginal, parenteral, pulmonary, intranasal, ophthalmic, and the like, as set forth in more detail below.
  • Useful dosages of the compounds of the invention for inclusion in the pharmaceutical compositions of the invention can be determined by comparing in vitro activity and in vivo activity of the compounds in appropriate animal models. Generally, the concentration of the compound(s) of the invention in a liquid composition will range from about 0.1% to about 95% by weight, preferably from about 0.5% to about 25% by weight. The concentration in a semi-solid or solid composition will range from about 0.1% to 100% by weight, preferably about 0.5% to about 5% by weight. Single doses for intravenous injection, subcutaneous, intramuscular or topical administration, infusion, ingestion or suppository will generally be from about 0.001 to about 5000 mg, and be administered from about 1 to about 3 times daily, to yield levels of about 0.01 to about 500 mg/kg, for adults.
  • The compounds can be co-administered with one or more other agents for the treatment or prevention of RSV infection. The other agents can be formulated separately, and administered either at the same or different time as the compounds of the instant invention. The other agents can be co-formulated with the compounds of the instant invention to give a combination dosage form.
  • Pharmaceutical Compositions and Modes of Administration
  • The invention also provides a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable vehicle, excipient or carrier, and the form of this composition can be suitable for a number of different modes of administration to a patient as set forth below.
  • The pharmaceutical composition may further comprise or be administered in combination with one or more other RSV antiviral agents such as Virazole®, BMS-4337715, TMC3531216, MDT-637 (formerly VP-14637), GS-5806, RSV604, ALNRSV01, AL-8176 (or ALS-8176) and/or other agents that may be developed as inhibitors of viral entry, assembly, replication, egress or host-virus interactions
  • The term “composition” is intended to include the formulation of an active ingredient with conventional vehicles, carriers and excipients, and also with encapsulating materials as the carrier, to give a capsule in which the active ingredient (with or without other carriers) is surrounded by the encapsulation carrier. Any carrier must be “pharmaceutically acceptable” meaning that it is compatible with the other ingredients of the composition and is not deleterious to a subject. The compositions of the present invention may contain other therapeutic agents as described above, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavours and the like) according to techniques such as those well known in the art of pharmaceutical formulation (see, for example, Remington: The Science and Practice of Pharmacy, 21st Ed., 2005, Lippincott Williams & Wilkins).
  • The pharmaceutical composition includes those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
  • The compounds of the invention, together with a conventional adjuvant, carrier, or diluent, may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • For preparing pharmaceutical compositions from the compounds of the present invention, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispensable granules. A solid carrier can be one or more substances which may also act as diluents, flavouring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • Suitable vehicles, carriers or excipients include magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter and the like. The term “preparation” is intended to include the formulation of the active compound with an encapsulating material as the carrier by providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions. For example, parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • Sterile liquid form compositions include sterile solutions, suspensions, emulsions, syrups and elixirs. The active ingredient can be dissolved or suspended in a pharmaceutically acceptable carrier, such as sterile water, sterile organic solvent or a mixture of both.
  • The compositions according to the present invention may thus be formulated for parenteral administration (for example, by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilising and/or dispersing agents. Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, for example, sterile, pyrogen-free water, before use.
  • Pharmaceutical forms suitable for injectable use include sterile injectable solutions or dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions. They should be stable under the conditions of manufacture and storage and may be preserved against oxidation and the contaminating action of microorganisms such as bacteria or fungi.
  • The solvent or dispersion medium for the injectable solution or dispersion may contain any of the conventional solvent or carrier systems for the compounds, and may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol and the like), suitable mixtures thereof, and vegetable oils.
  • Pharmaceutical forms suitable for injectable use may be delivered by any appropriate route including intravenous, intramuscular, intracerebral, intrathecal, epidural injection or infusion.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients such as these enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilised active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, preferred methods of preparation are vacuum drying or freeze-drying of a previously sterile-filtered solution of the active ingredient plus any additional desired ingredients.
  • When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers and the like.
  • The amount of active compound in therapeutically useful compositions should be sufficient that a suitable dosage will be obtained.
  • The tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such a sucrose, lactose or saccharin; or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound(s) may be incorporated into sustained-release preparations and formulations, including those that allow specific delivery of the active peptide to specific regions of the gut.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavours, stabilising and thickening agents, as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • Also included are solid form preparations that are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavours, stabilisers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilising agents and the like.
  • For topical administration to the epidermis the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch. Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray. The formulations may be provided in single or multidose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension.
  • In the case of a spray, this may be achieved for example by means of a metering atomising spray pump. To improve nasal delivery and retention the compounds according to the invention may be encapsulated with cyclodextrins, or formulated with other agents expected to enhance delivery and retention in the nasal mucosa.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurised pack with a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, a hydrofluorocarbon (HFC) for example hydrofluoroalkanes (HFA), carbon dioxide, or other suitable gas.
  • The aerosol may conveniently also contain a surfactant such as lecithin. The dose of drug may be controlled by provision of a metered valve.
  • Alternatively the active ingredients may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP). Conveniently the powder carrier will form a gel in the nasal cavity. The powder composition may be presented in unit dose form for example in capsules or cartridges of, for example gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 5 to 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • When desired, formulations adapted to give sustained release of the active ingredient may be employed.
  • The pharmaceutical preparations are preferably in unit dosage forms. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of viral infection in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail.
  • The invention also includes the compounds in the absence of carrier where the compounds are in unit dosage form.
  • Liquids or powders for intranasal administration, tablets or capsules for oral administration and liquids for intravenous administration are the preferred compositions.
  • The compounds as set forth above can be useful in a method of inhibiting RSV or in treating or preventing an RSV infection of other infections caused by related members of the paramyxovirus family such as mumps virus, human parainfluenzaviruses, and Nipah and hendra virus. The reference to RSV as used hereinbelow also include these related members or the paramyxovirus family compounds can also be used to treat an RSV disease or reduce exacerbation of an underlying or pre-existing respiratory disease wherein RSV infection is a cause of said exacerbation. The RSV disease may include bronchiolitis or pneumonia. The underlying or pre-existing respiratory diseases or conditions may include asthma, chronic obstructive pulmonary disease (COPD) and immunosuppression such as immunosuppression experienced by bone marrow transplant recipients.
  • Treatment may be therapeutic treatment or prophylactic treatment or prevention. Generally, the term “treating” means affecting a subject, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes: (a) inhibiting the viral infection or RSV disease, such as by arresting its development or further development; (b) relieving or ameliorating the effects of the viral infection or RSV disease, such as by causing regression of the effects of the viral infection or RSV disease; (c) reducing the incidence of the viral infection or RSV disease or (d) preventing the viral infection or RSV disease from occurring in a subject, tissue or cell predisposed to the viral infection or RSV disease or at risk thereof, but has not yet been diagnosed with a protective pharmacological and/or physiological effect so that the viral infection or RSV disease does not develop or occur in the subject, tissue or cell.
  • The term “subject” refers to any animal, in particular mammals such as humans, having a disease which requires treatment with the compound of formula (I). Particularly preferred treatment groups include at risk populations such as hospitalised subjects, the elderly, high-risk adults and infants. In one embodiment of the invention, an effective amount of the above compounds, or pharmaceutical compositions thereof, is administered to a patient or subject in need thereof.
  • The term “administering” or “administered” should be understood to mean providing a compound or pharmaceutical composition of the invention to a subject suffering from or at risk of the disease or condition to be treated or prevented.
  • As indicated above, although the invention has been described with particular reference to treating RSV infections and diseases, more particularly human and animal RSV infections or diseases, it will be appreciated that the invention may also be useful in the treatment of other viruses of the sub-family Pneumovirinae, more particularly, the genera Pneumovirus and Metapneumovirus.
  • Dosages
  • The term “therapeutically effective amount” refers to the amount of the compound of formula (I) that will elicit the biological or medical response of a subject, tissue or cell that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • By “effective amount” is generally considered that amount that will be effective to treat the condition sought to be treated, or to inhibit RSV, and this effective amount is variable based on a variety of factors including age, size and condition of the patient being treated. Accordingly, one skilled in the art would be readily able to determine the specific effective amount for each patient being treated for RSV, an RSV-related condition, or to inhibit RSV in a given case.
  • In the treatment of RSV infections or diseases, an appropriate dosage level will generally be about 0.01 to about 500 mg per kg subject body weight per day which can be administered in single or multiple doses. The dosage may be selected, for example, to any dose within any of these ranges, for therapeutic efficacy and/or symptomatic adjustment of the dosage to the subject to be treated.
  • As indicated above, it will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the subject undergoing therapy.
  • Methods of Preparation
  • The compounds of the invention may generally be prepared by the following methods. Unless otherwise stated, the groups of each of the compounds are as previously defined.
  • General information regarding method of preparation: In the syntheses of the present invention, all evaporations were carried out in vacuo with a rotary evaporator. Analytical samples were dried in vacuo (1-5 mmHg) at rt. Thin layer chromatography (TLC) was performed on silica gel plates, spots were visualized by UV light (214 and 254 nm). Purification by column and flash chromatography was carried out using silica gel (200-300 mesh). Solvent systems are reported as mixtures by volume. All NMR spectra were recorded on a Bruker 400 (400 MHz) spectrometer. 1H chemical shifts are reported in 5 values in ppm with the deuterated solvent as the internal standard. Data are reported as follows: chemical shift, multiplicity (s=singlet, d=doublet, t=triplet, q=quartet, br=broad, m=multiplet), coupling constant (Hz), integration.
  • As described further below, the general method for producing compounds in accordance with the invention is shown schematically as follows:
  • Figure US20210403444A1-20211230-C00073
    Figure US20210403444A1-20211230-C00074
  • Additional information regarding this general method is included in the Examples below.
  • Examples
  • The following examples are set forth below to illustrate the methods and results according to the disclosed subject matter. These examples are not intended to be inclusive of all aspects of the subject matter disclosed herein, but rather to illustrate representative methods, compositions, and results. These examples are not intended to exclude equivalents and variations of the present invention, which are apparent to one skilled in the art.
  • Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.) but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric. There are numerous variations and combinations of reaction conditions, e.g., component concentrations, temperatures, pressures, and other reaction ranges and conditions that can be used to optimize the product purity and yield obtained from the described process. Only reasonable and routine experimentation will be required to optimize such process conditions.
  • Example 1
  • General Method of Preparation of the Compounds of the Invention
  • In all of the experimental data as reported below, the following abbreviations are used:
  • rt: room temperature
  • UV: ultra violet
  • HPLC: high pressure liquid chromatography
  • Rt: retention time
  • LCMS: Liquid chromatography mass spectroscopy
  • NMR: Nuclear magnetic resonance spectroscopy
  • CC: column chromatography
  • TLC: thin layer chromatography
  • sat: saturated
  • aq: aqueous
  • DCM: dichloromethane
  • DCE: dichloroethane
  • DMF: dimethylformamide
  • DIPEA: diisopropylethylamine
  • EtOAc: ethyl acetate
  • TEA: triethylamine
  • THF: tetrahydrofurane
  • TFA: trifluoroacetic acid
  • t-BuOK: Potassium tert-butoxide
  • n-BuOH: n-Butanol
  • EtOH: Ethanol
  • HOAc: acetic acid
  • o/n: overnight
  • h: hour(s)
  • min: minutes
  • HATU: O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate
  • LiHMDS: Lithium bis(trimethylsilyl)amide
  • General information: All evaporations were carried out in vacuo with a rotary evaporator. Analytical samples were dried in vacuo (1-5 mmHg) at rt. Thin layer chromatography (TLC) was performed on silica gel plates, spots were visualized by UV light (214 and 254 nm). Purification by column and flash chromatography was carried out using silica gel (200-300 mesh). Solvent systems are reported as mixtures by volume. All NMR spectra were recorded on a Bruker 400 (400 MHz) spectrometer. 1H chemical shifts are reported in 5 values in ppm with the deuterated solvent as the internal standard. Data are reported as follows: chemical shift, multiplicity (s=singlet, d=doublet, t=triplet, q=quartet, br=broad, m=multiplet), coupling constant (Hz), integration.
  • General Synthetic Method
  • The general synthetic method for producing the compounds of the invention is set forth below:
  • Figure US20210403444A1-20211230-C00075
    Figure US20210403444A1-20211230-C00076
  • Exemplary Experimental Procedures for the General Method
  • 1. The synthesis of 2H-benzo[b][1,4]thiazin-3(4H)-one (C-2) is shown as follows:
  • Figure US20210403444A1-20211230-C00077
  • A mixture of C-1 (20.0 g, 159.8 mmol), methyl 2-chloroacetate (20.8 g, 191.7 mmol) and KOH (17.9 g, 319.5 mmol) in water (80 mL) and EtOH (400 mL) was stirred at 80° C. for 20 h. The mixture was concentrated in vacuo. The residue was diluted with water (150 mL) and stirred for several minutes. The resulting solid was collected by filtration, washed with water (100 mL×3) and dried in vacuo to get the desired compound C-2 (14.5 g, 55% yield) as a light brown solid.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 1.5 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.5 min, then under this condition for 0.5 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.1 min and under this condition for 0.1 min. Purity is 98.7% (in 254 nm). Rt=0.870 min; MS Calcd.: 165.1; MS Found: 166.3 [M+H]+.
  • 2. The synthesis of tert-butyl 2-(3-oxo-2H-benzo[b][1,4]thiazin-4(3H)-yl)acetate (C-3) is shown as follows:
  • Figure US20210403444A1-20211230-C00078
  • To a solution of C-2 (20.0 g, 121.1 mmol) in DMF (150 mL) was added NaH (60%, 5.3 g, 133.2 mmol) in portions and the temperature was maintained at 0° C. After being stirred for 10 min, tert-butyl 2-bromoacetate (28.3 g, 145.3 mmol) was added dropwise over 5 min. After the addition, the reaction mixture was stirred at room temperature for 2 h. The mixture was diluted with water (200 mL) and extracted with ethyl acetate (100 mL×3). The combined organic layers were washed with brine (100 mL×2), dried over sodium sulfate, and concentrated to dryness. The residue was purified by column chromatography on silica gel (petrol ether/EtOAc=5/1) to give C-3 (18.0 g, 53% yield) as yellow oil.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 2.0 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.6 min, then under this condition for 1.4 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.05 min and under this condition for 0.7 min. Rt=1.874 min; MS Calcd.: 279.1; MS Found: 302.2 [M+Na]+.
  • 3. The synthesis of ethyl 4-(2-tert-butoxy-2-oxoethyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]thiazine-2-carboxylate (C-4d) is shown as follows:
  • Figure US20210403444A1-20211230-C00079
  • To a solution of C-3 (10.0 g, 35.8 mmol) in THF (150 mL) at −78° C. was added LiHMDS (1 M in THF, 71.6 mL, 71.6 mmol) dropwise. After being stirred for 10 min at this temperature, ethyl carbonochloridate (3.9 g, 35.8 mmol) was added dropwise over 5 min and the reaction mixture was stirred at −78° C. for 2 h. The mixture was diluted with sat. NH4Cl solution (100 mL) and extracted with ethyl acetate (80 mL×3). The combined organic layers were washed with brine (100 mL×2), dried over sodium sulfate, and concentrated to dryness. The residue was purified by column chromatography on silica gel (petrol ether/EtOAc=4/1) to give C-4d (8.0 g, 64% yield) as brown oil.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 2.0 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.6 min, then under this condition for 1.4 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.05 min and under this condition for 0.7 min. Purity is 74.7%. Rt=1.923 min; MS Calcd.: 351.1; MS Found: 374.0 [M+Na]+.
  • 4. The synthesis of 2-(2-(ethoxycarbonyl)-3-oxo-2H-benzo[b][1,4]thiazin-4(3H)-yl)acetic acid (C-5f) is shown as follows:
  • Figure US20210403444A1-20211230-C00080
  • A mixture of C-4d (150 mg, 0.43 mmol) in TFA (2 mL) was stirred at room temperature for 1 h. The solvent was removed by concentration to give C-5f (120 mg, 95% yield) as yellow oil, which was used for next step directly.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 2.0 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.6 min, then under this condition for 1.4 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.05 min and under this condition for 0.7 min. Purity is 72.1%. Rt=1.526 min; MS Calcd.: 295.0; MS Found: 296.0 [M+H]+.
  • 5. The synthesis of ethyl 4-(2-(cyclohexylamino)-2-oxoethyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]thiazine-2-carboxylate (C-6f) is shown as follows:
  • Figure US20210403444A1-20211230-C00081
  • A mixture of C-5f (120 mg, 0.41 mmol), HATU (311 mg, 0.82 mmol), DIPEA (210 mg, 1.63 mmol) and cyclohexanamine (40 mg, 0.41 mmol) in THF (5 mL) was stirred at room temperature under nitrogen for 16 h. The mixture was diluted with water (30 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over sodium sulfate, and concentrated to dryness. The residue was purified by column chromatography on silica gel (petrol ether/EtOAc=2/1) to give C-6f (110 mg, 72% yield) as brown oil.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 um); Column Temperature: 40° C.; Flow Rate: 1.5 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.5 min, then under this condition for 0.5 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.1 min and under this condition for 0.1 min. Purity is >95% (in 254 nm). Rt=1.280 min; MS Calcd.: 376.1; MS Found: 377.4 [M+H]+.
  • 6. The synthesis of 4-(2-(cyclohexylamino)-2-oxoethyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]thiazine-2-carboxylic acid (C-7f) is shown as follows:
  • Figure US20210403444A1-20211230-C00082
  • To a solution of C-6f (100 mg, 0.27 mmol) in MeOH (6 mL) was added 2 N NaOH (0.27 mL, 0.54 mmol). The reaction mixture was stirred at room temperature for 1 h. The mixture was diluted with water (20 mL), then adjusted to pH=2-3 with diluted hydrochloric acid and extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over sodium sulfate, and concentrated to dryness to give C-7f (90 mg, 97% yield) as a white solid.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 um); Column Temperature: 40° C.; Flow Rate: 1.5 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 1.5 min, then under this condition for 0.5 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.1 min and under this condition for 0.1 min. Purity is >99% (in 254 nm). Rt=1.096 min; MS Calcd.: 348.1; MS Found: 349.3 [M+H]+.
  • 7. The synthesis of N-cyclohexyl-2-(3-oxo-2-(piperidine-1-carbonyl)-2H-benzo[b][1,4]thiazin-4(3H)-yl) acetamide (AVG-044) is shown as follows:
  • Figure US20210403444A1-20211230-C00083
  • A mixture of C-7f (90 mg, 0.26 mmol), HATU (160 mg, 0.42 mmol), DIPEA (134 mg, 1.03 mmol) and piperidine (22 mg, 0.26 mmol) in THF (4 mL) was stirred at room temperature under nitrogen for 16 h. The mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over sodium sulfate, and concentrated to dryness. The residue was purified by Prep-TLC (petrol ether/EtOAc=1/1) to give AVG-044 (23 mg, 21% yield) as a white solid.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 2.0 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 3.0 min, then under this condition for 1.0 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.05 min and under this condition for 0.7 min. Purity is 97.3%. Rt=2.684 min; MS Calcd.: 415.2; MS Found: 416.1 [M+H]+.
  • Agilent HPLC 1200; Column: L-column2 ODS (150 mm*4.6 mm*5.0 μm); Column Temperature: 40° C.; Flow Rate: 1.0 mL/min; Mobile Phase: from 95% [water+0.1% TFA] and 5% [CH3CN+0.1% TFA] to 0% [water+0.1% TFA] and 100% [CH3CN+0.1% TFA] in 10 min, then under this condition for 5 min, finally changed to 95% [water+0.1% TFA] and 5% [CH3CN+0.1% TFA] in 0.1 min and under this condition for 5 min. Purity is 97.3%. Rt=9.907 min.
  • 1H NMR (400 MHz, CDCl3) δ 8.03 (d, J=7.6 Hz, 1H), 7.33 (dd, J=7.8, 1.4 Hz, 1H), 7.26-7.24 (m, 1H), 7.06 (ddd, J=7.6, 7.6, 0.8 Hz, 1H), 6.94 (dd, J=8.4, 0.8 Hz, 1H), 5.39 (d, J=17.2 Hz, 1H), 4.41 (s, 1H), 3.86 (d, J=17.2 Hz, 1H), 3.83-3.76 (m, 1H), 3.57-3.51 (m, 2H), 3.41-3.16 (m, 2H), 1.84-1.81 (m, 2H), 1.69-1.65 (m, 6H), 1.58-1.55 (m, 2H), 1.43-1.35 (m, 1H), 1.33-1.25 (m, 2H), 1.20-1.03 (m, 3H).
  • 7. The synthesis of N-cyclohexyl-2-(2-(morpholine-4-carbonyl)-3-oxo-2H-benzo[b][1,4]thiazin-4(3H)-yl) acetamide (AVG-054) is shown as follows:
  • Figure US20210403444A1-20211230-C00084
  • A mixture of C-7f (90 mg, 0.26 mmol), HATU (160 mg, 0.42 mmol), DIPEA (134 mg, 1.03 mmol) and morpholine (23 mg, 0.26 mmol) in THF (4 mL) was stirred at room temperature under nitrogen for 16 h. The mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layers were washed with brine (20 mL×2), dried over sodium sulfate, and concentrated to dryness. The residue was purified by Prep-HPLC to give AVG-054 (24 mg, 22% yield) as a white solid.
  • Prep-HPLC Condition:
  • 1.1 Chromatographic Equipment
      • Gilson Prep-HPLC system: GX-281 sample manager, 306 pump, 806 Manometric module, 811 D DYNAMIC Mixer, UVNIS-156
  • 1.2 Chromatographic Condition
      • Column: Waters X-Bridge™ Prep C18 5 μm OBD™, 30×100 mm
      • Flowrate: 20 mL/min
      • Gradient:
  • Water (0.04%
    Time(min) MeOH NH4HCO3)
     0 55 45
     8.0 80 20
     9 95  5
    13.5 95  5
    13.6 55 45
    17.6 55 45
  • Wavelength: 214 nm and 254 nm.
  • Agilent LCMS 1200-6110, Column: Waters X-Bridge C18 (50 mm*4.6 mm*3.5 μm); Column Temperature: 40° C.; Flow Rate: 2.0 mL/min; Mobile Phase: from 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] to 0% [water+0.05% TFA] and 100% [CH3CN+0.05% TFA] in 3.0 min, then under this condition for 1.0 min, finally changed to 95% [water+0.05% TFA] and 5% [CH3CN+0.05% TFA] in 0.05 min and under this condition for 0.7 min. Purity is >99%. Rt=2.268 min; MS Calcd.: 417.2; MS Found: 418.0 [M+H]+.
  • Agilent HPLC 1200; Column: L-column2 ODS (150 mm*4.6 mm*5.0 μm); Column Temperature: 40° C.; Flow Rate: 1.0 mL/min; Mobile Phase: from 95% [water+0.1% TFA] and 5% [CH3CN+0.1% TFA] to 0% [water+0.1% TFA] and 100% [CH3CN+0.1% TFA] in 10 min, then under this condition for 5 min, finally changed to 95% [water+0.1% TFA] and 5% [CH3CN+0.1% TFA] in 0.1 min and under this condition for 5 min. Purity is >99%. Rt=8.103 min.
  • 1H NMR (400 MHz, CDCl3) δ 7.86 (d, J=8.0 Hz, 1H), 7.34 (dd, J=7.6, 1.2 Hz, 1H), 7.31-7.27 (m, 1H), 7.08 (ddd, J=7.6, 7.4, 0.8 Hz, 1H), 6.96 (dd, J=8.2, 1.0 Hz, 1H), 5.39 (d, J=17.2 Hz, 1H), 4.39 (s, 1H), 3.90 (d, J=17.2 Hz, 1H), 3.84-3.74 (m, 3H), 3.71-3.62 (m, 2H), 3.59-3.49 (m, 2H), 3.47-3.37 (m, 2H), 1.85-1.81 (m, 2H), 1.72-1.60 (m, 2H), 1.48-1.22 (m, 3H), 1.19-1.01 (m, 3H).
  • Example 2
  • Cell Lines and Transfections
  • Human carcinoma (HEp-2, ATCC CCL-23), human lung carcinoma (A549, ATCC CCL-185), human bronchial epithelial (BEAS-2B, ATCC CRL-9609), human embryonic kidney (293T, ATCC CRL-3216), and Madin Darby canine kidney (MDCK, ATCC CCL-34) cells were maintained at 37° C. and 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) supplemented with 7.5% fetal bovine serum. Lipofectamine 2000 or GeneJuice were used for all transient transfection reactions.
  • RSV Amplification and Virion Purification
  • recRSV stocks were grown on HEp-2 cells inoculated at a multiplicity of infection (MOI) of 0.01 pfu/cell. Infected cells were kept for 16 hours at 37° C., followed by incubation at 32° C. for five to seven days. Cell-associated progeny virus was released through one freeze/thaw cycle and titers determined by TCID50 titration on HEp-2 cells. Two alternative strategies were explored to remove contaminating luciferase proteins from virus stocks. Progeny virions in culture supernatants (IAV stocks) or released through one freeze/thaw cycle from infected cells (RSV stocks) were cleared (4,000×g for 20 minutes at 4° C.), then pelleted (60,000×g for 30 minutes at 4° C.). Pelleted material was resuspended in TNE buffer (50 mM Tris/Cl pH 7.2, 10 mM EDTA) and purified through a 20/60% one-step sucrose gradient in TNE buffer (100,000×g for 90 minutes at 4° C.). Virions were harvested from the gradient intersection.
  • Automated HTS Protocol in 384-Well Plate Format
  • BEAS-2B cells (8×103/well) were injected in 30 μl/well into barcoded white wall/clear bottom 384-well plates using a MultiFlo automated dispenser (BioTek) equipped with dual 10-μl peristaltic pump manifolds, collected (150×g for 90 seconds at 25° C.), and incubated for five hours at 37° C. and 5% CO2. Compound was added to a final concentration of 5 μM (20 nl/well) using a high-density pin tool (V&P Scientific) attached to the pipetting head of Hamilton Nimbus liquid handler, followed by infection in 10 μl/well using the MultiFlo dispenser unit, spin collection (150×g for 90 seconds at 25° C.), and incubation for 40 hours at 37° C. and 5% CO2. Final vehicle (DMSO) concentration was 0.05%. Barcodes of source and assay plates were automatically detected and recorded by the Nimbus unit at the time of stamping. Using a stacker unit with integrated barcode reader (Biotek) attached to an H1 Biotek synergy plate reader, plates were automatically loaded, luciferase substrates (15 μl/well) injected, and bioluminescence recorded after a three minute lag time. Readouts were automatically saved by plate barcode. For analysis of primary screen raw data, normalized relative inhibition values were calculated for each compound by subtracting each value from the average of the plate vehicle controls, followed by dividing the results by the difference between the means of plate vehicle and positive controls. Hits candidates were defined as compounds showing ≥75% inhibition of normalized signal intensity against either or both viral targets.
  • Dose-Response Counterscreens
  • Two-fold serial dilutions of hit candidates were prepared in 384-well plates in three replicates each using the Nimbus liquid handler. BEAS-2B cells (8×103/well) were then plated as before, serial dilutions transferred using the pin-tool, and cells infected with recombinant virus strains expressing distinct luciferase reporter proteins or left uninfected for cell viability assessment. Reporter signals were recorded as outlined above. To determine cell viability, PrestoBlue substrate (life technologies) was added after 40-hour incubation of cells at 37° C. (5 μl/well) and top-read fluorescence (excitation 560 nm; emission 590 nm; instrument gain 85) recorded after 45 minutes of incubation at 37° C. using the H1 synergy plate reader. Four-parameter variable slope regression modeling was applied to determine 50% active (EC50) and toxic (CC50) concentrations.
  • RSV Minigenome Reporter Assays
  • An RSV minigenome reporter plasmid (pHH-RSV-repl-firefly) was constructed under the control of the constitutive RNA pol I promoter by generating a firefly luciferase open reading frame flanked by the 5′- and 3′-non-coding regions and transfer into a pHH plasmid backbone harboring an RNA pol I promoter. Huh-7 cells were co-transfected with this plasmid and expression plasmids pRSV-L, pRSV-M2-1, pRSV-N and pRSV-P, which encode the RSV polymerase protein subunits. Test compounds were added in serial dilutions, luciferase reporter activities determined 40 hours post-transfection, and EC50 concentrations calculated if possible.
  • Virus Yield Reduction Assay
  • Hep2 cells were seeded in a 12-well plate format and exposed to serial dilutions of compound (3-fold, 20 μM highest), followed by infection with recombinant RSV-A2 harboring the fusion protein of the L19F isolate at a multiplicity of infection (MOI) of 0.1. Cell-associated progeny virions were harvested after 48-hour incubation at 37° C. and subjected to TCID50 titration on Hep2 cells. Viral titers were calculated based on the Spearman Karber method and inhibitory concentrations based on virus yields determined through four-parameter variable slope regression modeling.
  • Statistical Analysis
  • The Excel and Prism 6 (GraphPad) software packages were used for data analysis. Statistical significance of differences between sample groups were assessed by unpaired two-tailed t tests or one-way analysis of variance (ANOVA) in combination with Tukey's multiple comparison post-tests, respectively.
  • Assay results for RSV-172944 are presented in FIGS. 1-2.
  • Figure US20210403444A1-20211230-C00085
  • The compositions and methods of the appended claims are not limited in scope by the specific compositions and methods described herein, which are intended as illustrations of a few aspects of the claims and any compositions and methods that are functionally equivalent are intended to fall within the scope of the claims. Various modifications of the compositions and methods in addition to those shown and described herein are intended to fall within the scope of the appended claims. Further, while only certain representative compositions and method steps disclosed herein are specifically described, other combinations of the compositions and method steps also are intended to fall within the scope of the appended claims, even if not specifically recited. Thus, a combination of steps, elements, components, or constituents may be explicitly mentioned herein or less, however, other combinations of steps, elements, components, and constituents are included, even though not explicitly stated. The term “comprising” and variations thereof as used herein is used synonymously with the term “including” and variations thereof and are open, non-limiting terms. Although the terms “comprising” and “including” have been used herein to describe various embodiments, the terms “consisting essentially of” and “consisting of” can be used in place of “comprising” and “including” to provide for more specific embodiments of the invention and are also disclosed. Other than in the examples, or where otherwise noted, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood at the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, to be construed in light of the number of significant digits and ordinary rounding approaches.
  • Example 3: Additional Testing of the C Series Compounds
  • Additional testing was conducted to determine additional parameters of the compounds of the invention. In the following Table, a group of selected C series compounds was analyzed with regard to MS, LC, and NMR, and the results are provided below.
  • Compound Rt(min) Rt(Min)
    ID Structure M + 1 (LCMS) (HPLC) Method 1HNMR (400 MHz)
    AVG-044
    Figure US20210403444A1-20211230-C00086
    416.1 2.68 9.91 C (CDCl3) δ 8.03 (d, J = 7.6 Hz, 1H), 7.33 (dd, J = 7.8, 1.4 Hz, 1H), 7.26- 7.24 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 0.8 Hz, 1H), 6.94 (dd, J = 8.4, 0.8 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 3.86 (d, J = 17.2 Hz, 1H), 3.83-3.76 (m, 1H), 3.57-3.51 (m, 2H), 3.41-3.16 (m, 2H), 1.84- 1.81 (m, 2H), 1.69-1.65 (m, 6H), 1.58-1.55 (m, 2H), 1.43-1.35 (m, 1H), 1.33-1.25 (m, 2H), 1.20- 1.03 (m, 3H).
    AVG-045
    Figure US20210403444A1-20211230-C00087
    418.3 2.23 8.00 C (CDCl3) δ 8.32 (d, J = 7.2 Hz, 1H), 7.34 (d, J = 7.6 Hz, 1H), 7.29-7.28 (m, 1H), 7.08 (dd, J = 7.6, 7.2 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H), 5.43 (d, J = 17.2 Hz, 1H), 4.42 (s, 1H), 4.06-4.04 (m, 1H), 3.92-3.88 (m, 3H), 3.57-3.54 (m, 2H), 3.47-3.42 (m, 2H), 3.37- 3.35 (m, 1H), 3.26-3.25 (m, 1H), 1.78-1.71 (m, 3H), 1.69-1.64 (m, 3H), 1.52-1.48 (m, 2H), 1.40- 1.38 (m, 2H)
    AVG-046
    Figure US20210403444A1-20211230-C00088
    417.3 1.84 6.43 C (CDCl3) δ 8.25 (d, J = 7.6 Hz, 1H), 7.34 (dd, J = 7.8, 1.4 Hz, 1H), 7.29- 7.24 (m, 1H), 7.06 (ddd, J = 7.8, 7.4, 1.2 Hz, 1H), 6.92 (dd, J = 8.4, 0.8 Hz, 1H), 5.41 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 4.0-3.90 (m, 1H), 3.90 (d, J = 17.2 Hz, 1H), 3.60-3.53 (m, 2H), 3.38-3.30 (m, 1H), 3.26- 3.19 (m, 1H), 3.10-3.02 (m, 2H), 2.73-2.65 (m, 2H), 1.86-1.85 (m, 2H), 1.77-1.60 (m, 4H), 1.53- 1.50 (m, 2H), 1.45-1.35 (m, 3H).
    AVG-047
    Figure US20210403444A1-20211230-C00089
    431.3 1.97 7.07 C (CDCl3) δ 8.27 (d, J = 6.8 Hz, 1H), 7.34 (dd, J = 8.0, 1.2 Hz, 1H), 7.28- 7.24 (m, 1H), 7.6 (ddd, J = 7.8, 7.4, 1.2 Hz, 1H), 6.91 (dd, J = 8.4, 0.8 Hz, 1H), 5.42 (d, J = 17.6 Hz, 1H), 4.40 (s, 1H), 3.92-3.86 (m, 2H), 3.58-3.53 (m, 2H), 3.34- 3.33 (m, 1H), 3.26-3.21 (m, 1H), 2.86-2.83 (m, 2H), 2.31 (s, 3H), 2.18- 2.15 (m, 2H), 1.86-1.84 (m, 2H), 1.77-1.70 (m, 3H), 1.54-1.50 (m, 4H), 1.40-1.32 (m, 1H)
    AVG-048
    Figure US20210403444A1-20211230-C00090
    376.2 2.47 8.83 C (CDCl3) δ 8.10 (d, J = 6.8 Hz, 1H), δ 7.33 (dd, J = 7.8, 1.4 Hz, 1H), 7.26-7.24 (m, 1H), 7.06 (ddd, J = 7.6, 7.2, 1.2 Hz, 1H), 6.93 (dd, J = 8.4, 0.8 Hz, 1H), 5.38 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 4.12-4.10 (m, 1H), 3.88 (d, J = 17.2 Hz, 1H), 3.57-3.53 (m, 2H), 3.29-3.21 (m, 2H), 1.70-1.64 (m, 4H), 1.53-1.39 (m, 1H), 1.11- 1.09 (m, 1H), 1.10 (dd, J = 6.4, 0.8 Hz, 6H).
    AVG-049
    Figure US20210403444A1-20211230-C00091
    430.3 2.89, 2.91 10.28, 10.36 C (CDCl3) δ 8.04-7.96 (m, 1H), 7.37-7.30 (m, 1H), 7.29-7.25 (m, 1H), 7.08- 7.05 (m, 1H), 6.98-6.96 (m, 1H), 5.41-5.36 (m, 1H), 4.43-4.41 (m, 1H), 4.06-3.73 (m, 2H), 3.61- 3.44 (m, 2H), 3.42-3.18 (m, 2H), 1.89-1.75 (m, 1H), 1.69-1.64 (m, 4H), 1.54-1.50 (m,4H), 1.48- 1.32 (m, 3H), 1.32-1.22 (m, 2H), 1.26-1.14 (m, 1H), 1.09-0.88 (m, 1H), 0.88-0.84 (m, 1H), 0.83- 0.78 (m, 2H).
    AVG-050
    Figure US20210403444A1-20211230-C00092
    444.3 2.99 10.62 C (CDCl3) δ 8.10 (d, J = 8.4 Hz, 1H), 7.33 (dd, J = 7.8, 1.4 Hz, 1H), 7.26- 7.21 (m, 1H), 7.06 (dd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.95 (dd, J = 8.2, 1.0 Hz, 1H), 5.39 (d, J = 17.6 Hz, 1H), 4.42 (s, 1H), 3.89 (d, J = 17.2 Hz, 1H), 3.77 (m, 1H), 3.55 (m, 2H), 3.39-3.30 (m, 1H), 3.30-3.18 (m, 1H), 1.69-1.67 (m, 3H), 1.66-1.65 (m, 3H), 1.53- 1.46 (m, 1H), 1.45-1.36 (m, 2H), 1.36-1.32 (m 3H), 1.29-1.25 (m, 1H), 1.26-1.24 (m, 1H) , 0.87 (s, 3H), 0.82 (s, 3H).
    AVG-051
    Figure US20210403444A1-20211230-C00093
    402.3 2.6 9.25 C (CDCl3) δ 8.16 (d, J = 6.8 Hz, 1H), 7.33 (dd, J = 7.6, 1.6 Hz, 1H), 7.26- 7.20 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.93 (dd, J = 8.2, 0.6 Hz, 1H), 5.40 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 4.26-4.25 (m, 1H), 3.88 (d, J = 17.2 Hz, 1H), 3.59-3.46 (m, 2H), 3.37-3.34 (m, 1H), 3.26- 3.24 (m, 1H), 1.87 (m, 2H), 1.77-1.59 (m, 5H), 1.55-1.44 (m, 5H), 1.44- 1.24 (m, 2H).
    AVG-052
    Figure US20210403444A1-20211230-C00094
    424.2 2.71 9.66 C (CDCl3) δ 9.58 (s, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.36 (dd, J = 7.6, 1.6 Hz, 1H), 7.35-7.26 (m, 1H), 7.20-7.12 (m, 3H), 7.13-7.07 (m, 2H), 5.63 (d, J = 17.2 Hz, 1H), 4.47 (s, 1H), 4.09 (d, J = 17.6 Hz, 1H), 3.57-3.54 (m, 1H), 3.50-3.41 (m, 1H), 3.40-3.30 (m, 1H), 3.28-3.13 (m, 1H), 2.15 (s, 3H), 1.81-1.71 (m, 1H), 1.70-1.59 (m, 3H), 1.48-1.45 (m, 1H), 1.37- 1.32 (m, 1H).
    AVG-053
    Figure US20210403444A1-20211230-C00095
    424.1 2.78 10.32 C (CDCl3) δ 9.85 (s, 1H), 7.60 (s, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.36 (dd, J = 7.6, 1.6 Hz, 1H), 7.26- 7.22 (m, 1H), 7.15 (dd, J = 8.0, 7.6 Hz, 1H), 7.06 (ddd, J = 7.6, 7.6, 0.8 Hz, 1H), 7.01 (d, J = 8.4 Hz, 1H), 6.88 (d, J = 7.6 Hz, 1H), 5.61 (d, J = 17.2 Hz, 1H), 4.48 (s, 1H), 4.01 (d, J = 17.2 Hz, 1H), 3.70-3.57 (m, 2H), 3.36-3.26 (m, 2H), 2.30 (s, 3H), 2.01-1.71 (m, 3H), 1.71-1.63 (m, 1H), 1.53-1.29 (m, 2H).
    AVG-054
    Figure US20210403444A1-20211230-C00096
    418 2.27 8.1 C (CDCl3) δ 7.86 (d. J = 8.0 Hz, 1H), 7.34 (dd, J = 7.6, 1.2 Hz, 1H), 7.31- 7.27 (m, 1H), 7.08 (ddd, J = 7.6, 7.4, 0.8 Hz, 1H), 6.96 (dd, J = 8.2, 1.0 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.39 (s, 1H), 3.90 (d, J = 17.2 Hz, 1H), 3.84-3.74 (m, 3H), 3.71-3.62 (m, 2H), 3.59-3.49 (m, 2H), 3.47- 3.37 (m, 2H), 1.85-1.81 (m, 2H), 1.72-1.60 (m, 2H), 1.48-1.22 (m, 3H), 1.19-1.01 (m, 3H).
    AVG-055
    Figure US20210403444A1-20211230-C00097
    417.3 1.99 7.04 C (CDCl3) δ 7.93 (d, J = 7.2 Hz, 1H), 7.34 (dd, J = 7.8, 1.4 Hz, 1H), 7.31- 7.27 (m, 1H), 7.07 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.95 (dd, J = 8.2, 1.0 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 3.89 (d, J = 17.2 Hz, 1H), 3.84-3.75 (m, 1H), 3.66-3.54 (m, 2H), 3.48-3.29 (m, 2H), 3.10- 2.94 (m, 2H), 2.87-2.83 (m, 1H), 2.76-2.67 (m, 1H), 1.88-1.80 (m, 3H), 1.61-1.53 (m, 2H), 1.37- 1.24 (m, 2H), 1.19-1.01 (m, 3H).
    AVG-056
    Figure US20210403444A1-20211230-C00098
    431.1 1.77 6.44 C (DMSO-d6) δ 7.96 (d, J = 8.0 Hz, 1H), 7.44 (dd, J = 7.8, 1.4 Hz, 1H), 7.31 (ddd, J = 8.8, 8.4, 1.4 Hz, 1H), 7.08 (dd, J = 7.2, 6.8 Hz, 1H), 6.93 (d, J = 7.6 Hz, 1H), 5.14 (s, 1H), 4.69 (d, J = 16.8 Hz, 1H), 4.28 (d, J = 17.2 Hz, 1H), 3.63-3.52 (m, 2H), 3.48-3.38 (m, 2H), 3.30 (s, 2H), 2.39- 2.28 (m, 3H), 2.18 (s, 3H), 1.72-1.61 (m, 4H), 1.55-1.48 (m, 1H), 1.31- 1.17 (m, 2H), 1.13-1.01 (m, 3H).
    AVG-057
    Figure US20210403444A1-20211230-C00099
    376.3 2.43 8.67 C (CDCl3) δ 7.98 (d, J = 7.2 Hz, 1H), 7.31 (dd, J = 7.7, 1.4 Hz, 1H), 7.25- 7.23 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.95 (dd, J = 8.4, 0.8 Hz, 1H), 5.40 (d, J = 17.2 Hz, 1H), 4.40 (s, 1H), 3.90 (d, J = 17.2 Hz, 1H), 3.83-3.75 (m, 1H), 3.14 (s, 3H), 2.88 (s, 3H), 1.88-1.78 (m, 2H), 1.72-1.61 (m, 2H), 1.34-1.25 (m, 3H), 1.21- 1.12 (m, 2H), 1.10-0.98 (m, 1H).
    AVG-058
    Figure US20210403444A1-20211230-C00100
    430.2 2.8 10.4 C (CDCl3) δ 7.45 (d, J = 8.0 Hz, 1H), 7.35 (dd, J = 7.8, 1.4 Hz, 1H), 7.25- 7.22 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.94 (dd, J = 8.4, 0.8 Hz, 1H), 6.35 (d, J = 7.6 Hz, 1H), 5.40 (d, J = 16.8 Hz, 1H), 4.25 (s, 1H), 3.82-.378 (m, 2H), 3.55-3.53 (m, 1H), 1.92- 1.90 (m, 1H), 1.78-1.64 (m, 5H), 1.52-1.48 (m, 2H), 1.34-1.28 (m, 5H), 1.15-1.11 (m, 6H), 0.81- 0.77 (m, 1H).
    AVG-059
    Figure US20210403444A1-20211230-C00101
    430.3 2.62 9.42 C (CDCl3) δ 7.24 (dd, J = 7.8, 1.4 Hz, 1H), 7.24- 7.19 (m, 1H), 7.06 (ddd, J = 7.6, 7.2, 0.8 Hz, 1H), 6.88 (ddd, J = 7.6, 6.8, 0.8 Hz, 1H), 5.35-5.20 (m, 1H), 4.64-4.62 (m, 1H), 4.41-4.34 (m, 0.5H), 4.25-4.18 (m, 1H), 3.85-3.69 (m, 1H), 3.56-3.47 (m, 1H), 3.45- 3.28 (m, 2.5H), 2.94- 2.87 (m, 3H), 1.91-1.75 (m, 3H), 1.71-1.64 (m, 5H), 1.55-1.50 (m, 4H), 1.43-1.32 (m, 3H), 1.14- 1.07 (m, 1H).
    AVG-060
    Figure US20210403444A1-20211230-C00102
    402.1 2.27 8.65 C (CDCl3) δ 7.42 (dd, J = 7.8, 1.4 Hz, 1H), 7.30 (ddd, J = 7.8, 7.4, 1.2 Hz, 1H), 7.07-6.97 (m, 1H), 6.89 (dd, J = 8.2, 1.0 Hz, 1H), 5.26 (d, J = 16.2 Hz, 1H), 4.64 (s, 1H), 4.22 (d, J = 16.2 Hz, 1H), 3.77-3.75 (m, 1H), 3.59-3.56 (m, 2H), 3.45-3.44 (m, 3H), 3.44- 3.34 (m, 1H), 3.34-3.26 (m, 1H), 1.68-1.67 (m, 3H), 1.67-1.62 (m, 4H), 1.59-1.51 (m, 4H), 1.26 (s, 1H).
    AVG-061
    Figure US20210403444A1-20211230-C00103
    450.2 2.94 10.52 C (CDCl3) δ 8.04 (d, J = 8.0 Hz, 1H), 7.23-7.14 (m, 2H), 6.87 (dd, J = 8.0, 1.6 Hz, 1H), 5.38 (d, J = 17.2 Hz, 1H), 4.49 (s, 1H), 3.86 (d, J = 17.2 Hz, 1H), 3.82-3.74 (m, 1H), 3.72-3.59 (m, 2H), 3.39-3.24 (m, 1H), 3.21-3.07 (m, 1H), 1.91- 1.80 (m, 3H), 1.77-1.60 (m, 5H), 1.56-1.48 (m, 2H), 1.43-1.26 (m, 3H), 1.20-1.04 (m, 3H).
    AVG-062
    Figure US20210403444A1-20211230-C00104
    450.2 3 10.75 C (CDCl3) δ 8.00 (d, J = 7.2 Hz, 1H), 7.32 (d, J = 2.0 Hz, 1H), 7.22 (dd, J = 8.8, 2.0 Hz, 1H), 6.86 (d, J = 8.8 Hz, 1H), 5.40 (d, J = 17.6 Hz, 1H), 4.43 (s, 1H), 3.89-3.75 (m, 2H), 3.61-3.48 (m, 2H), 3.39-3.24 (m, 2H), 1.84-1.79 (m, 2H), 1.76- 1.61 (m, 6H), 1.56-1.40 (m, 3H), 1.33-1.22 (m, 2H), 1.19-1.03 (m, 3H).
    AVG-063
    Figure US20210403444A1-20211230-C00105
    450.2 2.86 10.29 C (CDCl3) δ 7.98 (d, J = 7.6 Hz, 1H), 7.25 (s, 1H), 7.03 (dd, J = 8.4, 2.0 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 5.37 (d, J = 17.6 Hz, 1H), 4.42 (s, 1H), 3.91-3.75 (m, 2H), 3.63-3.50 (m, 2H), 3.37- 3.20 (m, 2H), 1.98-1.78 (m, 2H), 1.78-1.58 (m, 7H), 1.55-1.49 (m, 1H), 1.47-1.38 (m, 1H), 1.36- 1.24 (m, 2H), 1.21-1.04 (m, 3H).
    AVG-064
    Figure US20210403444A1-20211230-C00106
    450.2 2.59 9.31 C (CDCl3) δ 7.37 (d, J = 7.2 Hz, 1H), 7.34 (dd, J = 8.0, 1.6 Hz, 1H), 7.05 (dd, J = 8.0, 7.6 Hz, 1H), 4.79 (d, J = 16.8 Hz, 1H), 4.53-4.42 (m, 2H), 3.85-3.71 (m, 1H), 3.67- 3.21 (m, 4H), 1.94-1.75 (m, 2H), 1.72-1.61 (m, 5H), 1.60-1.49 (m, 3H), 1.43-1.24 (m, 3H), 1.21- 1.08 (m, 3H).
    AVG-083
    Figure US20210403444A1-20211230-C00107
    432.3 2.2 8.32 C (CDCl3) δ 7.37 (dd, J = 7.6, 1.6 Hz, 1H), 7.30- 7.27 (m, 1H), 7.15 (d, J = 7.6 Hz, 1H), 7.07 (ddd, J = 7.6, 7.6, 0.8 Hz, 1H), 6.94 (dd, J = 8.0, 0.8 Hz, 1H), 6.57 (d, J = 7.6 Hz, 1H), 5.37 (d, J = 16.8 Hz, 1H), 4.26 (s, 1H), 3.90-3.74 (m, 5H), 3.46-3.22 (m, 2H), 1.94-1.91 (m, 1H), 1.84-1.81 (m, 1H), 1.77- 1.64 (m, 3H), 1.63-1.58 (m, 1H), 1.42-1.33 (m, 3H), 1.32-1.19 (m, 2H), 1.19-1.02 (m, 3H).
    AVG-084
    Figure US20210403444A1-20211230-C00108
    431.3 1.80 6.30 C (CDCl3) δ 7.37 (d, J = 7.6 Hz, 1H), 7.30-7.24 (m, 1H), 7.11 (d, J = 7.6 Hz, 1H), 7.07 (dd, J = 7.6 Hz, 1H), 6.93 (d, J = 8.0 Hz, 1H), 6.80 (d, J = 8.0 Hz, 1H), 5.36 (d, J = 16.8 Hz, 1H), 4.26 (s, 1H), 3.86 (d, J = 16.8 Hz, 1H), 3.86-3.66 (m, 2H), 3.07-2.96 (m, 2H), 2.73-2.58 (m, 2H), 1.95- 1.56 (m, 5H), 1.52-1.07 (m, 9H).
    AVG-085
    Figure US20210403444A1-20211230-C00109
    445.3 2.07 7.40 C (CDCl3) δ 7.36 (d, J = 7.6 Hz, 1H), 7.30-7.26 (m, 1H), 7.26-7.23 (m, 1H), 7.06 (dd, J = 7.6, 7.2 Hz, 1H), 6.93 (d, J = 8.4 Hz, 1H), 6.49 (d, J = 8.0 Hz, 1H), 5.38 (d, J = 16.8 Hz, 1H), 4.26 (s, 1H), 3.88-3.76 (m, 2H), 3.62-3.50 (m, 1H), 2.74- 2.65 (m, 1H), 2.61-2.50 (m, 1H), 2.23 (s, 3H), 2.10-2.00 (m, 1H), 1.98- 1.88 (m, 2H), 1.84-1.64 (m, 4H), 1.47-1.05 (m, 9H).
    AVG-086
    Figure US20210403444A1-20211230-C00110
    404.3 2.69 9.63 C (CDCl3) δ 7.45 (d, J = 8.0 Hz, 1H), 7.35 (dd, J = 7.8, 1.4 Hz, 1H), 7.26- 7.23 (m, 1H), 7.06 (ddd, J = 7.6, 7.2, 0.8 Hz, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.52 (s, 1H), 5.42 (d, J = 17.2 Hz, 1H), 4.27 (s, 1H), 3.88-3.75 (m, 2H), 3.28-3.24 (m, 1H), 3.03- 2.9 (m, 1H), 1.94-1.91 (m, 1H), 1.82-1.78 (m, 1H), 1.75-1.65 (m, 2H), 1.38-1.27 (m, 4H), 1.19- 1.04 (m, 3H), 1.02-0.79 (m, 3H), 0.73 (t, J = 7.2 Hz, 3H).
    AVG-087
    Figure US20210403444A1-20211230-C00111
    390.3 2.56 9.19 C (CDCl3) δ 7.48 (d, J = 7.6 Hz, 1H), 7.36 (dd, J = 7.6, 1.2 Hz, 1H), 7.26- 7.22 (m, 1H), 7.06 (ddd, J = 8.0, 7.6, 0.6 Hz, 1H), 6.94 (d, J = 8.4 Hz, 1H), 6.31 (d, J = 8.0 Hz, 1H), 5.40 (d, J = 17.2 Hz, 1H), 4.24 (s, 1H), 3.90- 3.74 (m, 3H), 1.96-1.88 (m, 1H), 1.84-1.76 (m, 1H), 1.75-1.65 (m, 2H), 1.43-1.27 (m, 4H), 1.19- 1.10 (m, 2H), 1.09-1.07 (m, 3H), 0.76 (d, J = 6.4 Hz, 3H).
    AVG-111
    Figure US20210403444A1-20211230-C00112
    408.2 2.60 9.24 C (CDCl3) δ 7.71-7.67 (m, 1H), 7.53-7.49 (m, 1H), 4.42 (dd, J = 7.6 Hz, 0.8 Hz, 1H), 7.36-7.31 (m, 2H), 7.26-7.23 (m, 2H), 7.07-7.03 (m, 1H), 5.72 (d, J = 16.8 Hz, 1H), 5.21 (d, J = 17.2 Hz, 1H), 4.66 (s, 1H), 3.66- 3.58 (m, 1H), 3.54-3.34 (m, 3H), 1.71-1.58 (m, 4H), 1.58-1.43 (m, 2H).
    AVG-112
    Figure US20210403444A1-20211230-C00113
    407.2 2.53 9.00 C (CDCl3) δ 11.73 (brs, 1H), 7.75 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 7.6 Hz, 1H), 7.36 (dd, J = 7.8, 1.4 Hz, 1H), 7.26-7.17 (m, 2H), 7.13 (ddd, J = 8.2, 7.6, 1.6 Hz, 1H), 7.03 (ddd, J = 8.0, 7.6, 0.8 Hz, 1H), 6.87 (dd, J = 8.0, 0.8 Hz, 1H), 6.30 (d, J = 17.2 Hz, 1H), 4.85 (d, J = 17.2 Hz, 1H), 4.54 (s, 1H), 3.72- 3.57 (m, 2H), 3.46-3.33 (m, 2H), 1.87-1.60 (m, 5H), 1.49-1.38 (m, 1H).
    AVG-113
    Figure US20210403444A1-20211230-C00114
    334.2 1.96 7.38 C (CDCl3) δ 8.29 (s, 1H), 7.38-7.32 (m, 1H), 7.29- 7.27 (m, 1H), 7.08 (dd, J = 7.6, 7.6 Hz, 1H), 7.01 (d, J = 8.4 Hz, 1H), 5.51-5.24 (m, 2H), 4.44 (s, 1H), 3.91 (d, J = 17.5 Hz, 1H), 3.60-3.47 (m, 2H), 3.41-3.32 (m, 1H), 3.29-3.20 (m, 1H), 1.87- 1.72 (m, 1H), 1.72-1.62 (m, 3H), 1.53-1.45 (m, 1H), 1.42-1.30 (m, 1H).
    AVG-114
    Figure US20210403444A1-20211230-C00115
    374.2 2.34 8.4 C (CDCl3) δ 8.32 (s, 1H), 7.33 (dd, J = 7.8, 1.4 Hz, 1H), 7.30-7.27 (m, 1H), 7.06 (ddd, J = 7.6, 7.4, 1.0 Hz, 1H), 6.92 (dd, J = 8.4, 0.8 Hz, 1H), 5.42 (d, J = 17.6 Hz, 1H), 4.40 (s, 1H), 3.90 (d, J = 17.6 Hz, 1H), 3.64-3.47 (m, 2H), 3.42- 3.16 (m, 2H), 2.91-2.77 (m, 1H), 1.81-1.72 (m, 1H), 1.72-1.61 (m, 3H), 1.56-1.47 (m, 1H), 1.43- 1.32 (m, 1H), 0.76-0.60 (m, 2H), 0.60-0.37 (m, 2H).
    AVG-137
    Figure US20210403444A1-20211230-C00116
    424.3 2.62 9.31 C (DMSO-d6) δ 8.72 (t, J = 5.8 Hz, 1H), 7.45 (dd, J = 8.0, 1.6 Hz, 1H), 7.33- 7.27 (m, 3H), 7.23-7.20 (m, 3H), 7.09 (dd, J = 7.6, 7.2 Hz, 1H), 7.02 (d, J = 8.0 Hz, 1H), 5.14 (s, 1H), 4.68 (d, J = 17.2 Hz, 1H), 4.51 (d, J = 16.8 Hz, 2H), 4.32 (d, J = 6.0 Hz, 2H), 3.51- 3.41 (m, 2H), 3.26-3.20 (m, 2H), 1.55-1.52 (m, 4H), 1.44-1.42 (m, 1H), 1.30-1.28 (m, 1H)
    AVG-138
    Figure US20210403444A1-20211230-C00117
    438.2 2.76 9.75 C (DMSO-d6) δ 8.75-8.55 (m, 1H), 7.31-7.23 (m, 1.5H), 7.23-7.19 (m, 2H), 7.16-7.08 (m, 3H), 7.06-6.92 (m, 2H), 6.65- 6.62 (m, 0.5H), 5.41- 5.34 (m, 1H), 5.11-5.02 (m, 1H), 4.38 (s, 1H), 3.89-3.76 (m, 1H), 3.54- 3.40 (m, 2H), 3.35-3.12 (m, 2H), 1.75-1.53 (m, 4H), 1.45-1.25 (m, 5H)
    AVG-139
    Figure US20210403444A1-20211230-C00118
    452.3 2.90 10.15 C (DMSO-d6) δ 8.35 (s, 1H), 7.45 (dd, J = 7.8, 1.2 Hz, 1H), 7.36 (dd, J = 7.8, 1.2 Hz, 1H), 7.31- 7.24 (m, 4H), 7.18-7.17 (m, 1H), 7.10 (dd, J = 7.6, 7.2 Hz, 1H), 6.99 (d, J = 8.4 Hz, 2H), 5.14 (s, 1H), 4.74 (d, J = 17.2 Hz, 1H), 4.30 (d, J = 16.8 Hz, 1H), 3.55-3.51 (m, 1H), 3.44-3.39 (m, 2H), 3.26-3.21 (m, 1H), 1.58-1.52 (m, 6H), 148- 1.42 (m, 4H), 1.28-1.26 (m, 2H)
    AVG-140
    Figure US20210403444A1-20211230-C00119
    390.3 2.66 9.48 C (CDCl3) δ 7.68 (s, 1H), 7.32 (dd, J = 7.8, 1.4 Hz, 1H), 7.31-7.27 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.96 (dd, J = 8.4, 0.8 Hz, 1H), 5.34 (d, J = 17.2 Hz, 1H), 4.41 (s, 1H), 3.80 (d, J = 17.2 Hz, 1H), 3.58-3.48 (m, 2H), 3.38- 3.18 (m, 2H), 1.86-1.61 (m, 4H), 1.59 (s, 9H), 1.54-1.46 (m, 1H), 1.43- 1.37 (m, 1H).
    AVG-141
    Figure US20210403444A1-20211230-C00120
    404.2 2.69 9.97 C (CDCl3) δ 7.70 (d, J = 7.2 Hz, 1H), 7.37 (dd, J = 7.6, 1.2 Hz, 1H), 7.30- 7.28 (m, 1H), 7.07 (ddd, J = 7.6, 7.6, 0.8 Hz, 1H), 6.94 (dd, J = 8.4, 0.8 Hz, 1H), 6.28 (s, 1H), 5.42 (d, J = 17.2 Hz, 1H), 4.22 (s, 1H), 3.88- 3.73 (m, 2H), 2.01-1.86 (m, 1H), 1.80-1.65 (m, 3H), 1.41-1.18 (m, 4H), 1.17-1.09 (m, 2H), 1.09 (s, 9H).
    AVG-142
    Figure US20210403444A1-20211230-C00121
    438.3 2.66 9.35 C (DMSO-d6) δ 8.98 (t, J = 5.8 Hz, 1H), 7.95 (d, J = 8.0 Hz, 1H), 7.45 (dd, J = 7.6, 1.2 Hz, 1H), 7.33 (ddd, J = 8.4, 8.4, 1.2 Hz, 1H), 7.17 (m, 3H), 7.12 (dd, J = 8.0, 7.2 Hz, 1H), 6.94-6.90 (m, 3H), 4.85 (d, J = 17.2 Hz, 1H), 4.58 (s, 1H), 4.24-4.20 (m, 3H), 3.55- 3.52 (m, 1H), 1.64-1.57 (m, 3H), 1.51-1.48 (m, 2H), 1.22-1.15 (m, 2H), 1.09-0.99 (m, 3H)
    AVG-143
    Figure US20210403444A1-20211230-C00122
    452.3 2.79, 2.86 9.68, 9.88 C (DMSO-d6) δ 7.40-7.26 (m, 4H), 7.17-6.95 (m, 4H), 6.90-6.78 (m, 2H), 6.67(d, J = 7.6 Hz, 1H), 5.46-5.38 (m, 1H), 4.92- 4.84 (m, 1H), 4.34-4.26 (m, 1H), 3.87-3.65 (m, 2H), 1.92-1.62 (m, 3H), 1.58-1.40 (m, 4H), 1.36- 1.05 (m, 5H), 0.75-0.60 (m, 1H).
    AVG-144
    Figure US20210403444A1-20211230-C00123
    466.2 2.79 10.31 C (CDCl3) δ 7.51 (dd, J = 7.6, 1.6 Hz, 1H), 7.43- 7.31 (m, 2H), 7.20 (ddd, J = 7.6, 7.6, 1.0 Hz, 1H), 7.11-6.96 (m, 3H), 6.89 (dd, J = 8.4, 0.8 Hz, 1H), 6.85 (s, 1H), 6.76-6.72 (m, 2H), 5.42 (d, J = 16.8 Hz, 1H), 4.29 (s, 1H), 3.72 (d, J = 16.8 Hz, 1H), 3.67-3.52 (m, 1H), 1.80-1.68 (m, 1H), 1.63 (s, 3H), 1.57-1.53 (m, 1H), 1.52-1.42 (m, 2H), 1.41 (s, 3H), 1.34- 1.26 (m, 1H), 1.25-1.08 (m, 2H), 1.01-0.80 (m, 2H), 0.51-0.32 (m, 1H).
    AVG-145
    Figure US20210403444A1-20211230-C00124
    446.3 2.79 9.88 C (CDCl3) δ 8.10 (d, J = 7.6 Hz, 1H), 7.20 (dd, J = 8.4, 8.0 Hz, 1H), 6.67 (d, J = 8.0 Hz, 1H), 6.60 (d, J = 8.4 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.43 (s, 1H), 3.94-3.84 (m, 4H), 3.83-3.73 (m, 1H), 3.67-3.45 (m, 2H), 3.45-3.20 (m, 2H), 1.90- 1.74 (m, 3H), 1.72-1.61 (m, 5H), 1.55-1.48 (m, 2H), 1.48-1.38 (m, 1H), 1.37-1.25 (m, 2H), 1.22- 1.12 (m, 2H), 1.11-1.03 (m, 1H).
    AVG-146
    Figure US20210403444A1-20211230-C00125
    446.3 2.78 9.67 C (CDCl3) δ 8.00 (d, J = 8.0 Hz, 1H), 6.88-6.84 (m, 2H), 6.78 (dd, J = 8.8, 2.8 Hz, 1H), 5.37 (d, J = 17.2 Hz, 1H), 4.40 (s, 1H), 3.86-3.76 (m, 5H), 3.60-3.49 (m, 2H), 3.38-3.25 (m, 2H), 1.87-1.80 (m, 2H), 1.76- 1.61 (m, 6H), 1.57-1.38 (m, 2H), 1.37-1.25 (m, 3H), 1.20-1.03 (m, 3H).
    AVG-147
    Figure US20210403444A1-20211230-C00126
    446.2 2.87 10.12 C (CDCl3) δ 7.94 (d, J = 7.2 Hz, 1H), 7.14 (d, J = 8.4 Hz, 1H), 6.54 (dd, J = 8.4, 2.4 Hz, 1H), 6.43 (d, J = 2.4 Hz, 1H), 5.29 (d, J = 17.2 Hz, 1H), 4.29 (s, 1H), 3.81 (d, J = 17.2 Hz, 1H), 3.77-3.67 (m, 1H), 3.70 (s, 3H), 3.57-3.44 (m, 2H), 3.29- 3.21 (m, 1H), 3.19-3.11 (m, 1H), 1.84-1.70 (m, 2H), 1.68-1.54 (m, 5H), 1.51-1.42 (m, 2H), 1.37- 1.16 (m, 4H), 1.15-0.95 (m, 3H).
    AVG-148
    Figure US20210403444A1-20211230-C00127
    446.3 2.61 9.29 C (CDCl3) δ 7.07-6.95 (m, 3H), 6.84 (dd, J = 8.0, 1.2 Hz, 1H), 4.83 (d, J = 16.4 Hz, 1H), 4.39 (s, 1H), 4.16 (d, J = 17.6 Hz, 1H), 3.82 (s, 3H), 3.82-3.74 (m, 1H), 3.66- 3.50 (m, 2H), 3.34-3.17 (m, 2H), 1.93-1.80 (m, 2H), 1.72-1.63 (m, 5H), 1.60-1.49 (m, 2H), 1.38- 1.24 (m, 4H), 1.23-1.09 (m, 3H).
    AVG-149
    Figure US20210403444A1-20211230-C00128
    494.2 3.133 10.91 C (CDCl3) δ 7.99 (d, J = 8.0 Hz, 1H), 7.46 (d, J = 2.4 Hz, 1H), 7.36 (dd, J = 8.8, 2.0 Hz, 1H), 6.80 (d, J = 8.8 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.42 (s, 1H), 3.83 (d, J = 17.2 Hz, 1H), 3.79-3.74 (m, 1H), 3.59-3.49 (m, 2H), 3.38-3.27 (m, 2H), 1.83-1.80 (m, 2H), 1.76- 1.62 (m, 6H), 1.56-1.55 (m, 2H), 1.49-1.43 (m, 1H), 1.36-1.25 (m, 2H), 1.19-1.06 (m, 3H).
    AVG-150
    Figure US20210403444A1-20211230-C00129
    434.3 2.90 10.18 C (CDCl3) δ 7.99 (d, J = 7.6 Hz, 1H), 7.07 (dd, J = 7.8, 3.0 Hz, 1H), 6.97 (ddd, J = 8.8, 8.0, 2.8 Hz, 1H), 6.90 (dd, J = 9.2, 4.4 Hz, 1H), 5.39 (d, J = 17.2 Hz, 1H), 4.43 (s, 1H), 3.84 (d, J = 17.2 Hz, 1H), 3.83-3.74 (m, 1H), 3.61-3.50 (m, 2H), 3.37-3.24 (m, 2H), 1.87-1.78 (m, 2H), 1.76- 1.63 (m, 6H), 1.60-1.51 (m, 2H), 1.48-1.38 (m, 1H), 1.37-1.24 (m, 2H), 1.20-1.03 (m, 3H).
    AVG-151
    Figure US20210403444A1-20211230-C00130
    430.3 2.92 10.38 C (CDCl3) δ 8.01 (d, J = 7.2 Hz, 1H), 7.12 (s, 1H), 7.05 (d, J = 8.8 Hz, 1H), 6.82 (d, J = 8.4 Hz, 1H), 5.38 (d, J = 17.2 Hz, 1H), 4.40 (s, 1H), 3.85 (d, J = 17.2 Hz, 1H), 3.83-3.78 (m, 1H), 3.60-3.48 (m, 2H), 3.39- 3.23(m, 2H), 2.29 (s, 3H), 1.87-1.78 (m, 2H), 1.77-1.61 (m, 6H), 1.57- 1.49 (m, 2H), 1.48-1.37 (m, 1H), 1.36-1.23 (m, 2H), 1.20-1.01 (m, 3H).
    AVG-152
    Figure US20210403444A1-20211230-C00131
    444.3 3.05 10.69 C (CDCl3) δ 8.02 (d, J = 7.6 Hz, 1H), 7.13 (d, J = 2.0 Hz, 1H), 7.07 (dd, J = 8.4, 2.0 Hz, 1H), 6.85 (d, J = 8.4 Hz, 1H), 5.37 (d, J = 17.2 Hz, 1H), 4.40 (s, 1H), 3.87 (d, J = 17.2 Hz, 1H), 3.83-3.72 (m, 1H), 3.63-3.42 (m, 2H), 3.42-3.20 (m, 2H), 2.62-2.57 (m, 2H), 1.88- 1.80 (m, 2H), 1.79-1.71 (m, 1H), 1.71-1.62 (m, 5H), 1.57-1.49 (m, 2H), 1.47-1.38 (m, 1H), 1.37- 1.25 (m, 2H), 1.22 (t, J = 7.6 Hz, 3H), 1.19-1.01 (m, 3H).
    AVG-153
    Figure US20210403444A1-20211230-C00132
    458.3 3.19 10.95 C (CDCl3) δ 8.01 (d, J = 7.6 Hz, 1H), 7.15 (d, J = 2.0 Hz, 1H), 7.09 (dd, J = 8.6, 1.8 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 5.36 (d, J = 17.2 Hz, 1H), 4.40 (s, 1H), 3.87 (d, J = 17.2 Hz, 1H), 3.83-3.72 (m, 1H), 3.63-3.45 (m, 2H), 3.43-3.21 (m, 2H), 2.91-2.77 (m, 1H), 1.91- 1.79 (m, 2H), 1.78-1.71 (m, 1H), 1.71-1.59 (m, 5H), 1.57-1.47 (m, 2H), 1.45-1.36 (m, 1H), 1.35- 1.25 (m, 2H), 1.23 (d, J = 7.2 Hz, 6H), 1.18-1.10 (m, 2H), 1.10-0.99 (m, 1H).
    AVG-154
    Figure US20210403444A1-20211230-C00133
    492.3 3.15 11.08 C (CDCl3) δ 8.07 (d, J = 7.6 Hz, 1H), 7.60-7.52 (m, 3H), 7.51-7.41 (m, 3H), 7.39-7.33 (m, 1H), 7.01 (d, J = 8.4 Hz, 1H), 5.42 (d, J = 17.6 Hz, 1H), 4.46 (s, 1H), 3.93 (d, J = 17.6 Hz, 1H), 3.86-3.73 (m, 1H), 3.60- 3.49 (m, 2H), 3.41-3.24 (m, 2H), 1.91-1.78 (m, 2H), 1.77-1.61 (m, 6H), 1.53-1.42 (m, 2H), 1.40- 1.22 (m, 3H), 1.22-1.01 (m, 3H).
    AVG-155
    Figure US20210403444A1-20211230-C00134
    417.3 2.39 8.59 C (CDCl3) δ 8.50 (s, 1H), 8.43 (d, J = 5.6 Hz, 1H), 8.05 (d, J = 7.2 Hz, 1H), 6.82 (d, J = 6.0 Hz, 1H), 5.43 (d, J = 17.6 Hz, 1H), 4.49 (s, 1H), 3.87 (d, J = 17.6 Hz, 1H), 3.81-3.72 (m, 1H), 3.61- 3.50 (m, 2H), 3.41-3.23 (m, 2H), 1.88-1.74 (m, 3H), 1.74-1.60 (m, 6H), 1.54-1.37 (m, 2H), 1.35- 1.26 (m, 2H), 1.18-1.02 (m, 3H).
    AVG-156
    Figure US20210403444A1-20211230-C00135
    484.2 2.94 10.77 C (CDCl3) δ 8.00 (d, J = 8.4 Hz, 1H), 7.59 (d, J = 1.6 Hz, 1H), 7.51 (dd, J = 8.8, 1.6 Hz, 1H), 7.03 (d, J = 8.8 Hz, 1H), 5.43 (d, J = 17.6 Hz, 1H), 4.48 (s, 1H), 3.89 (d, J = 17.6 Hz, 1H), 3.84-3.73 (m, 1H), 3.61-3.48 (m, 2H), 3.41-3.25 (m, 2H), 1.83 (d, J = 12.1 Hz, 2H), 1.78-1.61 (m, 6H), 1.56-1.47 (m, 2H), 1.47- 1.39 (m, 1H), 1.37-1.26 (m, 2H), 1.19-1.03 (m, 3H).
    AVG-182
    Figure US20210403444A1-20211230-C00136
    423.2 2.52 8.82 C (CDCl3) δ11.65 (brs, 1H), 7.76 (d, J = 6.8 Hz, 1H), 7.45-7.34 (m, 2H), 7.26-7.14 (m, 3H), 7.08 (dd, J = 7.6, 7.2 Hz, 1H), 7.00-6.91 (m, 1H), 6.26- 6.08 (m, 1H), 5.65 (brs, 1H), 5.03 (d, J = 16.4 Hz, 1H), 4.06-3.97 (m, 1H), 3.83-3.75 (m, 1H), 3.59-3.51 (m, 1H), 3.43- 3.34 (m, 1H), 1.82-1.60 (m, 4H), 1.60-1.52 (m, 1H), 1.44-1.34 (m, 1H).
    AVG-215
    Figure US20210403444A1-20211230-C00137
    456.3 3.15 10.82 C (CDCl3) δ 8.03 (d, J = 7.6 Hz, 1H), 7.40 (d, J = 1.6 Hz, 1H), 7.35 (dd, J = 8.8, 1.6 Hz, 1H), 6.90 (d, J = 8.4 Hz, 1H), 5.46-5.35 (m, 2H), 5.10 (s, 1H), 4.42 (s, 1H), 3.88 (d, J = 16.8 Hz, 1H), 3.83-3.75 (m, 1H), 3.57-3.50 (m, 2H), 3.39- 3.29 (m, 2H), 2.12 (s, 3H), 1.93-1.80 (m, 2H), 1.78-1.62 (m, 6H), 1.61- 1.58 (m, 1H), 1.51-1.39 (m, 2H), 1.34-1.26 (m, 2H), 1.20-1.04 (m, 3H).
    AVG-216
    Figure US20210403444A1-20211230-C00138
    430.3 2.83 10.421 C (CDCl3) δ 8.19 (brs, 1H), 7.33 (dd, J = 8.0, 1.6 Hz, 1H), 7.29-7.25 (m, 1H), 7.06 (ddd, J = 7.6, 7.6, 1.2 Hz, 1H), 6.95 (d, J = 8.0 Hz, 1H), 5.42 (d, J = 17.6 Hz, 1H), 4.42 (s, 1H), 3.92 (d, J = 17.2 Hz, 2H), 3.56-3.49 (m, 2H), 3.39- 3.33 (m, 1H), 3.30-3.24 (m, 1H), 3.22-3.16 (m, 1H), 3.06-3.00 (m, 1H),
    1.78-1.62 (m, 9H), 1.55-
    1.35 (m, 3H), 1.17-1.08
    (m, 3H), 0.89-0.80 (m,
    2H).
    AVG-217
    Figure US20210403444A1-20211230-C00139
    464.3 3.21 11.23 C (DMSO-d6) δ 8.11 (t, J = 5.6 Hz, 1H), 7.57 (d, J = 2.4 Hz, 1H), 7.36 (dd, J = 8.6, 2.2 Hz, 1H), 6.97 (d, J = 8.8 Hz, 1H), 5.19 (s, 1H), 4.65 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 3.54-3.26 (m, 4H), 2.99-2.87(m, 2H), 1.66-1.49 (m, 9H), 1.48-1.26 (m, 3H), 1.18- 1.01 (m, 3H), 0.88-0.72 (m, 2H).
    AVG-218
    Figure US20210403444A1-20211230-C00140
    464.2 3.18 11.10 C (CDCl3) δ 7.43 (d, J = 7.6 Hz, 1H), 7.36 (d, J = 2.4 Hz, 1H), 7.23 (dd, J = 9.2, 2.4 Hz, 1H), 6.87 (d, J = 8.8 Hz, 1H), 6.30 (d, J = 8.0 Hz, 1H), 5.40 (d, J = 17.2 Hz, 1H), 4.25 (s, 1H), 3.85-3.74 (m, 2H), 3.63-3.52 (m, 1H), 1.94-1.87 (m, 1H), 1.82-1.63 (m, 5H), 1.62- 1.51 (m, 2H), 1.44-1.05 (m, 11H), 0.95-0.84 (m, 1H).
    AVG-219
    Figure US20210403444A1-20211230-C00141
    458.2 3.01 10.55 C (CDCl3) δ 9.53 (s, 1H), 7.40 (d, J = 7.6 Hz, 1H), 7.36 (d, J = 2.4 Hz, 1H), 7.31-7.27 (m, 1H), 7.22- 7.14 (m, 2H), 7.13-7.09 (m, 1H), 7.06 (d, J = 8.4 Hz, 1H), 5.63 (d, J = 17.2 Hz, 1H), 4.49 (s, 1H), 4.04 (d, J = 17.2 Hz, 1H), 3.56-3.36 (m, 3H), 3.33-3.20 (m, 1H), 2.14 (s, 3H), 1.79-1.60 (m, 4H), 1.52-1.46 (m, 1H), 1.45-1.33 (m, 1H).
    AVG-220
    Figure US20210403444A1-20211230-C00142
    444.2 2.82 9.86 C (CDCl3) δ 9.64 (s, 1H), 7.69 (d, J = 8.0 Hz, 1H), 7.40-7.34 (m, 2H), 7.33- 7.30 (m, 1H), 7.25-7.21 (m, 2H), 7.14-7.07 (m, 2H), 5.51 (d, J = 17.2 Hz, 1H), 4.49 (s, 1H), 4.23 (d, J = 17.6 Hz, 1H), 3.59-3.45 (m, 2H), 3.39-3.28 (m, 1H), 3.28- 3.14 (m, 1H), 1.78-1.59 (m, 5H), 1.39-1.25 (m, 1H).
    AVG-221
    Figure US20210403444A1-20211230-C00143
    440.3 2.67 9.46 C (CDCl3) δ 9.06 (s, 1H), 7.99 (d, J = 6.8 Hz, 1H), 7.37 (d, J = 7.6 Hz, 1H), 7.32-7.27 (m, 1H), 7.20 (d, J = 8.4 Hz, 1H), 7.12-7.04 (m, 2H), 6.93 (dd, J = 7.6, 7.6 Hz, 1H), 6.84 (d, J = 8.0 Hz, 1H), 5.28 (d, J = 18.0 Hz, 1H), 4.53 (s, 1H), 4.38 (d, J = 16.4 Hz, 1H), 3.75 (s, 3H), 3.57-3.48 (m, 2H), 3.38-3.29 (m, 2H), 1.73-1.60 (m, 4H), 1.52-1.47 (m, 1H), 1.45- 1.35 (m, 1H).
    AVG-222
    Figure US20210403444A1-20211230-C00144
    441.2 2.46 8.71 C (CDCl3) δ 10.08 (s, 1H), 8.31 (dd, J = 4.6, 1.0 Hz, 1H), 8.21 (d, J = 8.4 Hz, 1H), 7.68 (ddd, J = 8.8, 8.8, 1.8 Hz, 1H), 7.35 (dd, J = 7.6, 1.2 Hz, 1H), 7.31-7.27 (m, 1H), 7.10-6.96 (m, 3H), 5.59 (d, J = 17.6 Hz, 1H), 4.51 (s, 1H), 4.12 (d, J = 17.6 Hz, 1H), 3.80-3.73 (m, 1H), 3.65- 3.58 (m, 1H), 3.36-3.21 (m, 2H), 1.79-1.65 (m, 2H), 1.63-1.60 (m, 2H), 1.46-1.24 (m, 2H).
    AVG-223
    Figure US20210403444A1-20211230-C00145
    411.2 2.45 8.57 C (DMSO-d6) δ 10.35 (s, 1H), 8.75 (d, J = 2.4 Hz, 1H), 8.28 (m, J = 3.2 Hz, 1H), 8.06 (dd, J = 6.8, 1.6 Hz, 1H), 7.48 (dd, J = 7.6, 1.2 Hz, 1H), 7.38- 7.29 (m, 2H), 7.14-7.08 (m, 2H), 5.22 (s, 1H), 4.91 (d, J = 17.6 Hz, 1H), 4.66 (d, J = 17.6 Hz, 1H), 3.56-3.49 (m, 2H), 3.39-3.38 (m, 2H), 1.57-1.47 (m, 4H), 1.32- 1.29 (m, 2H)
    AVG-224
    Figure US20210403444A1-20211230-C00146
    441.2 2.37 8.47 C (CDCl3) δ 10.24 (s, 1H), 8.45 (s, 2H), 7.72 (d, J = 4.8 Hz, 2H), 7.38 (dd, J = 7.8, 1.4 Hz, 1H), 7.32- 7.28 (m, 1H), 7.09 (dd, J = 7.6, 7.6 Hz, 1H), 6.94 (d, J = 8.0 Hz, 1H), 5.63 (d, J = 18.0 Hz, 1H), 4.50 (s, 1H), 4.03 (d, J = 17.6 Hz, 1H), 3.80-3.60 (m, 2H), 3.48-3.23 (m, 2H), 1.85-1.64 (m, 4H), 1.56-1.21 (m, 2H).
  • Example 4: Potency Range of Certain Compounds (EC50 Values)
  • The potency range of selected compounds (EC50 values) was tested and the results are provided below:
  • Potency
    Compound # Range*
    AVG-044 B
    AVG-045 D
    AVG-046 D
    AVG-047 D
    AVG-048 D
    AVG-049 B
    AVG-050 B
    AVG-051 B
    AVG-052 B
    AVG-053 D
    AVG-054 B
    AVG-055 D
    AVG-056 C
    AVG-057 B
    AVG-058 B
    AVG-059 D
    AVG-060 D
    AVG-061 B
    AVG-062 A
    AVG-063 B
    AVG-064 C
    AVG-083 D
    AVG-084 D
    AVG-085 D
    AVG-086 C
    AVG-087 C
    AVG-111 D
    AVG-112 C
    AVG-113 D
    AVG-114 D
    AVG-137 B
    AVG-138 B
    AVG-139 B
    AVG-140 B
    AVG-141 B
    AVG-142 C
    AVG-143 D
    AVG-144 B
    AVG-145 B
    AVG-146 B
    AVG-147 C
    AVG-148 C
    AVG-149 B
    AVG-150 B
    AVG-151 C
    AVG-152 B
    AVG-153 B
    AVG-154 D
    AVG-155 B
    AVG-156 B
    AVG-182 D
    AVG-215 B
    AVG-216 B
    AVG-217 B
    AVG-218 B
    AVG-219 A
    AVG-220 B
    AVG-221 B
    AVG-223 C
    AVG-224 C
    *Potency Range (EC50 values)
    A >0.1-1.0 μM
    B >1.0-10.0 μM
    C >10.0-20.0 μM
    D >20.0 μM

Claims (24)

What is claimed is:
1. A method of inhibiting RSV, comprising administering to a patient in need thereof, an effective amount of a compound of Formula 1:
Figure US20210403444A1-20211230-C00147
or a pharmaceutically acceptable salt thereof, wherein
X is selected from O, S, or NR7;
z is selected from 0, 1, 2, 3, 4, 5, and 6;
y is selected from 0, 1, 2, 3, 4, 5, and 6;
R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, C(═O)Ra, C(═O)ORa, C(═O)N(Ra)2, C(═O)SRa;
R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein when multiple Ra groups are present, said Ra groups may together form a ring;
R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
wherein either of R3 and R4, or R5 and R6 may together form a double bond
wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin; and
wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring; and
wherein Ra is in each case independently selected from hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl.
2. The method according to claim 1, wherein X is S.
3. The method according to claim 1, wherein R3 and R4 together form a ring.
4. The method according to claim 1, wherein R3 and R4 together form an aryl or heteroaryl ring.
5. The method according to claim 1, wherein R3 and R4 can together form a phenyl ring, to give a compound of Formula 1b or 1b-i:
Figure US20210403444A1-20211230-C00148
wherein a is selected from 0, 1, 2, 3 and 4; and
R8 is independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2,
wherein Ra has the meanings as set forth in claim 1.
6. The method according to claim 1, wherein z is 0.
7. The method according to claim 1, wherein y is 1.
8. The method according to claim 1, wherein R1 is N(Ra)2.
9. The method according to claim 1, wherein R1 is selected from
Figure US20210403444A1-20211230-C00149
b is selected from 0, 1, 2, 3 and 4.
R9 is independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2, wherein Ra has the meanings given above, and any two or more R9 groups may together form a ring;
wherein R10 when present, is independently selected from Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2, wherein R10 may, with any one or more of R1, R2, R3, R4, R5, R6 or R7, R8, or Ra groups, together form a ring.
10. The method according to claim 1, wherein R1 is a group of the formula:
Figure US20210403444A1-20211230-C00150
11. The method according to claim 1, wherein R2 is C(═O)NHRa1, wherein said Ra1 is selected from C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, and C3-12 heteroaryl.
12. The method according to claim 1, wherein R2 is C(═O)NH—C3-8 cycloalkyl.
13. The method according to claim 1, wherein R2 is C(═O)NHcyclohexyl.
14. The method according to claim 1 wherein the compound of formula 1 is:
Figure US20210403444A1-20211230-C00151
15. A method of treating or preventing an RSV infection, comprising administering to a patient in need thereof, an effective amount of a compound of Formula 1:
Figure US20210403444A1-20211230-C00152
or a pharmaceutically acceptable salt thereof, wherein
X is selected from O, S, or NR7;
z is selected from 0, 1, 2, 3, 4, 5, and 6;
y is selected from 0, 1, 2, 3, 4, 5, and 6;
R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, C(═O)Ra, —C(═O)ORa, C(═O)N(Ra)2, C(═O)SRa;
R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein when multiple Ra groups are present, said Ra groups may together form a ring;
R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
wherein either of R3 and R4, or R5 and R6 may together form a double bond
wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin; and
wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring; and
wherein Ra is in each case independently selected from hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl.
16. The method of treating or preventing an RSV infection according to claim 15 wherein the compound is administered via a route of administration selected from the group consisting of buccal, oral, intravenous, inhalation, intradermal, intramuscular, topical, subcutaneous, rectal, vaginal, parenteral, pulmonary, intranasal, and ophthalmic.
17. The method of treating or preventing an RSV infection according to claim 15 wherein the compound of Formula 1 is selected from the group consisting of:
Figure US20210403444A1-20211230-C00153
18. The method of treating or preventing an RSV infection according to claim 15 wherein the compound of Formula 1 is:
Figure US20210403444A1-20211230-C00154
19. A compound of Formula 1:
Figure US20210403444A1-20211230-C00155
or a pharmaceutically acceptable salt thereof, wherein
X is selected from O, S, or NR7;
z is selected from 0, 1, 2, 3, 4, 5, and 6;
y is selected from 0, 1, 2, 3, 4, 5, and 6;
R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, C(═O)Ra, C(═O)ORa, C(═O)N(Ra)2, C(═O)SRa;
R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein when multiple Ra groups are present, said Ra groups may together form a ring;
R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
wherein either of R3 and R4, or R5 and R6 may together form a double bond
wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin; and
wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring; and
wherein Ra is in each case independently selected from hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl.
20. The compound according to claim 19 wherein the compound is selected from the group consisting of:
Figure US20210403444A1-20211230-C00156
21. A pharmaceutical composition comprising the compound of claim 19 and a pharmaceutically acceptable carrier, vehicle, or excipient.
22. A pharmaceutical composition comprising a compound of Formula 1:
Figure US20210403444A1-20211230-C00157
or a pharmaceutically acceptable salt thereof, wherein
X is selected from O, S, or NR7;
z is selected from 0, 1, 2, 3, 4, 5, and 6;
y is selected from 0, 1, 2, 3, 4, 5, and 6;
R1 and R2 are independently selected from Ra, ORa, N(Ra)2, SRa, C(═O)Ra, C(═O)ORa, C(═O)N(Ra)2, C(═O)SRa;
R3, R4, R5, and R6 are independently selected from independently selected from —Ra, —ORa, —N(Ra)2, —SRa, —SO2Ra, —SO2N(Ra)2; —C(O)Ra, OC(O)Ra, —COORa, —C(O)N(Ra)2, —OC(O)N(Ra)2, —N(Ra)C(O), —N(Ra)C(O)N(Ra)2, —F, —Cl, —Br, —I, —CN, —NO2; wherein when multiple Ra groups are present, said Ra groups may together form a ring;
R7, when present, is Ra, C(O)Ra, SO2Ra, COORa, C(O)N(Ra)2,
wherein either of R3 and R4, or R5 and R6 may together form a double bond
wherein either of R4 and R6 or R3 and R5 may together form a carbonyl, imine or olefin; and
wherein any of two or more of R1, R2, R3, R4, R5, R6 or R7, or Ra groups may together form a ring; and
wherein Ra is in each case independently selected from hydrogen, C1-8 alkyl, C3-8 cycloalkyl, C2-8 heterocyclyl, C6-12 aryl, C3-12 heteroaryl, C1-8 alkyl-C3-8 cycloalkyl, C1-8 alkyl-C2-8 heterocyclyl, C1-8 alkyl-C6-12 aryl, and C1-8 alkyl-C3-12 heteroaryl;
and a pharmaceutically acceptable carrier, vehicle, or excipient.
23. The pharmaceutical composition according to claim 22 wherein the compound is selected from the group consisting of:
Figure US20210403444A1-20211230-C00158
24. The pharmaceutical composition according to claim 22 wherein the compound is:
Figure US20210403444A1-20211230-C00159
US17/370,586 2016-05-10 2021-07-08 Heterocyclic derivatives for the treatment of rsv Abandoned US20210403444A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/370,586 US20210403444A1 (en) 2016-05-10 2021-07-08 Heterocyclic derivatives for the treatment of rsv

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662333996P 2016-05-10 2016-05-10
PCT/US2017/031945 WO2017196970A1 (en) 2016-05-10 2017-05-10 Heterocyclic derivatives for the thratment of rsv
US201816099957A 2018-11-08 2018-11-08
US17/370,586 US20210403444A1 (en) 2016-05-10 2021-07-08 Heterocyclic derivatives for the treatment of rsv

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/031945 Division WO2017196970A1 (en) 2016-05-10 2017-05-10 Heterocyclic derivatives for the thratment of rsv
US16/099,957 Division US11084796B2 (en) 2016-05-10 2017-05-10 Heterocyclic derivatives for the treatment of RSV

Publications (1)

Publication Number Publication Date
US20210403444A1 true US20210403444A1 (en) 2021-12-30

Family

ID=60267422

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/099,957 Active US11084796B2 (en) 2016-05-10 2017-05-10 Heterocyclic derivatives for the treatment of RSV
US17/370,586 Abandoned US20210403444A1 (en) 2016-05-10 2021-07-08 Heterocyclic derivatives for the treatment of rsv

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/099,957 Active US11084796B2 (en) 2016-05-10 2017-05-10 Heterocyclic derivatives for the treatment of RSV

Country Status (5)

Country Link
US (2) US11084796B2 (en)
EP (1) EP3454896A4 (en)
CN (1) CN109641042B (en)
CA (1) CA3023922C (en)
WO (1) WO2017196970A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3023922C (en) * 2016-05-10 2023-04-18 Georgia State University Research Foundation, Inc. Heterocyclic derivatives for the treatment of rsv
US10847705B2 (en) 2018-02-15 2020-11-24 Intel Corporation Reducing crosstalk from flux bias lines in qubit devices
US11177912B2 (en) 2018-03-06 2021-11-16 Intel Corporation Quantum circuit assemblies with on-chip demultiplexers
US11355623B2 (en) 2018-03-19 2022-06-07 Intel Corporation Wafer-scale integration of dopant atoms for donor- or acceptor-based spin qubits
US11183564B2 (en) 2018-06-21 2021-11-23 Intel Corporation Quantum dot devices with strain control
US11616126B2 (en) 2018-09-27 2023-03-28 Intel Corporation Quantum dot devices with passive barrier elements in a quantum well stack between metal gates
US11699747B2 (en) 2019-03-26 2023-07-11 Intel Corporation Quantum dot devices with multiple layers of gate metal
US11682701B2 (en) 2019-03-27 2023-06-20 Intel Corporation Quantum dot devices
US11387324B1 (en) 2019-12-12 2022-07-12 Intel Corporation Connectivity in quantum dot devices
CN116425754A (en) * 2021-12-30 2023-07-14 苏州爱科百发生物医药技术有限公司 Aromatic heterobicyclic compounds and antiviral uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11084796B2 (en) * 2016-05-10 2021-08-10 Georgia State University Research Foundation, Inc. Heterocyclic derivatives for the treatment of RSV

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2841582A (en) 1958-07-01 Aminoalkyl esters of
US3767653A (en) 1971-06-28 1973-10-23 Squibb & Sons Inc Thiazines
US4397849A (en) * 1980-12-30 1983-08-09 Fujisawa Pharmaceutical Co., Ltd. Benzothiazine derivatives
US6774134B2 (en) 2000-12-20 2004-08-10 Bristol-Myers Squibb Company Heterocyclic substituted 2-methyl-benzimidazole antiviral agents
JP4196678B2 (en) 2001-04-26 2008-12-17 味の素株式会社 Heterocyclic compounds
AU2003225705A1 (en) 2003-03-07 2004-09-30 Ribapharm Inc. Cytidine analogs and methods of use
CN101723949B (en) * 2003-12-24 2013-10-23 生物区科学管理控股有限公司 Polycyclic agents for treatment of respiratory syncytial virus infections
CN101443320A (en) * 2006-03-14 2009-05-27 阿斯利康(瑞典)有限公司 Bezothiazol derivatives as beta2 adrenoreceptor agonists
EP2002835A1 (en) 2007-06-04 2008-12-17 GenKyo Tex Pyrazolo pyridine derivatives as NADPH oxidase inhibitors
DK2464664T3 (en) * 2009-08-13 2016-01-18 Crucell Holland Bv ANTIBODIES AGAINST HUMAN RESPIRATORY SYNCYTIAL VIRUS (RSV) AND METHODS FOR USING IT
US10100372B2 (en) 2014-06-18 2018-10-16 Georgia State University Research Foundation, Inc. Recombinant RSV reporter virus

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11084796B2 (en) * 2016-05-10 2021-08-10 Georgia State University Research Foundation, Inc. Heterocyclic derivatives for the treatment of RSV

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Ryabokon et al. Khimiko-Farmatsevticheskii Zhurnal (1975), 9(4), 15-18. English translation of this Russian Journal submitted (pages 227-230, 4 pages submitted) (Year: 1975) *

Also Published As

Publication number Publication date
CN109641042B (en) 2023-06-09
CA3023922A1 (en) 2017-11-16
EP3454896A1 (en) 2019-03-20
CN109641042A (en) 2019-04-16
CA3023922C (en) 2023-04-18
US11084796B2 (en) 2021-08-10
US20190135770A1 (en) 2019-05-09
EP3454896A4 (en) 2020-03-25
WO2017196970A1 (en) 2017-11-16

Similar Documents

Publication Publication Date Title
US20210403444A1 (en) Heterocyclic derivatives for the treatment of rsv
DK2776416T3 (en) Pharmaceutical compounds
TWI515193B (en) Compounds for treating respiratory syncytial virus infections
CN114057702B (en) Novel inhibitor of coronavirus main protease and preparation method and application thereof
US11384090B2 (en) Spiropyrrolidine derived antiviral agents
US11352363B1 (en) Spiropyrrolidine derived antiviral agents
JP2007516983A (en) Polycyclic drugs for the treatment of respiratory syncytial virus infection
CN115960088A (en) Novel coronavirus main protease inhibitor, preparation method and application thereof
JP7031002B2 (en) Crystal form, salt type and method for producing the pyridinoimidazole compound
KR20200051646A (en) AHR inhibitors and uses thereof
US10906899B2 (en) Bicyclic fused pyrazole derivatives for the treatment of RSV
CN111675661B (en) Diaryl pyrimidine HIV-1 reverse transcriptase inhibitor containing trans double bond and preparation method and application thereof
CN112771048A (en) Inhibitors of influenza virus replication and intermediates and uses thereof
CN114014866A (en) 5, 7-dihydrofuro [3,4-d ] pyrimidine compound and preparation method and application thereof
ES2905650T3 (en) pharmaceutical compounds
EP3753942B1 (en) Crystal form of 3,4-dihydrothieno[3,2-d]pyrimidine compound and preparation method therefor
CN111465600A (en) Pharmaceutical compounds
CN115141206A (en) Alpha-lipoic acid lycorine conjugate and preparation method and application thereof
CN116621918A (en) Spiro compound and preparation method and application thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION