CN117881403A - Thionucleosides as antiviral agents - Google Patents

Thionucleosides as antiviral agents Download PDF

Info

Publication number
CN117881403A
CN117881403A CN202280041407.1A CN202280041407A CN117881403A CN 117881403 A CN117881403 A CN 117881403A CN 202280041407 A CN202280041407 A CN 202280041407A CN 117881403 A CN117881403 A CN 117881403A
Authority
CN
China
Prior art keywords
alkyl
optionally substituted
substituted
cycloalkyl
unsubstituted
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.)
Pending
Application number
CN202280041407.1A
Other languages
Chinese (zh)
Inventor
雷蒙德·希纳兹
弗兰克·安布拉德
陈哲
基万·赞迪
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.)
Emory University
Original Assignee
Emory University
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 Emory University filed Critical Emory University
Priority claimed from PCT/US2022/024290 external-priority patent/WO2022217155A2/en
Publication of CN117881403A publication Critical patent/CN117881403A/en
Pending legal-status Critical Current

Links

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compounds, compositions and methods for preventing, treating or curing coronavirus infections in a human subject or other animal host. In one embodiment, the compounds can be used to treat infections with severe acute respiratory syndrome viruses such as human coronavirus 229E, SARS, MERS, SARS-CoV-1, OC43, and SARS-CoV-2. In another embodiment, the method is for treating a patient infected with a flavivirus, picornavirus, togavirus or bunyavirus.

Description

Thionucleosides as antiviral agents
Technical Field
Compounds, methods and compositions for treating or preventing coronavirus infections are disclosed. More specifically, certain nucleoside and nucleotide analogs, pharmaceutically acceptable salts or other derivatives thereof, and their use in the treatment of coronaviruses, particularly SARS-CoV-2, are disclosed.
Background
Coronaviruses are viruses belonging to the subfamily coronaviridae, the family coronaviridae, and are enveloped viruses having a positive-stranded single-stranded RNA genome and a helically symmetric nucleocapsid.
Coronaviruses primarily infect the upper respiratory and gastrointestinal tract of mammals and birds, but several known strains can also infect humans. Coronaviruses are believed to account for a significant proportion of all common colds in adults and children.
Coronaviruses cause cold in humans, mainly in winter and early spring. Coronaviruses can also cause pneumonia (direct viral pneumonia or secondary bacterial pneumonia), bronchitis (direct viral bronchitis or secondary bacterial bronchitis), and Severe Acute Respiratory Syndrome (SARS).
Coronaviruses can also cause a range of diseases in farm animals and domestic pets, some of which can be severe and pose a threat to the agricultural product processing industry. In chickens, infectious Bronchitis Virus (IBV) is a coronavirus, directed not only to the respiratory tract but also to the genitourinary tract. The virus can be transmitted to different organs of chicken.
Coronaviruses of economic importance to farm animals include porcine coronavirus (transmissible gastroenteritis coronavirus, TGE) and bovine coronavirus, both of which cause diarrhea in young animals. Feline coronavirus: there are two forms of feline enterocoronavirus, a less clinically significant pathogen, but spontaneous mutation of the virus can lead to Feline Infectious Peritonitis (FIP), a disease associated with high mortality. Canine coronavirus (CCoV) is of two types, one of which causes mild gastrointestinal disease and the other of which has been found to cause respiratory disease. Mouse Hepatitis Virus (MHV) is a coronavirus that causes murine epidemics and has high mortality, especially in laboratory mouse communities.
Certain strains of MHV cause progressive demyelinating encephalitis in mice that have been used as a mouse model of multiple sclerosis.
Recently, coronavirus pandemics pose a double threat to health and economy in the united states and the world. Covd-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Common symptoms of the disease include fever (88%), dry cough (68%), shortness of breath (19%) and loss of sense of smell (15% to 30%). Complications may include pneumonia, viral sepsis, acute respiratory distress syndrome, diarrhea, kidney disease, heart problems, and encephalitis. By 1 month 2022, the total number of global infections exceeds 3.06 million, at least 550 tens of thousands die, and nearly 6000 tens of thousands of coronaviruses tested positive in the united states and above based on the university of john hopkins coronavirus resource center. Eighty thousand people have died from the disease. More than 200 countries have recorded local transmission of the disease. Risk factors include travel and virus exposure, social distance and isolation help prevent.
Current treatments for these infections are primarily supportive, minimizing symptoms, rather than treating potential viral infections. For example, patients may be treated with analgesics to relieve pain, and complications of enteroviral heart disease patients such as cardiac arrhythmias, pericardial effusion and heart failure may be treated.
It would be advantageous to provide new antiviral agents, compositions comprising these agents, and methods of treatment using these agents to treat coronaviruses. The present disclosure provides such agents, compositions and methods.
Disclosure of Invention
The present disclosure relates to compounds, methods, and compositions for treating or preventing coronavirus and/or other viral infections in a host. The method comprises administering a therapeutically or prophylactically effective amount of at least one compound described herein to treat or prevent a coronavirus infection or other viral infection (including but not limited to SARS-CoV-2, MERS, SARS and OC-43), or an amount sufficient to reduce the biological activity of the virus. In other embodiments, the compounds described herein are useful for treating or preventing flaviviridae (flavoviruses), picornaviridae (Picornaviridae), togaviridae (togaviridae), and Bunyaviridae (Bunyaviridae).
In one embodiment, the present disclosure relates to methods of using potent selective antiviral agents to target coronaviruses and other viral infections and thus help eliminate and/or treat infections in patients infected with these viruses.
In one aspect of this embodiment, the compound used comprises one or more of the specific nucleoside inhibitors described herein.
In another aspectIn one embodiment, a pharmaceutical composition comprising one or more compounds described herein, which in one embodiment comprises a combination of cytidine and uridine analogs, and a pharmaceutically acceptable carrier or excipient, is disclosed. These compositions are useful for treating a host infected with coronavirus or other virus, preventing one of these infections, and/or reducing the biological activity of one of these viruses. The composition may comprise a combination of one or more compounds described herein, optionally together with other antiviral compounds or biological agents, including anti-SARS-CoV 2 compounds and biological agents, fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides (e.g., redeSivir), GS-441524, N 4 Hydroxycytidine and other compounds disclosed in U.S. patent No.9,809,616 and prodrugs thereof), viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV-2 specific human monoclonal antibodies including CR3022, and agents of different or unknown mechanisms.
In yet another embodiment, the present disclosure relates to a method for preparing a particular nucleoside compound described herein.
In some embodiments, the compounds described herein are deuterated at one or more positions. When the compound is a nucleoside, deuteration may be present at any position at one or more positions on the sugar portion of the compound, the base portion of the compound, and/or the prodrug portion of the compound.
In some embodiments, the ester prodrug is prepared to allow more of the drug to reach the plasma upon oral administration and not become trapped in the gut as a triphosphate.
In another embodiment, ester prodrugs are prepared to enhance the oral bioavailability of the drug.
The disclosure will be better understood with reference to the following detailed description.
Drawings
FIG. 1 is a graph showing plasma levels over time following IV administration (15 mg/kg) of compound 8.
Detailed Description
The compounds described herein show inhibitory activity against the coronaviridae family in a cell-based assay. Thus, the compounds are useful for treating or preventing coronaviridae infections in a host, or for reducing the biological activity of a virus. The host may be a mammal, in particular a human, infected with a virus of the coronaviridae family. The compounds are also effective against flaviviridae, picornaviridae, togaviridae and bunyaviridae viruses. The method comprises administering an effective amount of one or more compounds described herein.
Also disclosed are pharmaceutical formulations comprising one or more compounds described herein in combination with a pharmaceutically acceptable carrier or excipient. In one embodiment, the formulation comprises at least one compound described herein and at least one additional therapeutic agent.
The present disclosure will be better understood with reference to the following definitions.
I. Definition of the definition
The term "independently" is used herein to mean that independently applied variables vary independently from application to application. Thus, in a compound such as R "XYR", where R "is independently carbon or nitrogen, two R" may be carbon, two R "may be nitrogen, or one R" may be carbon and the other R "may be nitrogen.
As used herein, the term "enantiomerically pure" refers to a compound composition comprising at least about 95%, preferably about 97%, 98%, 99% or 100% of a single enantiomer of the compound.
As used herein, the term "substantially free" or "substantially absent" refers to a compound composition comprising at least 85 to 90 wt%, preferably 95 to 98 wt%, even more preferably 99 to 100 wt% of the specified enantiomer of the compound. In a preferred embodiment, the compounds described herein are substantially free of enantiomers.
Similarly, the term "isolated" refers to a compound composition comprising at least 85 to 90 wt%, preferably 95 to 98 wt%, even more preferably 99 to 100 wt% of the compound, the remainder comprising other chemicals or enantiomers.
The term "alkyl" as used herein, unless otherwise indicated, refers to saturated, straight, branched or cyclic primary, secondary or tertiary hydrocarbons, including substituted and unsubstituted alkyl groups. The alkyl groups may be optionally substituted with any moiety that does not interfere with the reaction or provide process improvements, including but not limited to halo, C 1-6 Haloalkyl, hydroxy, carboxyl, C 1-6 Acyl, aryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-6 -alkylamino, arylamino, C 1-6 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrazine, carbamate, phosphonic acid, phosphonate, as known to those skilled in the art, unprotected or protected as desired, e.g., as taught in: greene et al, Protective Groupsin Organic SynthesisJohn Wiley and Sons, second edition, 1991, incorporated herein by reference. Specifically include CF 3 And CH (CH) 2 CF 3
In this document, whenever the term C (alkyl range) is used, the term includes each member of the class independently as if specifically and individually set forth. The term "alkyl" includes C 1-22 An alkyl moiety, and the term "lower alkyl" includes C 1-6 An alkyl moiety. Those of ordinary skill in the art will appreciate that the relevant alkyl groups are named by replacing the suffix "-alkane" with the suffix "-base".
As used herein, "bridged alkyl" refers to a bicycloalkane or tricycloalkane, such as 2:1:1 bicyclohexane.
As used herein, "spiroalkyl" refers to two rings attached at a single (quaternary) carbon atom.
The term "alkenyl" refers to an unsaturated straight or branched hydrocarbon group, provided that it contains one or more double bonds. The alkenyl groups disclosed herein may be optionally substituted with any moiety that does not adversely affect the reaction process, including but not limited to those moieties described for substituents on the alkyl moiety. Non-limiting examples of alkenyl groups include ethylene, methylethylene, isopropylene, 1, 2-ethane-diyl, 1-ethane-diyl, 1, 3-propane-diyl, 1, 2-propane-diyl, 1, 3-butane-diyl, and 1, 4-butane-diyl.
The term "alkynyl" refers to an unsaturated straight or branched chain acyclic hydrocarbon group, so long as it contains one or more triple bonds. Alkynyl groups may be optionally substituted with any moiety that does not adversely affect the reaction process, including but not limited to those described for alkyl moieties. Non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, hydroxypropionyl, butyn-1-yl, butyn-2-yl, pentyn-1-yl, pentyn-2-yl, 4-methoxypentan-2-yl, 3-methylbutan-1-yl, hexyn-2-yl and hexyn-3-yl, 3-dimethylbutyyn-1-yl.
The term "alkylamino" or "arylamino" refers to an amino group having one or two alkyl or aryl substituents, respectively.
The term "fatty alcohol" as used herein refers to a linear primary alcohol having from 4 to 26 carbons in the chain, preferably from 8 to 26 carbons in the chain, most preferably from 10 to 22 carbons in the chain. The exact chain length will vary from source to source. Representative fatty alcohols include lauryl alcohol, stearyl alcohol and oleyl alcohol. They are colorless oily liquids (for smaller carbon numbers) or waxy solids, although impure samples may be yellow in color. Fatty alcohols typically have an even number of carbon atoms and a single alcohol group (-OH) attached to the terminal carbon. Some are unsaturated and some are branched. They are widely used in industry. As with fatty acids, they are generally collectively referred to by the number of carbon atoms in the molecule, e.g. "C 12 Alcohols ", i.e. alcohols having 12 carbons, such as dodecanol.
The term "protected" as used herein, unless otherwise defined, refers to a group that is added to an oxygen, nitrogen, or phosphorus atom to prevent further reaction or for other purposes. Various oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis and are described, for example, in Greene et al, protective Groups in Organic Synthesis, supra.
The term "aryl", alone or in combination, refers to a carbocyclic aromatic system containing one, two, or three rings, wherein such rings may be linked together in a pendent manner or may be fused. Non-limiting examples of aryl groups include phenyl, biphenyl, or naphthyl, or other aromatic groups that remain after hydrogen is removed from the aromatic ring. The term aryl includes both substituted and unsubstituted moieties. Aryl groups may be optionally substituted with any moiety that does not adversely affect the reaction process, including but not limited to those described for alkyl moieties. Non-limiting examples of substituted aryl groups include heteroarylamino, N-aryl-N-alkylamino, N-heteroarylamino-N-alkylamino, heteroarylalkoxy, arylamino, aralkylamino, arylthio, monoarylamidosulfonyl, arylsulfonamido, diarylaminosulfonyl, monoarylamidosulfonyl, arylsulfinyl, arylsulfonyl, heteroarylthio, heteroarylsulfinyl, heteroarylsulfonyl, aroyl, heteroarylacyl, aralkanoyl, heteroarylalkanoyl, hydroxyarylalkyl, hydroxyheteroarylalkyl, haloalkoxyalkyl, aryl, aralkyl, aryloxy, aralkoxy, aryloxyalkyl, saturated heterocyclyl, partially saturated heterocyclyl, heteroaryl, heteroaryloxy, heteroaryloxyalkyl, arylalkyl, heteroarylalkyl, arylalkenyl, and heteroarylalkenyl, carboaralkoxy.
The term "alkylaryl" or "alkylaryl" refers to an alkyl group having an aryl substituent. The term "aralkyl" or "arylalkyl" refers to an aryl group having an alkyl substituent.
The term "halo" as used herein includes chloro, bromo, iodo and fluoro.
The term "acyl" refers to a carboxylic acid ester wherein the non-carbonyl moiety of the ester group is selected from the group consisting of: straight, branched or cyclic alkyl or lower alkyl, alkoxyalkyl (including but not limited to methoxymethyl), aralkyl (including but not limited to benzyl), aryloxyalkyl (e.g., phenoxymethyl), aryl (including but not limited to phenyl, optionally halogenated(F, cl, br or I), alkyl (including but not limited to C 1 、C 2 、C 3 And C 4 ) Or alkoxy (including but not limited to C 1 、C 2 、C 3 And C 4 ) Substituted), sulfonates (e.g., alkyl or aralkylsulfonyl, including but not limited to methylsulfonyl), mono-, di-or triphosphate, trityl or monomethoxytrityl, substituted benzyl, and trialkylsilyl (e.g., dimethyl t-butylsilyl or diphenylmethylsilyl). The aryl group in the ester preferably comprises a phenyl group. The term "lower acyl" refers to an acyl group wherein the non-carbonyl moiety is lower alkyl.
The terms "alkoxy" and "alkoxyalkyl" encompass straight or branched chain oxygen containing groups having an alkyl moiety, such as methoxy. The term "alkoxyalkyl" also encompasses alkyl groups having one or more alkoxy groups attached to the alkyl group, i.e., forming monoalkoxyalkyl and dialkoxyalkyl groups. The "alkoxy" group may be further substituted with one or more halogen atoms (e.g., fluorine, chlorine, or bromine) to provide a "haloalkoxy" group. Examples of such groups include fluoromethoxy, chloromethoxy, trifluoromethoxy, difluoromethoxy, trifluoroethoxy, fluoroethoxy, tetrafluoroethoxy, pentafluoroethoxy and fluoropropoxy.
The term "alkylamino" means "mono-alkylamino" and "dialkylamino" containing one or two alkyl groups, respectively, attached to an amino group. The term arylamino refers to "monoarylamino" and "diarylamino" groups that contain one or two aryl groups attached to an amino group, respectively. The term "aralkylamino" encompasses aralkyl groups attached to an amino group. The term aralkylamino refers to "monoarylamino" and "diarylamino" containing one or two aralkyl groups, respectively, attached to an amino group. The term aralkylamino also denotes a "monoarylalkylmonoalkylamino" group containing one aralkyl group and one alkyl group attached to the amino group.
The term "heteroatom" as used herein refers to oxygen, sulfur, nitrogen and phosphorus.
The term "heteroaryl" or "heteroaromatic" as used herein refers to an aromatic group that includes at least one sulfur, oxygen, nitrogen, or phosphorus in an aromatic ring.
The terms "heterocycle", "heterocyclyl" and cycloheteroalkyl refer to a non-aromatic cyclic group in which at least one heteroatom, such as oxygen, sulfur, nitrogen or phosphorus, is present in the ring.
Non-limiting examples of heteroaryl and heterocyclyl groups include furyl (furyl), pyridyl, pyrimidinyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, benzofuryl, benzothienyl, quinolinyl, isoquinolinyl, benzothienyl, isobenzofuranyl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2, 4-thiadiazolyl, isoxazolyl, pyrrolyl, quinazolinyl, cinnolinyl, phthalazinyl, xanthinyl, hypoxanthyl, thiophen, furan, pyrrole, polypyrrole, pyrazole, imidazole, 1,2, 3-triazole, 1,2, 4-triazole, oxazole, isoxazole, thiazole, isothiazole, pyrimidine or pyridazine, and pteridinyl, aziridine, thiazole, isothiazole, 1,2, 3-oxadiazole, thiazine, pyridine, pyrazine, piperazine, pyrrolidine, oxazinane (oxazirane), phenazine, phenothiazine, morpholinyl, pyrazolyl, pyridazinyl, pyrazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracil, triazolopyridinyl, imidazopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, adenine, N 6 Alkylpurine, N 6 Benzyl purine, N 6 Halogenated purines, N 6 Vinyl purine, N 6 Alkynylpurine, N 6 -acylpurines, N 6 Hydroxy alkyl purines, N 6 Sulfanyl purine, thymine, cytosine, 6-azapyrimidine, 2-mercaptopyrimidine, uracil, N 5 Alkylpyrimidines, N 5 -benzyl pyrimidine, N 5 -halogenated pyrimidines, N 5 Vinyl pyrimidine, N 5 Alkynopyrimidines, N 5 -acyl pyrimidines, N 5 Hydroxyalkyl purines and N 6 -thioalkyl purines and isoxazolyl. The heteroaromatic group may be optionally substituted as described above for aryl. The heterocyclic or heteroaromatic groups may optionally be interrupted by one or more substituentsA substituent substitution of the group consisting of: halogen, haloalkyl, alkyl, alkoxy, hydroxy, carboxyl derivatives, amido, amino, alkylamino and dialkylamino. The heteroaromatic groups may be partially or fully hydrogenated as desired. As a non-limiting example, dihydropyridines may be used in place of pyridines. Functional oxygen and nitrogen groups on the heterocycle or heteroaryl may be protected as needed or desired. Suitable protecting groups are well known to those skilled in the art and include trimethylsilyl, dimethylhexylsilyl, t-butyldimethylsilyl and t-butyldiphenylsilyl, trityl or substituted trityl, alkyl, acyl groups, such as acetyl and propionyl, methanesulfonyl and p-toluenesulfonyl. The heterocyclic or heteroaromatic group may be optionally substituted with any moiety that does not adversely affect the reaction, including but not limited to those moieties described above for aryl groups.
The term "host" as used herein refers to a single-or multicellular organism in which a virus can replicate, including but not limited to cell lines and animals, preferably humans. Alternatively, the host may carry a portion of the viral genome, the replication or function of which may be altered by the compounds described herein. The term host refers specifically to infected cells, cells transfected with all or part of the viral genome, and animals, particularly primates (including but not limited to chimpanzees) and humans. In most animal applications of the present disclosure, the host is a human. However, the present disclosure expressly contemplates veterinary applications in certain indications (e.g., for treating chimpanzees).
The term nucleoside also includes ribonucleosides, representative ribonucleosides are disclosed, for example, in Journal of Medicinal Chemistry,43 (23), 4516-4525 (2000), antimicrobial Agents and Chemotherapy,45 (5), 1539-1546 (2001), and PCT WO 2000069876.
The term "peptide" refers to a natural or synthetic compound containing two to one hundred amino acids linked to the amino group of one amino acid through the carboxyl group of another amino acid.
The term "pharmaceutically acceptable salt or prodrug" is used throughout the specification to describe any pharmaceutically acceptable form (e.g., ester) of a compound that provides the compound when administered to a patient. Pharmaceutically acceptable salts include salts derived from pharmaceutically acceptable inorganic or organic bases and inorganic or organic acids. Suitable salts include those derived from alkali metals (e.g., potassium and sodium), alkaline earth metals (e.g., calcium and magnesium), and many other acids well known in the pharmaceutical arts.
Pharmaceutically acceptable prodrugs refer to compounds which are metabolized (e.g., hydrolyzed or oxidized) in the host to form the compounds described herein. Typical examples of prodrugs include compounds having a biostable protecting group on the functional moiety of the active compound. Prodrugs include compounds that may be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, or dephosphorylated to produce the active compound. The prodrug forms of the compounds described herein may have antiviral activity, may be metabolized to form compounds that exhibit such activity, or both.
Active compounds
In one embodiment, the compound is a compound of formula (a):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R 1 is H, deuterium, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 2-8 Alkynyl or N 3
R 2 And R is 2’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR 7 And SR (Surfural) 7
Each R 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
provided that R is 2 And R is 2’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy, carboxyl, C 1-12 Acyl, aryl, heteroaryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, alkyl sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphono, phosphinyl, phosphoryl,Phosphines, thioesters, thioethers, acid halides, anhydrides, oximes, hydrazines, carbamates, phosphonic acids, phosphonates, boric acid and borates;
R 3 and R is 3’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR 7 And SR (Surfural) 7 Wherein each R is 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy,Carboxyl, C 1-12 Acyl, aryl, heteroaryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrazine, carbamate, phosphonic acid, phosphonate, boric acid, and borate;
Provided that R is 3 And R is 3’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R 4 Selected from the group consisting of: H. deuterium, CN, halo, N 3 Substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl, substituted or unsubstituted (C 1-8 ) Haloalkyl and N 3
R 5 Is and R 5’ H, CH independently 3 、CH 2 F、CHF 2 Or CF (CF) 3 Wherein when R is 5 Is CH 3 When it is attached to carbon, it may be all or part of R or S or any mixture thereof, or R 5 And R is 5’ Can be combined to form C 3-7 A cycloalkyl ring;
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (N) R ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group;
the base is selected from the group consisting of:
y is H or a halogen group,
x is N or CH, and the number of the N or the CH,
w is O or S, and the R is selected from the group consisting of O and S,
X 1 and X 1’ Is independently CH, C- (C) 1-6 ) Alkyl, C- (C) 2-6 ) Alkenyl, C- (C) 2-6 ) Alkynyl, C- (C) 3-7 ) Cycloalkyl, C- (C) 1-6 ) Haloalkyl, C- (C) 1-6 ) Hydroxyalkyl, C-OR 22 、C-N(R 22 ) 2 C-halo, C-CN or N,
X 2 and X 2’ Independently H, halo, OR 9’ Or NR (NR) 10 R 10’
R 9’ H, L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl) esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
R 10 and R is 10’ Independently are H, OH, L-amino acid amide, D-amino acid amide, (acyloxybenzyl) amine, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, PEG amide, PEG carbamate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', lipid amide, optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Or lipid carbamates wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy) with the proviso that R 10 And R is 10’ And cannot be OH.
In another embodiment, the compound is a compound of formula B:
Or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 2 、R 2’ 、R 3 、R 4 、R 5 R 5’ 、R 7 And R is 8 As defined in formula A, A is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkyne, carbon chain derived from fatty alcohol orC substituted below 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acids R 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl group, and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
In another embodiment, the compound is a compound of formula C:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 2 、R 2’ 、R 3 And R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein when chiral at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or tri-phosphate, wherein, when chiral is present at the phosphorus centre When it is complete or partial R p Or S p Or any mixture thereof, or a mixture of any of them,
R 7 is an L-amino acid ester, a D-amino acid ester, an N-substituted L-amino acid ester, an N-substituted D-amino acid ester, an N, N-disubstituted L-amino acid ester, an N, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, an optionally substituted bis-acyloxybenzyl ester, an optionally substituted (acyloxybenzyl) ester, an optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 Cycloalkyl group, and
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 Wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
In another embodiment, the compound is a compound of formula D:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is selected from the group consisting of:
and is also provided with
X 1 、X 1’ 、X 2’ 、X 2 、R 2 、R 2’ 、R 3 、R 3’ 、R 4 、R 5 、R 5’ And R is 6 As defined in formula a.
In another embodiment, the compound is a compound of formula E:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is selected from the group consisting of:
/>
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 、R 4 、R 5 and R is 5’ As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl or substitutedHeteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl group, and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
In another embodiment, the compound is a compound of formula F:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is selected from the group consisting of:
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 and R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein, when present at the phosphorus centreIn the case of chirality, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluorine、C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
In one embodiment of the compounds of formula A, R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
In one embodiment of the compounds of formula B, wherein R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
In one embodiment of the compounds of formula C, R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H.
In one embodiment of the compounds of formula D, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
In one embodiment of the compounds of formula E, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
In one embodiment of the compounds of formula F, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H
In one embodiment of the compounds of formula A, R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
In one embodiment of the compound of formula BIn the scheme, R 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
In one embodiment of the compounds of formula C, R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
In one embodiment of the compounds of formula D, R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
In one embodiment of the compounds of formula E, R 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
In one embodiment of the compounds of formula F, R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
Representative compounds of formula a include the following:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt or prodrug thereof.
Representative compounds of formula B include the following:
/>
/>
or a pharmaceutically acceptable salt or prodrug thereof.
Representative compounds of formula D include the following:
or a pharmaceutically acceptable salt or prodrug thereof.
In any of these embodiments, the compound can exist in the β -D or β -L configuration.
III stereoisomers and polymorphs
The compounds described herein may have asymmetric centers and exist as racemates, racemic mixtures, individual diastereomers or enantiomers, all isomeric forms being encompassed in the present disclosure. The compounds described herein having chiral centers may exist and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. The present disclosure encompasses racemic, optically-active, polymorphic, or stereoisomeric forms, or mixtures thereof, of the compounds described herein, which possess the useful properties described herein. The optically active forms can be prepared, for example, by resolution of the racemic forms by recrystallization techniques, by synthesis from optically active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase, or by enzymatic resolution. One can purify the corresponding compound and then derivatize the compound to form the compound described herein, or purify the compound itself.
Optically active forms of the compounds can be prepared using any method known in the art, including, but not limited to, resolution of the racemic form by recrystallization techniques, synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase.
Examples of the method of obtaining an optically active material include at least the following methods.
i)Physical separation of crystals: techniques for manual separation of macroscopic crystals of individual enantiomers. This technique can be used if crystals of individual enantiomers are present, i.e. the material is an aggregate, and the crystals are visually distinct;
ii)simultaneous crystallization:a technique for crystallizing individual enantiomers from a solution of racemates separately, which is only possible when the latter are solid aggregates;
iii)enzymatic resolution:techniques for partially or completely separating racemates by differential reaction rates of enantiomers with enzymes;
iv)enzymatic asymmetric synthesis:a synthesis technique wherein at least one step of the synthesis uses an enzymatic reaction to obtain enantiomerically pure or enriched synthesis precursors of the desired enantiomer;
v)chemical asymmetric synthesis: A synthesis technique for synthesizing a desired enantiomer from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which can be accomplished using chiral catalysts or chiral auxiliary;
vi)diastereoisomeric separation:techniques for converting individual enantiomers to diastereomers by reacting a racemic compound with an enantiomerically pure reagent (chiral auxiliary). Then separating them according to the resulting diastereoisomers, which are reflected in more pronounced structural differences, using chromatography or crystallization, followed by removal of the chiral auxiliary to obtain the desired enantiomer;
vii)primary and secondary asymmetric transformations:a technique by which diastereomers in the racemate equilibrate to produce advantages in solutions of the diastereomers in the desired enantiomer or preferential crystallization of the diastereomers in the desired enantiomer upsets the equilibrium so that in principle all material is converted from the desired enantiomer to the crystalline diastereomer. The desired enantiomer is then released from the diastereomer;
viii)kinetic resolution:the technology refers toPartial or complete resolution of the racemate (or further resolution of the partially resolved compound) is accomplished under kinetic conditions by unequal reaction rates of the enantiomer with chiral non-racemic reagents or catalysts;
ix)Enantiospecific synthesis from non-racemic precursors:a synthetic technique by which the desired enantiomer is obtained from an achiral starting material and stereochemical integrity is not compromised or only minimally compromised during synthesis;
x)chiral liquid chromatography:techniques for separation in a liquid mobile phase by different interactions of enantiomers of racemates with a stationary phase (including but not limited to by chiral HPLC). The stationary phase may be made of chiral material or the mobile phase may contain additional chiral material to induce different interactions;
xi)chiral gas chromatography:a technique for volatilizing racemates and separating enantiomers using different interactions in the gaseous mobile phase by means of a chromatographic column containing a fixed non-racemic chiral adsorbent phase;
xii)extraction with chiral solvent:a technique for separating enantiomers by preferentially dissolving one enantiomer in a specific chiral solvent;
xiii)transport across chiral membranes:a technique in which the racemate is contacted with a thin film barrier. The barrier typically separates two miscible fluids, one of which contains racemates, and the driving force of concentration or pressure differential, etc., results in preferential transport across the membrane barrier. Separation occurs due to the non-racemic chiral nature of the membrane, which allows only one enantiomer of the racemate to pass through.
Chiral chromatography is used in one embodiment, including but not limited to simulated moving bed chromatography. A variety of chiral stationary phases are commercially available.
IV salt or prodrug formulations
Where the compound is basic or acidic enough to form a stable non-toxic acid or base salt, it may be appropriate to administer the compound as a pharmaceutically acceptable salt. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids forming physiologically acceptable anions, such as tosylate, mesylate, acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, alpha-ketoglutarate and alpha-glycerophosphate. Suitable inorganic salts may also be formed, including, but not limited to, sulfate, nitrate, bicarbonate, and carbonate. For certain transdermal applications, it may be preferable to use fatty acid salts of the compounds described herein. Fatty acid salts can help penetrate the stratum corneum. Examples of suitable salts include salts of the compounds with stearic acid, oleic acid, linoleic acid, palmitic acid, caprylic acid and capric acid.
Pharmaceutically acceptable salts can be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid that provides a physiologically acceptable anion. Where the compound includes a plurality of amine groups, salts may be formed with any number of amine groups. Alkali metal (e.g., sodium, potassium, or lithium) or alkaline earth metal (e.g., calcium) salts of carboxylic acids may also be prepared.
A prodrug is a pharmacological substance that is administered in an inactive (or significantly reduced) form and subsequently metabolized in vivo to an active metabolite. Allowing more drug to reach the desired target at lower doses is generally the rationale for using prodrugs, often due to better absorption, distribution, metabolism and/or excretion (ADME) properties. Prodrugs are generally intended to increase oral bioavailability and malabsorption in the gastrointestinal tract is often the limiting factor. Furthermore, the use of prodrug strategies can increase the selectivity of a drug for its intended target, thereby reducing the likelihood of off-target effects.
V. therapeutic methods
In one embodiment, the compounds described herein are useful for preventing, treating or curing coronavirus infections, including in particular SARS-CoV2 infections, such as SARS-CoV-2, MERS, SARS and OC-43. In other embodiments, the compounds described herein are useful for preventing, treating, or curing flaviviridae, picornaviridae, togaviridae, and bunyaviridae infections.
The method comprises administering a therapeutically or prophylactically effective amount of at least one compound as described herein to treat, cure, or prevent a coronavirus infection or a flavivirus, picornavirus, togavirus or bunyavirus infection, or an amount sufficient to reduce the biological activity of the virus.
In another embodiment, the compounds described herein are useful for inhibiting coronavirus, flavivirus, picornavirus, togavirus or bunyavirus protease in a cell. The method comprises contacting the cell with an effective amount of a compound described herein.
Hosts, including but not limited to humans infected with coronaviruses, flaviviruses, picornaviruses, togaviruses, or bunyaviruses, or gene fragments thereof, may be treated by administering to a patient an effective amount of an active compound or a pharmaceutically acceptable prodrug or salt thereof in the presence of a pharmaceutically acceptable carrier or diluent. The active substance may be administered by any suitable route, for example orally, parenterally, intravenously, intradermally, transdermally, subcutaneously or topically, in liquid or solid form.
There are a number of species in the genus coronavirus, including but not limited to the middle east respiratory syndrome coronavirus (MERS-CoV), SARS coronavirus (SARS-CoV) and SARS-CoV2. In some embodiments, the compounds described herein may ameliorate and/or treat MERS-CoV infection, SARS-CoV infection, or SARS-CoV2 infection. An effective amount of a compound described herein may be administered to a subject infected with these viruses, and/or by contacting cells infected with these viruses with an effective amount of a compound described herein. In some embodiments, the compounds described herein may inhibit replication of these viruses. In some embodiments, the compounds described herein may ameliorate one or more symptoms of these infections. Symptoms include, but are not limited to, extreme fatigue, malaise, headache, high fever (e.g., >100.4°f), somnolence, confusion, rash, loss of appetite, myalgia, cold, diarrhea, dry cough, runny nose, sore throat, shortness of breath, dyspnea, gradual decline in blood oxygen levels (e.g., hypoxia), and pneumonia.
Some embodiments disclosed herein relate to a method of treating and/or ameliorating an infection caused by a togaviridae virus, which may include administering to a subject an effective amount of one or more compounds described herein or a pharmaceutical composition including a compound described herein. Some embodiments described herein relate to the use of one or more compounds described herein for the manufacture of a medicament for ameliorating and/or treating an infection caused by a togaviridae virus, which may include administering to a subject an effective amount of one or more compounds described herein.
Some embodiments disclosed herein relate to methods of ameliorating and/or treating an infection caused by a togaviridae virus, which may include contacting a cell infected with the virus with an effective amount of one or more compounds described herein or a pharmaceutical composition comprising one or more compounds described herein. Other embodiments described herein relate to the use of one or more compounds described herein for the manufacture of a pharmaceutical togaviridae virus for ameliorating and/or treating an infection caused thereby, which may comprise contacting a cell infected with said virus with an effective amount of said compound.
In some embodiments, the togaviridae virus may be an alphavirus. One species of alphavirus is venezuelan equine encephalitis virus (Venezuelan equine encephalitis virus, VEEV). In some embodiments, the compounds described herein can ameliorate and/or treat VEEV infection. In other embodiments, one or more of the compounds described herein may be manufactured as a medicament for ameliorating and/or treating an infection caused by a VEEV, which may include contacting a cell infected with the virus with an effective amount of the compound. In other embodiments, one or more of the compounds described herein may be used to ameliorate and/or treat an infection caused by VEEV, which may include contacting a cell infected with the virus with an effective amount of the compound. In some embodiments, the VEEV may be an animal epidemic subtype. In some embodiments, the VEEV may be a endemic animal disease subtype. As described herein, venezuelan equine encephalitis virus complex includes multiple subtypes further divided by antigen variant. In some embodiments, the compounds described herein are effective against more than one VEEV subtype, e.g., 2, 3, 4, 5, or 6 subtypes. In some embodiments, the compounds are useful for the treatment, amelioration and/or prevention of VEEV subtype I. In some embodiments, the compounds described herein are effective against more than one VEEV antigen variant. In some embodiments, the compound may ameliorate one or more symptoms of VEEV infection. Examples of symptoms exhibited by subjects infected with VEEV include influenza-like symptoms such as high fever, headache, myalgia, fatigue, vomiting, nausea, diarrhea, and pharyngitis. Subjects with encephalitis showed one or more of the following symptoms: somnolence, convulsions, mental confusion, photophobia, coma, and bleeding in the brain, lungs and/or gastrointestinal tract. In some embodiments, the subject may be a human. In other embodiments, the subject may be a horse.
Chikungunya virus (Chikungunya, CHIKV) is another alphavirus species. In some embodiments, the compounds described herein may ameliorate and/or treat CHIKV infection. In other embodiments, one or more of the compounds described herein may be manufactured as a medicament for ameliorating and/or treating an infection caused by CHIKV, which may include contacting a cell infected with the virus with an effective amount of the compound. In other embodiments, one or more of the compounds described herein may be used to ameliorate and/or treat infections caused by CHIKV, which may include contacting cells infected with the virus with an effective amount of the compound. In some embodiments, one or more symptoms of CHIKV infection may be ameliorated by administering an effective amount of a compound to a subject infected with CHIKV and/or by contacting cells infected with CHIKV with an effective amount of a compound described herein. Clinical symptoms of CHIKV infection include fever, rashes (e.g., petechiae and/or maculopapules), muscle pain, joint pain, fatigue, headache, nausea, vomiting, conjunctivitis, loss of taste, photophobia, insomnia, disabling joint pain, and arthritis.
Other species of alphaviruses include the Ba Ma Senlin virus (Barmah Forest virus), the Ma Yaluo virus (Mayaro virus, MAYV), the Ornithine permanent virus (O 'nyong' nyong virus), the Ross River Virus (RRV), the Semliki forest virus (Semliki Forest virus), the Sindbis virus (SINV), the Wuna virus (Una virus), the eastern equine encephalitis virus (EEE), and the Western Equine Encephalomyelitis (WEE). In some embodiments, one or more of the compounds described herein may be used to ameliorate and/or treat an infection caused by an alphavirus, which may include contacting cells infected with the virus with an effective amount of one or more of the compounds and/or administering an effective amount of one or more of the compounds to a subject (e.g., a subject infected with the virus), wherein the alphavirus may be selected from the group consisting of ba Ma Senlin virus, ma Yaluo virus (MAYV), orney permanent virus, ross River Virus (RRV), semliki forest virus, sindbis virus (SINV), han virus, eastern equine encephalitis virus (EEE), and Western Equine Encephalomyelitis (WEE).
Another genus of coronaviridae is rubella virus. Some embodiments disclosed herein relate to methods of ameliorating and/or treating an infection caused by a rubella virus, which may include contacting a cell infected with the virus with an effective amount of one or more compounds described herein or a pharmaceutical composition including one or more compounds described herein. Other embodiments described herein relate to the use of one or more compounds described herein for the manufacture of a medicament for ameliorating and/or treating an infection caused by a rubella virus, which may include contacting a cell infected with the virus with an effective amount of the compound. Other embodiments described herein relate to one or more compounds described herein that can be used to ameliorate and/or treat an infection caused by a rubella virus by contacting a cell infected with the virus with an effective amount of the compound.
Some embodiments disclosed herein relate to a method of treating and/or ameliorating an infection caused by a bunyaviridae virus, which may include administering to a subject an effective amount of one or more compounds described herein or a pharmaceutical composition including the compounds described herein. Other embodiments disclosed herein relate to a method of treating and/or ameliorating an infection caused by a bunyaviridae virus, which may include administering to a subject identified as suffering from a viral infection an effective amount of one or more compounds described herein or a pharmaceutical composition including the compounds described herein.
Some embodiments disclosed herein relate to methods of ameliorating and/or treating an infection caused by a bunyaviridae virus, which may include contacting a cell infected with the virus with an effective amount of one or more compounds described herein or a pharmaceutical composition comprising one or more compounds described herein. Other embodiments described herein relate to the use of one or more compounds described herein for the manufacture of a medicament for ameliorating and/or treating an infection caused by a bunyaviridae virus, which may comprise contacting a cell infected with the virus with an effective amount of the compound. Other embodiments described herein relate to one or more compounds described herein that are useful for ameliorating and/or treating an infection caused by a bunyaviridae virus by contacting a cell infected with the virus with an effective amount of the compound.
Some embodiments disclosed herein relate to methods of inhibiting replication of a bunyaviridae virus, which may include contacting a cell infected with the virus with an effective amount of one or more compounds described herein or a pharmaceutical composition comprising one or more compounds described herein. Other embodiments described herein relate to the use of one or more compounds described herein for the manufacture of a medicament for inhibiting replication of a bunyaviridae virus, which may comprise contacting a cell infected with the virus with an effective amount of the compound. Other embodiments described herein relate to compounds described herein that are useful for inhibiting bunyaviridae virus replication by contacting a cell infected with the virus with an effective amount of the compound. In some embodiments, the compounds described herein inhibit RNA-dependent RNA polymerase of bunyaviridae viruses, thereby inhibiting RNA replication. In some embodiments, the polymerase of the bunyaviridae virus may be inhibited by contacting a cell infected with the bunyaviridae virus with a compound described herein.
In some embodiments, the bunyaviridae virus may be a bunyavirus. In other embodiments, the bunyaviridae virus may be Hantavirus (hantavir). In other embodiments, the bunyaviridae virus may be a norovirus (Nairovirus). In other embodiments, the bunyaviridae virus may be sand fly virus (Phlebovirus). In some embodiments, the bunyaviridae virus may be an Orthobunyavirus (Orthobunyavirus). In other embodiments, the bunyaviridae virus may be tomato spotted wilt virus (tospirus).
One species of the genus sand fly virus is rift valley fever virus. In some embodiments, the compounds described herein can ameliorate and/or treat rift valley fever virus infection. In other embodiments, one or more of the compounds described herein may be manufactured as a medicament for ameliorating and/or treating an infection caused by a rift valley fever virus, which may include contacting a cell infected with the virus with an effective amount of the compound. In other embodiments, one or more of the compounds described herein may be used to ameliorate and/or treat an infection caused by a rift valley fever virus, which may include contacting a cell infected with the virus with an effective amount of the compound. In some embodiments, a compound described herein can inhibit replication of a rift valley fever virus, wherein the compound is administered to a subject infected with a rift valley fever virus and/or wherein the compound contacts a cell infected with a rift valley fever virus.
In some embodiments, the compounds described herein can improve, treat, and/or inhibit replication of one or more of the ocular form, meningoencephalitis form, or hemorrhagic fever form of the rift valley fever virus. In some embodiments, one or more symptoms of a rift valley fever virus infection may be ameliorated. Symptoms of rift valley fever virus infection include headache, muscle pain, joint pain, neck stiffness, light sensitivity, loss of appetite, vomiting, myalgia, fever, fatigue, back pain, dizziness, weight loss, eye-form symptoms (e.g., retinopathy, blurred vision, vision loss and/or permanent vision loss), meningoepithymia-form symptoms (e.g., severe headache, memory loss, hallucinations, confusion, disorientation, dizziness, convulsions, somnolence and coma), and hemorrhagic fever-form symptoms (e.g., jaundice, hematemesis, hematochezia, purpura rash, ecchymosis, nose and/or gum bleeding, menorrhagia and venipuncture site bleeding).
Another species of sand fly virus is thrombocytopenia syndrome virus. In some embodiments, the compounds described herein may improve, treat, and/or inhibit thrombocytopenia syndrome virus replication. In some embodiments, the compounds may ameliorate and/or treat severe fever with thrombocytopenia syndrome (SFTS). In some embodiments, the compounds described herein can ameliorate one or more symptoms of SFTS. Clinical symptoms include the following: fever, vomiting, diarrhea, multiple organ failure, thrombocytopenia, leukopenia and elevated liver enzyme levels.
Critimia-Congo hemorrhagic fever virus (CCHF) is a species of the genus North. In some embodiments, the compounds described herein may improve, treat, and/or inhibit the replication of crimia-congo hemorrhagic fever virus. Subjects infected with CCHF have one or more of the following symptoms: influenza-like symptoms (e.g., high fever, headache, myalgia, fatigue, vomiting, nausea, diarrhea, and/or pharyngitis), bleeding, mood swings, dysphoria, confusion, throat stagnation, nose bleeding, hematuria, vomiting, black stool, liver swelling and/or pain, disseminated intravascular coagulation, acute renal failure, shock, and acute respiratory distress syndrome. In some embodiments, the compounds described herein can ameliorate one or more symptoms of CCHF.
California encephalitis virus (California encephalitis) is another virus of the bunyaviridae family, a member of the n-bunyavirus. Symptoms of california encephalitis virus infection include, but are not limited to, fever, chills, nausea, vomiting, headache, abdominal pain, somnolence, focal nervous system findings, focal motor abnormalities, paralysis, somnolence, lack of mental alertness and directionality, and seizures. In some embodiments, the compounds described herein can improve, treat, and/or inhibit california encephalitis virus replication. In some embodiments, the compounds described herein can ameliorate one or more symptoms of a california encephalitis virus infection.
Viruses within the Hantavirus genus may cause Hantavirus kidney syndrome Hemorrhagic Fever (HFRS) (caused by viruses such as Hantaan river virus (Hantaan River virus), duobuva-Belglade virus (Dobrava-Belgrad virus), saarema virus (Saarema virus), head virus (Seoul virus) and pramla virus (Puumala virus)) and Hantaan virus lung syndrome (HPS). Viruses that can cause HPS include, but are not limited to, black canal virus (Black Creek Canal virus, BCCV), new York Virus (NYV), xin Nuobu mole virus (Sin Nombre virus, SNV). In some embodiments, the compounds described herein can improve and/or treat HFRS or HPS. Clinical symptoms of HFRS include redness of the cheeks and/or nose, fever, chills, palmar sweating, diarrhea, discomfort, headache, nausea, abdominal and back pain, respiratory problems, gastrointestinal problems, tachycardia, hypooximetry, renal failure, proteinuria and polyuria. Clinical symptoms of HPS include influenza-like symptoms (e.g., cough, myalgia, headache, somnolence, and shortness of breath, potentially worsening to acute respiratory failure). In some embodiments, the compounds described herein can ameliorate one or more symptoms of HFRS or HPS.
Various indicators for determining the effectiveness of methods for treating and/or ameliorating a viral infection of the coronaviridae, togaviridae, hepaciviridae and/or bunyaviridae families are known to those skilled in the art. Examples of suitable indicators include, but are not limited to, a decrease in viral load, a decrease in viral replication, a decrease in serum conversion time (no virus detected in patient serum), a decrease in morbidity or mortality in clinical outcome, and/or other indicators of disease response. Further indicators include one or more overall quality of life health indicators, such as reduced duration of disease, reduced severity of disease, reduced time to resume normal health and normal activity, and reduced time to remission of one or more symptoms. In some embodiments, a compound described herein can cause a reduction, alleviation or positive indication of one or more of the foregoing indicators, as compared to an untreated subject.
VI combination or alternation therapy
In one embodiment, the compounds described herein may be used with at least one other active agent, which may be an antiviral agent. In one aspect of this embodiment, the at least one additional active agent is selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors (e.g., PF-07304814 (Pfizer) or PF-07321332 (Pfizer), optionally co-administered with relatively low doses of ritonavir (ritonavir)), polymerase inhibitors, antiviral nucleosides (e.g., ritonavir, GS-441524, AT-527 (ATEA), N4-hydroxycytosine, mo Nuola vir (Molnupiravir) (N4-hydroxycytosine prodrug) and other compounds disclosed in U.S. Pat. No.9,809,616 and prodrugs thereof), 4' -fluorouridine and prodrugs thereof, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, and agents of different or unknown mechanisms.
Wu Mifei norvir (Umifenovir), also known as Arbidol, is a representative fusion inhibitor.
Representative entry inhibitors include Camostat (castostat), luteolin (luteolin), MDL28170, SSAA09E2, SSAA09E1 (acting as a cathepsin L inhibitor), SSAA09E3, and tetra-O-galloyl- β -D-glucose (TGG). The chemical formulas of certain compounds are provided below:
other entry inhibitors include the following:
redexivir, sofosbuvir (Sofosbuvir), ribavirin (ribavirin), IDX-184 and GS-441524 have the formula:
/>
/>
in addition, compounds that inhibit cytokine storms, anticoagulants and/or platelet aggregation inhibitors that address the clotting problem, compounds that sequester iron ions released from hemoglobin by viruses such as covd-19, cytochrome P-450 (CYP 450) inhibitors, and/or NOX inhibitors may be administered.
Representative NOX inhibitors are disclosed in PCT/US2018/067674 and include AEBSF; apocynin (apocynin); DPI; GK-136901; ML171; plumbagin (Plumbagin); s17834; VAS2870; VAS3947; GKT-831; GKT771; GTL003 or an amidothiadiazole derivative thereof as described in AU2015365465, EP20140198597 and WO 2015/59659; schisandrin B (Schisandrin B), as described in CN104147001 and CN 20131179455); biaromatic and tri-aromatic compounds described in U.S. publication nos. 2015045387, GB 20110016017, and WO 201200725; methoxy flavone derivatives described in JP 2015227329, JP 20140097875 and JP 20150093939; peptides, such as NOX2ds-tat and PR-39, as described in U.S. publication No.2015368301, TN 2015000295, U.S. publication No.201514689803, U.S. publication No.201462013916, PCT WO 201450063 and EP 20130150187; piperazine derivatives described in U.S. publication No.2014194422, U.S. patent No.9428478, U.S. publication No.201214123877, U.S. publication No.201161496161, and PCT WO2012 US 41988; pyrazole derivatives disclosed in KR101280198, KR20110025151 and KR 20090082518; pyrazoline dione derivatives disclosed in HK1171748, PCT WO201054329 and EP 20090171466; pyrazolopiperidine derivatives disclosed in KR20130010109, KR20130002317, EP20100153927, PCT WO201150667, EP20100153929 and PCT WO2011IB 50668; pyrazolopyridine derivatives described in KR20170026643, HK1158948, HK1141734, HK1159096, HK1159092, EP20080164857, PCT WO200954156, PCT WO200954150, EP20080164853, PCT WO200853390, U.S. publication No.20070896284, EP20070109555, PCT WO 200954148, EP20080164847, PCT WO200954155 and EP 20080164849; quinazoline and quinoline derivatives as disclosed in EP2886120, U.S. publication No.2014018384, U.S. publication No.20100407925, EP20110836947, GB20110004600 and PCT WO 201250586; tetrahydroindole derivatives disclosed in U.S. publication No.2010120749, U.S. patent No.8,288,432, U.S. publication No.20080532567, EP20070109561, U.S. publication No.20070908414, and PCT WO 200853704; tetrahydroisoquinoline derivatives disclosed in U.S. publication No.2016083351, U.S. publication No.201414888390, U.S. publication No.201361818726, and PCT WO 201436402; scopoletin (Scopoletin) described in TW201325588 and TW 20110147671; and 2, 5-disubstituted benzoxazole and benzothiazole derivatives disclosed in TW201713650 and PCT WO 201554662. Representative NOX inhibitors also include those disclosed in PCT WO 2011062864.
Exemplary Nox inhibitors also include 2-phenylbenzo [ d ] isothiazol-3 (2H) -one, 2- (4-methoxyphenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (benzo [ d ] [ l,3] dioxol-5-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (4-fluorophenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) -5-fluorobenzo [ d ] isothiazol-3 (2H) -one, 5-fluoro-2- (4-fluorophenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (2-chloro-6-methylphenyl) -5-fluorobenzo [ d ] isothiazol-3 (2H) -one, 5-fluoro-2-phenylbenzo [ d ] isothiazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) -5-fluorobenzoic acid [ d ] isothiazol-3 (2H) -one, 5-fluoro-2- (4-fluorophenyl) benzo [ d ] isothiazol-3 (2H) -one, 5-fluoro-2-fluorophenyl) -3 (2H) -one, 5-fluoro-benzo [ d ] isothiazol-3 (2H) -one, methyl 4- (5-fluoro-3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, ethyl 4- (3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, tert-butyl 4- (3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, methyl 2-methoxy-4- (3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, methyl 3-chloro-4- (3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, methyl 2- (4-acetylphenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (4-nitrobenzo [ d ] isothiazol-2 (3H) -yl) benzoate, methyl 4-chloro-4- (3-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, methyl 2- (4-oxybenzo [ d ] isothiazol-2 (3H) -yl) benzoate, 6- (3-oxo-benzo [ d ] isothiazol-2 (3H) -yl) nicotinonitrile, 2- (4- (hydroxymethyl) phenyl) benzo [ d ] isothiazol-3 (2H) -one, 2-benzyl benzo [ d ] isothiazol-3 (2H) -one, N-methyl-4- (3-oxo-benzo [ d ] isothiazol-2 (3H) -yl) benzamide, 2- (4-hydroxyphenyl) benzo [ d ] isothiazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) -l-methyl-lH-indazol-3 (2H) -one, 2- (4-fluorophenyl) -1-methyl-1H-indazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) -lH-indazol-3 (2H) -one, 1-methyl-2-phenyl-1H-indazol-3 (2H) -one, 2- (l, 3-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (2, 4-dimethylphenyl) -l-2- (2, 4-phenyl) -3 (2H) -one, 5-thioxo-3 (2H) -one, 3, 4-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (5- (methylthio) -l,3, 4-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 5-fluoro-2- (l, 3, 4-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (5- (tert-butyl) -l,3, 4-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (5- (4-bromophenyl) -l,3, 4-thiadiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one 2- (4-methylthiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (4, 5-dimethylthiazol-2-yl) benzo [ d ] isothiazol-3 (2H) -one, 2- (benzo [ d ] [ l,3] dioxol-5-yl) -4, 5-difluorobenzo [ d ] [ l,2] selenazol-3 (2H) -one, 2- (benzo [ d ] [ l,3] dioxol-5-yl) -5-fluorobenzo [ d ] [ l,2] selenazol-3 (2H) -one, 2- (2, 3-dihydrobenzo [ b ] [ l,4] dioxin-6-yl) -5-fluorobenzo [ d ] [ l,2] selenazol-3 (2H) -2- (4- (l, 3-dioxol-2-yl) phenyl) benzo [ d ] [ l,2] selenazol-3 (2H) -one, 2- (benzo [ d ] [ l,3] dioxol-5-yl) -6, 7-dimethoxybenzo [ d ] [ l,2] selenazol-3 (2H) -one, 4- (3-oxobenzo [ d ] [ l,2] selenazol-2 (3H) -yl) benzoic acid methyl ester, 4- (3-oxoisothiazolo [5,4-b ] pyridin-2 (3H) -yl) benzoic acid methyl ester and 4- (3-oxoisothiazol-2 (3H) -yl) benzoic acid ethyl ester, and pharmaceutically acceptable salts and prodrugs thereof.
Additional representative NOX inhibitors include:
specific examples of these compounds include
Their deuterated analogs or their pharmaceutically acceptable salts or prodrugs.
In one embodiment, the NOX inhibitor is Ebselen (Ebselen), neopterin (Neopterin), APBA, biarubicin (diapocin) or deuterated analogs thereof or a pharmaceutically acceptable salt or prodrug thereof.
In another embodiment, are those NOX compounds disclosed in PCT WO 2010/035221.
In another embodiment, the compound is a NOX inhibitor disclosed in PCT WO 2013/068972 selected from the group consisting of:
4- (2-fluoro-4-methoxyphenyl) -2- (2-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (4-methoxyphenyl) -5- (pyrazin-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (4-chlorophenyl) -2- (2-methoxyphenyl) -5- (pyrazin-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2-fluoro-4-methoxyphenyl) -5- [ (l-methyl-lH-pyrazol-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (2-fluoro-5-methoxyphenyl) -2- (2-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -5- [ (2-methoxypyridin-4-yl) methyl ] -4-methyl-1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-methoxyphenyl) -4-methyl-5- (pyridin-3-ylmethyl) -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
4- (4-chloro-2-fluorophenyl) -2- (2-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (5-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-3-ylmethyl) -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-chlorophenyl) -5- [ (6-methoxypyridin-3-yl) methyl ] -4-methyl-1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
4- (4-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-3-ylmethyl) -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
4- (5-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (2-fluoro-5-methoxyphenyl) -2- (2-methoxyphenyl) -5- [ (1-methyl-1H-pyrazol-1-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
4- (5-chloro-2-fluorophenyl) -2- (2-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4-methyl-5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (4-chlorophenyl) -5- (pyrazin-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2-fluorophenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (4-chlorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (4-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-methoxyphenyl) -4- (3-methoxyphenyl) -5- [ (1-methyl-1H-pyrazol-1-3-yl) methyl ] -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-chlorophenyl) -4- (2-fluoro-4-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (2-fluoro-4-methoxyphenyl) -2- (2-methoxyphenyl) -5- [ (1-methyl-1H-pyrazol-1-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-methoxyphenyl) -4- (4-methoxyphenyl) -5- [ (1-methyl-1H-pyrazol-1-3-yl) methyl ] -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-methoxyphenyl) -4- (3-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (4-chlorophenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (4-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- [ (2-methoxypyridin-4-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2-fluoro-4-methoxyphenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2, 6-difluorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2-fluorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4-methyl-5- [ (l-methyl-lH-pyrazol-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (3-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -5-methyl-4- [3- (methylamino) phenyl ] -1H-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-methoxyphenyl) -4- (4-methoxyphenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2-fluorophenyl) -5- (pyridin-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2, 5-difluorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (4-chlorophenyl) -5- (l, 3-thiazol-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- [3- (dimethylamino) phenyl ] -5- [ (1-methyl-1H-pyrazol-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione;
2- (2-chlorophenyl) -4- (3, 5-dichlorophenyl) -5- (pyridin-4-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (3-chloro-2-fluorophenyl) -2- (2-chlorophenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- [3- (dimethylamino) phenyl ] -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2, 6-difluorophenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
4- (2-fluoro-5-methoxyphenyl) -2- (2-methoxyphenyl) -5- (pyrazin-2-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione;
2- (2-chlorophenyl) -4- (2, 5-difluorophenyl) -5- (pyridin-3-ylmethyl) -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2 h,5 h) -dione; and
2- (2-chlorophenyl) -4- [3- (dimethylamino) phenyl ] -5- [ (1-methyl-1H-pyrazol-3-yl) methyl ] -lH-pyrazolo [4,3-c ] pyridine-3, 6 (2H, 5H) -dione.
Representative CYP450 inhibitors include, but are not limited to, amiodarone (amiodarone), amlodipine (amiodipine), apigenin (apigenin), aprepitant (aprepatan), bergamot (grapefruit), buprenorphine (buprofezin), caffeine, cafestol (cafestol), cannabidiol (cannabiol), celecoxib (celecoxib), chloramphenicol (chloramphenicol), chlorpheniramine (chlorpheniramine), chlorpromazine (chlorpromazine), ciprofloxacin (ciprofloxacin), citalopram (citalopram), citalopram (clarituxin), clenbuterol (chlorpyrifosamite), chlorpyrifosamite (codetin), ketotifen (ketotifen), ketotifen (thiofidaline), xanthone (picloramine), xanthophylline (picloram), and other drugs (picloram), and combinations thereof Fluoroquinolones (fluoroquinolones) (e.g., ciprofloxacin), fluoxetine (fluoxoline), fluvoxamine (fluvoxamine), fluconazole (fluconazole), fluvastatin (fluvastatin), gabapentin (gabapentin), gemfibrozil, gestodene, halofantrine (halofantrine), haloperidol (haloperidol), oxazine, imatinib (imatinib), indomethacin (indomethacin), indinavir (indinavir), interferon, isoniazid (isoniazid), itraconazole (itraconazole), JWH-018, ketoconazole (ketconazole), letrozole (letrozole), lovastatin (lovastatin), levomethapromazine (levomethaprozine), memantine (mevalinate), methide (methymate), methylpiperazine (methylpiperazine). Metoclopramide, methadone, methimazole, methoxsalen, metrafenone, milbezadil, mibefradil, miconazole, midodrine, mifepristone, milk thistle, mollobemide, modafinil, montelukast, mollobemide, naringenin, nefazodone, nelfinavir, nicotinic acid, nicotinamide, nilutamide, norfloxacin, oxyphenamine, paroxetine, and pharmaceutical compositions containing the same, perphenazine, pilocarpine, piperine, phenylbutazone, probenecid, promethazine, proton pump inhibitors such as lansoprazole, omeprazole, pantoprazole, rabeprazole, quercetin, quinidine, ranitidine, risperidone, ritonavir, saquinavir, selegiline, sertraline, caraway, holjohn's word, sulconazole, fluquindine, and the like Sulfamethoxazole (Sulfamethoxazole), sulfabenzzole (Sulfaphenazole), telithromycin (telithromycin), teniposide (teniposide), terbinafine (terbinafine), thiazolidinediones, thioridazine (thioridazine), ticlopidine (ticlopidine), tioconazole (tioconazole), thiotepa (thiotepa), trimethoprim (trimethoprim), topiramate (topiramate), tranylcypromine (tranylcypromine), qu Bi (tripelennamine), valerian, valproic acid, verapamil, voriconazole (voriconazole), zafirlukast (zafirlukast) and zuclothianol (zuclomiphil).
Representative ACE-2 inhibitors include sulfhydryl-containing agents such as alapril (alaepril), captopril (captopril) and zefmopril (zefmopril); dicarboxylic acid ester-containing agents such as enalapril (vasotec), ramipril (ramipri) (altace), quinapril (quinapril) (accupril), perindopril (perindopril) (coversyl), lisinopril (lisril), benazepril (lotensin), imidaclopril (tanatril), trandolapril (mavik) and cilazapril (inhibace), and phosphonate-containing agents such as fosinopril (fosinopril) (fosinopril/monopril).
For example, when used to treat or prevent an infection, the active compound or a prodrug or pharmaceutically acceptable salt thereof may be administered in combination or alternation with another antiviral agent, including, but not limited to, those of the above formula. Generally, in combination therapy, an effective dose of two or more agents are administered together, while during alternation therapy, an effective dose of each agent is administered serially. The dosage will depend on the absorption, inactivation, and excretion rates of the drug, as well as other factors known to those of skill in the art. Notably, the dosage value will also vary with the severity of the condition to be alleviated. It will also be appreciated that for any particular subject, the particular dosage regimen and schedule should be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions.
Ghosh et al, "Drug Development and Medicinal Chemistry Efforts Toward SARS-Coronavirus and Covid-19 Therapeutics," chemMed chem 10.1002/cmdc.202000223 discloses a number of agents in combination with the compounds described herein.
Non-limiting examples of antiviral agents that can be used in combination with the compounds disclosed herein include those listed below.
Compounds that inhibit cytokine storm
During activation, the inflammatory response must be regulated to prevent destructive systemic inflammation, also known as "cytokine storms". This is the case for many cytokines with anti-inflammatory properties, such as IL-10 and transforming growth factor beta (TGF-beta). Each cytokine acts on a different part of the inflammatory response. For example, the products of a Th2 immune response may suppress a Th1 immune response and vice versa.
By addressing inflammation, collateral damage to surrounding cells can be minimized with little or no long-term damage to the patient. Thus, in addition to using the compounds described herein to inhibit viral infection, one or more compounds that inhibit cytokine storms may be co-administered.
Compounds that inhibit cytokine storms include compounds directed against primary immune pathways, such as the chemokine network and cholinergic anti-inflammatory pathways.
JAK inhibitors, such as JAK1 and JAK2 inhibitors, can inhibit cytokine storms and in some cases also have antiviral effects. Representative JAK inhibitors include those disclosed in U.S. patent No.10,022,378, such as, for example, strapdown scrupulously and respectfully, tofacitinib and baritinib, and LY3009104/INCB28050, pactinib/SB 1518, VX-509, GLPG0634, INC424, R-348, CYT387, TG 10138, AEG 3482, and pharmaceutically acceptable salts and prodrugs thereof.
Other examples include CEP-701 (Lestaurtinib), AZD1480, INC424, R-348, CYT387, TG 10138, AEG 3482, 7-iodo-N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (4-aminophenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acrylamide, 7- (3-aminophenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acrylamide, N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-3-yl) amine, 2-morpholino [3,2-d ] thieno-7-yl) thieno [3,2-d ] pyrimidin-7-yl) phenyl ] amide, 5-pyrrolo [ 2-d ] pyrimidin-7-yl ] pyrimidine-amine, 5-yl ] pyrrolo [3,2-d ] amino ] pyrimidine-2-yl ] amide 7- (4-amino-3-methoxyphenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzene-sulfonamide, N-dimethyl-3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, 1-ethyl-3- (2-methoxy-4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) urea, N- (4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, 2-methoxy-4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) pheno-l, 2-cyano-N- (3- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl ] acetamide, N- (cyanomethyl) -2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidine-7-carboxamide, N- (3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, 1-ethyl-3- (4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) -2- (trifluoromethoxy) phenyl) urea, N- (3-nitrophenyl) -7-phenylthieno [3,2-d ] pyrimidin-2-amine, 7-iodo-N- (3-nitrophenyl) thieno [3,2-d ] pyrimidin-2-amine, N1- (7- (2-ethylphenyl) thieno [3,2-d ] pyrimidin-2-yl) benzene-1, 3-diamine, N-tert-butyl-3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, N1- (7-iodo-3, 2-d ] pyrimidin-2-yl) benzo [3,2-d ] pyrimidin-2-yl ] thiophen-amine, 7- (4-amino-3- (trifluoromethoxy) phenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (2-ethylphenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, N- (cyanomethyl) -N- (3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, N- (cyanomethyl) -N- (4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, N- (3- (5-methyl-2- (4-morpholinophenylamino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, 4- (5-methyl-2- (4-morpholinophenylamino) -5-pyrrolo [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, N- (4- (5-methyl-2- (4-morpholinophenylamino) -5H-pyrrolo [3,2-d ] pyrimidin-7-yl) phenyl) methanesulfonamide, 7-iodo-N- (4-morpholinophenyl) -5H-pyrrolo [3,2-d ] pyrimidin-2-amine, 7- (2-isopropylphenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 7-bromo-N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, N7- (2-isopropylphenyl) -N2- (4-morpholinophenyl) thieno [3,2-d ] pyrimidine-2, 7-diamine, N7- (4-isopropylphenyl) -N2- (4-morpholinophenyl) thieno [3,2-d ] pyrimidine-2, 7-diamine, 7- (5-amino-2-methylphenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (2-d) thieno [3,2-d ] pyrimidine-amine, N- (2-isopropylphenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidine-amine, N- (4-morpholino) amino-2-4-morpholino-yl) thieno [3,2-d ] thieno [ 2-methyl-2-amino ] amino-2-carbonyl) -N-amino-7-amino-thieno [ 4-2-yl-carbonyl ] amino-2-carbonyl-amino-thieno-2-amine 7-iodo-N- (3-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (4-amino-3-nitrophenyl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (2-methoxypyridin-3-yl) -N- (4-morpholinophenyl) thieno [3,2-d ] pyrimidin-2-amine, (3- (7-iodothieno [3,2-d ] pyrimidin-2-ylamino) phenyl) methanol, N-tert-butyl-3- (2- (3-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, N-tert-butyl-3- (2- (3- (hydroxymethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, N- (4-morpholinophenyl) -7- (4-nitrobenzylthio) -5H-pyrrolo [3,2-d ] pyrimidin-2-amine, N-tert-butyl-3- (2- (3-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, 7- (4-amino-3-nitrophenyl) -N- (3, 4-dimethoxyphenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3, 4-dimethoxyphenyl) -7- (2-methoxypyridin-3-yl) thieno [3,2-d ] pyrimidin-2-amine, N-tert-butyl-3- (2- (3, 4-dimethoxyphenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, 7- (2-aminopyrimidin-5-yl) -N- (3, 4-dimethoxyphenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3, 4-dimethoxyphenyl) -7- (2, 6-dimethoxypyridin-3-yl) thieno [3,2-d ] -pyrimidin-2-amine, N- (3, 4-dimethoxyphenyl) -7- (2, 4-dimethoxypyrimidin-5-yl) thieno [3,2-d ] pyrimidin-2-amine, 7-iodo-N- (4-methylphenyl) thieno [3,2-d ] morpholino ] amine, N-tert-butyl-3- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, 2-cyano-N- (4-methyl-3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, 3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzoic acid ethyl ester, 7-bromo-N- (4- (2- (pyrrolidin-1-yl) ethoxy) phenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3- (2- (4- (2- (pyrrolidin-1-yl) ethoxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, N- (cyanomethyl) -3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzamide, N-tert-butyl-3- (2- (4-morpholinophenyl) pyrimidin-7-yl) phenyl) thieno [3,2-d ] pyrimidin-7-yl ] benzamide, N-tert-butyl-3- (2- (4- (1-ethylpiperidin-4-yloxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, tert-butyl-4- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) -1H-pyrazole-1-carboxylate, 7-bromo-N- (4- ((4-ethylpiperazin-1-yl) methyl) phenyl) thieno [3,2-d ] pyrimidine-2-amine, N-tert-butyl-3- (2- (4- ((4-ethylpiperazin-1-yl) methyl) phenylamino) -thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, N- (4- ((4-ethylpiperazin-1-yl) methyl) phenyl) -7- (1H-pyrazol-4-yl) thieno [3,2-d ] pyrimidin-2-amine, N- (cyanomethyl) -3- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzamide, N-tert-butyl-3- (2- (4- (2- (pyrrolidin-1-yl) ethoxy) phenylamino) thieno [3,2-d ] -pyrimidin-7-yl) benzenesulfonamide, pyrrolidin-1-yl) ethoxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzyl carbamic acid tert-butyl ester, 3- (2- (4- (2- (pyrrolidin-1-yl) ethoxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzenesulfonamide, 7- (3-chloro-4-fluorophenyl) -N- (4- (2- (pyrrolidin-1-yl) ethoxy) phenyl) thieno- [3,2-d ] pyrimidin-2-amine, 4- (2- (4- (1-ethylpiperidin-4-yloxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl-) -1H-pyrazole-1-carboxylic acid tert-butyl ester, 7 (benzo [ d ] [1,3] dioxol-5-yl) -N- (4-methyl) thieno [3,2-d ] morpholino ] amine, tert-butyl 5- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) -1H-indole-1-carboxylate, tert-butyl 7- (2-aminopyrimidin-5-yl) -N- (4- (morpholinomethyl) phenyl) thieno [3,2-d ] pyrimidin-2-amine, 4- (2- (-4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) -5, 6-dihydropyridin-1 (2H) -carboxylate, tert-butyl morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzyl carbamate, tert-butyl N- (3- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, N- (4- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, N- (3- (4- (morpholinomethyl) phenyl) pyrimidin-7-yl) phenyl) thieno [3, 3-d ] thio ] methyl ] thioamide, 7- (4- (4-methylpiperazin-1-yl) phenyl) -N- (4- (morpholinomethyl) phenyl) thieno- [3,2-d ] pyrimidin-2-amine, N- (2-methoxy-4- (2- (4- (morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) acetamide, 7-bromo-N- (3, 4, 5-trimethoxyphenyl) thieno [3,2-d ] pyrimidin-2-amine, (3- (2- (3, 4, 5-trimethoxyphenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanol (4- (2- (3, 4, 5-trimethoxyphenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanol, (3- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanol, (4- (2- (4-morpholinophenylamino) thieno [3,2-d ] pyrimidin-7-yl) phenyl) methanol, N- (pyrrolidin-1-yl) ethoxy) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzyl) methanesulfonamide, morpholinomethyl) phenylamino) thieno [3,2-d ] pyrimidin-7-yl) benzyl carbamic acid tert-butyl ester, N- (4- (morpholinomethyl) phenyl) -7- (3- (piperazin-1-yl) phenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (6- (2-morpholinoethyl-amino) pyridin-3-yl) -N- (3, 4, 5-trimethoxyphenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (2-ethylphenyl) -N- (4- (2- (pyrrolidin-1-yl) ethoxy) phenyl) thieno [3,2-d ] pyrimidin-2-amine, 7- (4- (aminomethyl) phenyl) -N- (4- (morpholinomethyl) phenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (4- (1-ethylpiperidin-4-yloxy) phenyl) -7- (1H-pyrazol-4-yl) thieno [3,2-d ] pyrimidin-2-amine, N- (2, 4-diphenyl) -7-thieno [3,2-d ] pyrimidin-2-amine, 7-bromo-N- (3, 4-dimethoxyphenyl) thieno [3,2-d ] pyrimidin-2-amine, N- (3, 4-dimethoxyphenyl) -7-phenylthieno [3,2-d ] pyrimidin-2-amine, and pharmaceutically acceptable salts and prodrugs thereof.
An HMGB1 antibody and a COX-2 inhibitor may be used, which down-regulates the cytokine storm. Examples of such compounds include Actemra (Roche). Celebrex (celecoxib), a COX-2 inhibitor, may be used. Inhibitors of IL-8 (CXCL 8) may also be used.
Chemokine receptor CCR2 antagonists such as PF-04178903 can reduce pulmonary immune lesions.
Selective alphase:Sub>A 7Ach receptor agonists, such as GTS-21 (DMXB-A) and CNI-1495, may be used. These compounds reduce TNF-alpha. Late mediator of sepsis, HMGB1, down regulates the IFN- γ pathway and prevents LPS-induced inhibition of IL-10 and STAT3 mechanisms.
Compounds for treating or preventing blood clots
Viruses that cause respiratory tract infections, including coronaviruses such as Covid-19, may be associated with pulmonary blood clots, which may also damage the heart.
The compounds described herein may be co-administered with a compound that inhibits clot formation (e.g., a blood diluent) or a compound that breaks down an existing clot (e.g., tissue Plasminogen Activator (TPA), integrilin (eptifibatide), abciximab (ReoPro), or tirofiban (Aggrastat)).
The blood thinner prevents the formation of blood clots and prevents the enlargement of existing blood clots. Blood diluents are mainly of two types. Anticoagulants, such as heparin or warfarin (also known as Coumadin), slow down the biological processes that produce the clot, while anti-platelet aggregation drugs, such as plamix, aspirin (aspirin), prevent blood cells, known as platelets, from aggregating together to form the clot.
For example, the processing steps may be performed,typically at a dose of 180mcg/kg, administered as an intravenous bolus as soon as possible after diagnosis, with continuous infusion of 2mcg/kg/min (after initial bolus) for up to 96 hours of treatment.
Representative platelet aggregation inhibitors include glycoprotein IIB/IIIA inhibitors, phosphodiesterase inhibitors, adenosine reuptake inhibitors, and Adenosine Diphosphate (ADP) receptor inhibitors. These may optionally be administered in combination with an anticoagulant.
Representative anticoagulants include coumarin (vitamin K antagonists); heparin and its derivatives, including plain heparin (unfractionated heparin, UFH), low Molecular Weight Heparin (LMWH), and Ultra Low Molecular Weight Heparin (ULMWH); factor Xa synthetic pentasaccharide inhibitors including Fang Dapa forest (Fondaparinux), ectropin (Idraparinux) and ectropin (idrabiotap arinux); direct acting oral anticoagulants (DAOC), such as dabigatran (dabigatran), rivaroxaban (rivaroxaban), apixaban (apixaban), edoxaban (edoxaban) and betrixaban (betrixaban), and antithrombin protein therapeutics/thrombin inhibitors, such as the bivalent drugs hirudin, lepirudin (lepirudin) and bivalirudin (bivalirudin) and monovalent argatroban (argatroban).
Representative platelet aggregation inhibitors include pravastatin (pravastatin), plavix (clopidogrel bisulfate (clopidogrel bisulfate)), platal (cilostazol), effect (prasugrel), aggrenox (aspirin and dipyridamole), briinta (ticagrelor), carboxilizumab, kentreal (cangrel), persantine (dipyridamole), ticlopidine (ticlopidine), yospralla (aspirin and omeprazole).
Small molecule covalent CoV 3CLpro inhibitors
Representative small molecule covalent CoV 3CLpro inhibitors include the following compounds:
non-covalent CoV 3CLpro inhibitors
Representative non-covalent CoV 3CLpro inhibitors include the following:
/>
SARS-CoV PLpro inhibitors
Representative SARS-Cov PLpro inhibitors include the following:
/>
additional compounds include the following:
additional compounds that may be used
Additional compounds and classes of compounds useful in combination therapies include: antibodies, including monoclonal antibodies (mAbs), abidol (Wu Mifei nomovir), actemra (tolizumab)), APN01 (Aperion Biologics), ARMS-1 (including cetylpyridinium chloride (CPC)), ASC09 (Ascletis Pharmaceutical), AT-001 (Applied Therapeutics company) and other Aldose Reductase Inhibitors (ARI), ATYR1923 (aTyr Pharmacopeia), aviptadil (Relief Therapeutics), azvudine (Azvudine), bei Sheng tinib (Bemcinninib), BLD-2660 (Blade Therapeutics), bevacizumab), britissimab (Bruncatib), calquezene (acalyptib), carboline mesylate (TMPRSS 2 inhibitor), caririlizumab (Camrelizumab), CAP-1002 (Capricor Therapeutics), CD24Fcm, clevidine (Coueing), 4620-C, devaldecoco (CRA), devaldecoco-52, devaldecoco (DSV), devaldecoco-52, devaldecoco (Devaldecoco) and UK (Devaldecovals) (52), devaldecovaldecoco (Devaldecoco) 5) and UK (Devals) (Devaldecovals) (52) are included Fadracilib (CYC 065) and Sinumber Li Xili (selicillib) (roscovitine) (cyclin dependent kinase (CDK) inhibitors), farxika (dapagliflozin), farvipravir (Favlavir)/Faviravir/T-705/Avigan, ganovo (danopropanvir), gilenya (fingolimod) 1-phosphate sphingosine receptor modulators, boschirumab (Gimsilomab), IFX-1, ilanabinin (canakineumbab), intravenous immunoglobulins, ivermectin (Ivermexicam) (input protein alpha/beta inhibitors), katra/Aluvia (LAAvirin)/Lithovir), ganovo (Danopropanavir) 5, for example, tasugrel (Danopropanvir), levalproin (Levalprokine) and Levalproin (Levalproin) receptor modulators, levalprokine (Levalproimide (Levalproir), levalprokine (Levalprokine) and Levalprozid (Levalsartan) 2 (Levalsartan) and Levalproin (Levalsartan) receptor modulators, levalprojen (Levalsartan) and Levalsartan (Levalsartan) receptor antagonists (Levalsartan) of Levalproir) (Levalproinflammatory receptor inhibitors, levalsartan (Levalsartan) and Levalsartan (Levalsartan) Levalat (Levalsartan) Levamide (Levamide) 6) Metablok (LSALT peptide, a DPEP1 inhibitor), methylprednisolone and other corticosteroids, MN-166 (ibudilast), macrophage Migration Inhibitory Factor (MIF) inhibitor), MRx-4DP0004 (Bifidobacterium breve strain, 4D Pharma), nafamostat (a serine protease inhibitor), neuraminidase inhibitors (e.g. Tamiflu (oseltamivir)), nitazoxanide (nucleocapsid (N) protein inhibitor), nivolumab (Nivolumab), OT-101 (Mateon), novaferon (artificial interferon), opaonib (yeliva) (sphingokinase 2 inhibitor), octreotide Li Shan anti (Otilimab), PD-1 blocking antibodies, polyethylene glycol interferons (e.g. polyethylene glycol interferon), petidine (famotidine (monostatin), piotalin (P) receptor (P) and Puvidone receptor (P.sub.3) agonist (P.sub.2), toll-like receptor (TLR) binding agent), rebif (interferon beta-1 a), RHB-107 (Wu Mosi he (upamostat)) (serine protease inhibitor, redHill Biopharma limited), plug Li Nisuo (Selinexor) (nuclear export Selective Inhibitor (SINE)), SNG001 (synairin, inhaled interferon beta-1 a), solitaide (Solnatide), stem cells (including mesenchymal stem cells, multiStem (Athersys) and PLX (Pluristem Therapeutics)), sylvant (siltuximab), thymosin, TJM2 (TJ 003234), tradipitant (neurokinin-1 receptor antagonist), truvada (emtricitabine) and tenofovir), ultopiris (elkulizumab-cwvz), vazegepant (CGRP receptor antagonist or blocker) and Xofluza (balo Sha Weima bosch (baloxavir marboxil)).
Antiviral agent for readjustment of use
Many agents, including agents active against other viruses, have been evaluated against Covid-19 and found to be active. Any of these compounds may be combined with the compounds described herein. Representative compounds include lopinavir, ritonavir, niclosamide, promazine, PNU, UC2, cinnarin (cinanserin) (SQ 10,643), carboimidazole (calmidozolium) (C3930), tannic acid, 3-isotheaflavin-3-gallate, theaflavin-3, 3' -digallic acid, glycyrrhizin, S-nitroso-N-acetylpenicillamine, nelfinavir, niclosamide, chloroquine, hydroxychloroquine, 5-benzyloxymine (5-benzooxygramine), ribavirin, interferons (e.g., interferon (IFN) - α, IFN- β and pegylated forms thereof), and combinations of these compounds with ribavirin, chlorpromazine hydrochloride, trifluopromazine hydrochloride, gemcitabine (gemcitabine), imatinib mesylate (imatinib mesylate), dasatinib (dasatinib), and imatinib (imatinib).
VIII pharmaceutical composition
Hosts, including but not limited to humans infected with coronaviridae viruses or other viruses described herein, may be treated by administering to a patient an effective amount of an active compound or a pharmaceutically acceptable prodrug or salt thereof in the presence of a pharmaceutically acceptable carrier or diluent. The active substance may be administered by any suitable route, for example orally, parenterally, intravenously, intradermally, subcutaneously or topically, in liquid or solid form.
The preferred dosage of the compound will range between about 0.01 and about 10mg/kg of the recipient's body weight per day, more typically between about 0.1 and 5mg/kg, and preferably between about 0.5 and about 2mg/kg, until the patient is rehabilitated. In some cases, the compounds may be administered in doses up to 10 μm, which may be considered a relatively high dose if administered over a prolonged period of time, but which is acceptable when administered during one or more viral infections described herein, typically on the order of days to weeks.
The effective dosage range of the pharmaceutically acceptable salts and prodrugs can be calculated based on the weight of the parent compound to be delivered. If the salt or prodrug itself exhibits activity, the weight of the salt or prodrug may be used as described above or the effective dose may be estimated by other means known to those skilled in the art.
The compounds are suitably administered in any suitable dosage form including, but not limited to, dosage forms containing from 7 to 600mg, preferably from 70 to 600mg, of active ingredient per unit dosage form. Oral doses of 5-400mg are generally convenient.
The concentration of the active compound in the pharmaceutical composition will depend on the absorption, inactivation, and excretion rates of the drug, as well as other factors known to those of skill in the art. Notably, the dosage value will also vary with the severity of the condition to be alleviated. It should also be understood that the specific dosage regimen for any particular subject should be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentrations set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions. The active ingredient may be administered at one time or may be divided into a plurality of smaller doses for administration at different time intervals.
The preferred mode of administration of the active compounds is oral. Oral compositions typically include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For oral administration of the therapeutic agent, the active compound may be mixed with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binders and/or adjuvant materials may be included as part of the composition.
Tablets, pills, capsules, troches and the like may contain any of the following ingredients or compounds of similar nature: a binder, such as microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example starch or lactose; disintegrants, for example alginic acid, primogel or corn starch; lubricants, such as magnesium stearate or Sterotes; glidants, such as colloidal silicon dioxide; sweeteners, such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil. In addition, the unit dosage form may contain various other materials that alter the physical form of the dosage unit, such as coatings of sugar, shellac, or other enteric agents.
The compounds may be administered as a component of elixirs, suspensions, syrups, wafers, chewing gums and the like. In addition to the active compounds, syrups may contain sucrose as a sweetener, as well as certain preservatives, dyes, colorants and flavors.
The compound or pharmaceutically acceptable prodrug or salt thereof may also be admixed with other active substances which do not impair the desired effect, or with substances which supplement the desired effect (e.g. antibiotics, antifungals, anti-inflammatory agents or other antiviral compounds). Solutions or suspensions for parenteral, intradermal, subcutaneous, or topical administration may include the following components: sterile diluents, such as water for injection, saline solutions, fixed oils, polyethylene glycols, glycerol, propylene glycol or other synthetic solvents; antimicrobial agents, such as benzyl alcohol or methylparaben; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediamine tetraacetic acid; buffers such as acetate, citrate or phosphate; and agents for modulating tonicity, such as sodium chloride or dextrose. Parenteral formulations may be packaged in ampules, disposable syringes or multiple dose vials made of glass or plastic.
If administered intravenously, the preferred carrier is physiological saline or Phosphate Buffered Saline (PBS).
Transdermal preparation
In some embodiments, the composition is in the form of a transdermal formulation, for example, a formulation for use in an FDA approved agonist Luo Tiji (rotigitine) transdermal formulation (Neupro patch). Another suitable formulation is the formulation described in U.S. publication No.20080050424, entitled "Transdermal Therapeutic System for Treating Parkinsonism". The formulation includes a silicone or acrylate based binder and may include an additive having increased solubility for the active agent in an amount effective to increase the dissolution capacity of the matrix for the active agent.
The transdermal formulation may be a single-phase matrix comprising a backing layer, a self-adhesive matrix containing the active substance and a protective film that is removed prior to use. More complex embodiments contain a multi-layer matrix that may also contain a non-adhesive layer and a control film. If a polyacrylate adhesive is used, it can be crosslinked with multivalent metal ions such as zinc, calcium, aluminum or titanium ions (e.g., aluminum acetylacetonate and titanium acetylacetonate).
When silicone adhesives are used, they are typically polydimethylsiloxanes. However, in principle other organic residues may be present, for example ethyl or phenyl instead of methyl. Because the active compound is an amine, it is desirable to use an amine-resistant binder. A representative amine-resistant adhesive is described, for example, in EP 0 180 377.
Representative acrylate-based polymeric binders include acrylic acid, acrylamide, hexyl acrylate, 2-ethylhexyl acrylate, hydroxyethyl acrylate, octyl acrylate, butyl acrylate, methyl acrylate, glycidyl acrylate, methacrylic acid, methacrylamide, hexyl methacrylate, 2-ethylhexyl methacrylate, octyl methacrylate, methyl methacrylate, glycidyl methacrylate, vinyl acetate, vinyl pyrrolidone, and combinations thereof.
The adhesive must have a suitable dissolution capacity for the active substance and the active substance is preferably able to migrate within the matrix and pass through the contact surface to the skin. Transdermal formulations with suitable transdermal transport of the active substance can be readily formulated by the person skilled in the art.
Certain pharmaceutically acceptable salts are often more preferred for transdermal formulations because they can assist the passage of the active agent across the stratum corneum barrier. Examples include fatty acid salts such as stearates and oleates. Oleate and stearate are relatively lipophilic and may even act as penetration enhancers for the skin.
Penetration enhancers may also be used. Representative penetration enhancers include fatty alcohols, fatty acids, fatty acid esters, fatty acid amides, glycerol or fatty acid esters thereof, N-methylpyrrolidone, terpenes such as limonene, alpha-pinene, alpha-terpineol, carvone, carveol, limonene oxide, pinene oxide, and 1, 8-eucalyptol.
The patch may be generally prepared by dissolving or suspending the active agent in ethanol or another suitable organic solvent, followed by the addition of the binder solution with stirring. Additional auxiliary substances may be added to the binder solution, the active substance solution or the active substance-containing binder solution. The solution may then be applied to a suitable sheet, the solvent removed, the backing layer laminated to the matrix layer, and the patch punched out of the overall laminate.
Nanoparticle compositions
The compounds described herein may also be administered in the form of nanoparticle compositions. In one embodiment, a controlled release nanoparticle formulation comprises a nanoparticle active agent to be administered and a rate controlling polymer that extends the release of the active agent after administration. In this embodiment, the composition may release the active agent for a period of about 2 to about 24 hours or up to 30 days or more after administration. Representative controlled release formulations comprising active agents in nanoparticle form are described, for example, in U.S. patent No.8,293,277.
Nanoparticle compositions can comprise particles of an active agent described herein, the surface of which is adsorbed or associated with a non-crosslinked surface stabilizer.
The average particle size of the nanoparticles is typically less than about 800nm, more typically less than about 600nm, more typically less than about 400nm, less than about 300nm, less than about 250nm, less than about 100nm, or less than about 50nm. In one aspect of this embodiment, at least 50% of the active agent particles have an average particle size of less than about 800nm, 600nm, 400nm, 300nm, 250nm, 100nm, or 50nm, respectively, as measured by light scattering techniques.
A variety of surface stabilizers are commonly used with nanoparticle compositions to prevent agglomeration or aggregation of the particles. Representative surface stabilizers are selected from the group consisting of: gelatin, lecithin, dextran, gum arabic, cholesterol, tragacanth, stearic acid, benzalkonium chloride (bezalkonium chloride), calcium stearate, glyceryl monostearate, cetostearyl alcohol, polysilritol emulsifying wax, sorbitan esters, polyoxyethylene alkyl ethers, polyoxyethylene castor oil derivatives, polyoxyethylene sorbitan fatty acid esters, polyethylene glycol, polyoxyethylene stearates, colloidal silica, phosphate salts, sodium lauryl sulfate, calcium carboxymethyl cellulose, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose phthalate, amorphous cellulose, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol, polyvinylpyrrolidone, tyloxapol (tyloxapol) poloxamers (poloxamers), poloxamers 908, sodium dialkyl sulfosuccinates, sodium dodecyl sulfate, alkylaryl polyether sulfonates, mixtures of sucrose stearate and sucrose distearate, p-isononyl phenoxy poly (glycidol), SA9OHCO, decanoyl-N-methyl glucamide, N-decyl-D-glucopyranoside, N-decyl-D-maltopyranoside, N-dodecyl-D-glucopyranoside, N-dodecyl-D-maltoside, heptanoyl-N-methyl glucamide, N-heptyl-D-glucopyranoside, N-heptyl-D-thioglucoside, N-hexyl-D-glucopyranoside, nonanoyl-N-methyl glucamide, N-nonyl-D-glucopyranoside, octanoyl-N-methyl glucamide, N-octyl-D-glucopyranoside and octyl-D-thiopyranoside. Lysozyme can also be used as a surface stabilizer for nanoparticle compositions. Certain nanoparticles, such as poly (lactic-co-glycolic acid) (PLGA) nanoparticles, are known to target the liver when administered Intravenously (IV) or Subcutaneously (SQ).
Representative rate controlling polymers from which nanoparticles may be formulated include chitosan, polyethylene oxide (PEO), polyvinyl acetate phthalate, gum arabic, agar, guar gum, cereal gum, dextran, casein, gelatin, pectin, carrageenan, wax, shellac, hydrogenated vegetable oil, polyvinylpyrrolidone, hydroxypropyl cellulose (HPC), hydroxyethyl cellulose (HEC), hydroxypropyl methylcellulose (HPMC), sodium carboxymethyl cellulose (CMC), poly (ethylene oxide), alkyl cellulose, ethyl cellulose, methyl cellulose, carboxymethyl cellulose, hydrophilic cellulose derivatives, polyethylene glycol, polyvinylpyrrolidone, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, polyvinyl acetate phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetal diethylaminoacetate, poly (methacrylic acid alkyl ester), poly (vinyl acetate), polymers derived from acrylic or methacrylic acid and their respective esters, and copolymers derived from acrylic or methacrylic acid and their respective esters.
Methods of preparing nanoparticle compositions are described, for example, in U.S. Pat. nos.5,518,187 and5,862,999, both being "Method of Grinding Pharmaceutical Substances"; U.S. Pat. No.5,718,388, both of which are "Continuous Method of Grinding Pharmaceutical Substances"; and U.S. Pat. No.5,510,118, "Process of Preparing Therapeutic Compositions Containing Nanoparticles".
Nanoparticle compositions are also described, for example, in U.S. patent No.5,298,262, "Use of Ionic Cloud Point Modifiers to Prevent Particle Aggregation During Sterilization"; U.S. Pat. No.5,302,401, "Method to Reduce Particle Size Growth During Lyophilization"; U.S. Pat. No.5,318,767, "X-Ray Contrast Compositions Useful in Medical Imaging"; U.S. Pat. No.5,326,552, "Novel Formulation For Nanoparticulate X-Ray Blood Pool Contrast Agents Using High Molecular Weight Non-ionic Surfactants"; U.S. Pat. No.5,328,404, "Method of X-Ray Imaging Using Iodinated Aromatic Propanedioates"; U.S. Pat. No.5,336,507, "Use of Charged Phospholipids to Reduce Nanoparticle Aggregation"; U.S. Pat. No.5,340,564, formulations Comprising Olin 10-G to Prevent Particle Aggregation and Increase Stability "; U.S. Pat. No.5,346,702, "Use of Non-Ionic Cloud Point Modifiers to Minimize Nanoparticulate Aggregation During Sterilization"; U.S. Pat. No.5,349,957, "Preparation and Magnetic Properties of Very Small Magnetic-Dextran Particles"; U.S. Pat. No.5,352,459, "Use of Purified Surface Modifiers to Prevent Particle Aggregation During Sterilization"; U.S. Pat. nos.5,399,363and5,494,683, both being "Surface Modified Anticancer Nanoparticles"; U.S. Pat. No.5,401,492, "Water Insoluble Non-Magnetic Manganese Particles as Magnetic Resonance Enhancement Agents"; U.S. Pat. No.5,429,824, "Use of Tyloxapol as a Nanoparticulate Stabilizer"; U.S. Pat. No.5,447,710, "Method for Making Nanoparticulate X-Ray Blood Pool Contrast Agents Using High Molecular Weight Non-ionic Surfactants"; U.S. Pat. No.5,451,393, "X-Ray Contrast Compositions Useful in Medical Imaging"; U.S. Pat. No.5,466,440, "Formulations of Oral Gastrointestinal Diagnostic X-Ray Contrast Agents in Combination with Pharmaceutically Acceptable Clays"; U.S. Pat. No.5,470,583, "Method of Preparing Nanoparticle Compositions Containing Charged Phospholipids to Reduce Aggregation"; U.S. Pat. No.5,472,683, "Nanoparticulate Diagnostic Mixed Carbamic Anhydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging"; U.S. Pat. No.5,500,204, "Nanoparticulate Diagnostic Dimers as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging"; U.S. Pat. No.5,518,738, "Nanoparticulate NSAID Formulations"; U.S. Pat. No.5,521,218, "Nanoparticulate Iododipamide Derivatives for Use as X-Ray Contrast Agents"; U.S. Pat. No.5,525,328, "Nanoparticulate Diagnostic Diatrizoxy Ester X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging"; U.S. Pat. No.5,543,133, "Process of Preparing X-Ray Contrast Compositions Containing Nanoparticles"; U.S. Pat. No.5,552,160, "Surface Modified NSAID Nanoparticles"; U.S. Pat. No.5,560,931, "Formulations of Compounds as Nanoparticulate Dispersions in Digestible Oils or Fatty Acids"; U.S. Pat. No.5,565,188, "Polyalkylene Block Copolymers as Surface Modifiers for Nanoparticles"; U.S. Pat. No.5,569,448, "Sulfated Non-ionic Block Copolymer Surfactant as Stabilizer Coatings for Nanoparticle Compositions"; U.S. Pat. No.5,571,536, "Formulations of Compounds as Nanoparticulate Dispersions in Digestible Oils or Fatty Acids"; U.S. Pat. No.5,573,749, "Nanoparticulate Diagnostic Mixed Carboxylic Anydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging"; U.S. Pat. No.5,573,750, "Diagnostic Imaging X-Ray Contrast Agents"; U.S. Pat. No.5,573,783, "Redispersible Nanoparticulate Film Matrices With Protective Overcoats"; U.S. Pat. No.5,580,579, "Site-specific Adhesion Within the GI Tract Using Nanoparticles Stabilized by High Molecular Weight, linear Polymers"; U.S. Pat. No.5,585,108, "Formulations of Oral Gastrointestinal Therapeutic Agents in Combination with Pharmaceutically Acceptable Clays"; U.S. Pat. No.5,587,143, "butyl Oxide-Ethylene Oxide Block Copolymers Surfactants as Stabilizer Coatings for Nanoparticulate Compositions"; U.S. Pat. No.5,591,456, "Milled Naproxen with Hydroxypropyl Cellulose as Dispersion Stabilizer"; U.S. Pat. No.5,593,657, "Novel Barium Salt Formulations Stabilized by Non-ionic and Anionic Stabilizers"; U.S. Pat. No.5,622,938, "Sugar Based Surfactant for Nanocrystals"; U.S. Pat. No.5,628,981, "Improved Formulations of Oral Gastrointestinal Diagnostic X-Ray Contrast Agents and Oral Gastrointestinal Therapeutic Agents"; U.S. Pat. No.5,643,552, "Nanoparticulate Diagnostic Mixed Carbonic Anhydrides as X-Ray Contrast Agents for Blood Pool and Lymphatic System Imaging"; U.S. Pat. No.5,718,388, "Continuous Method of Grinding Pharmaceutical Substances"; U.S. Pat. No.5,718,919, "Nanoparticles Containing the R (-) Enantiomer of Ibuprofen"; U.S. Pat. No.5,747,001, "Aerosols Containing Beclomethasone Nanoparticle Dispersions"; U.S. Pat. No.5,834,025, "Reduction of Intravenously Administered Nanoparticulate Formulation Induced Adverse Physiological Reactions"; U.S. Pat. No.6,045,829"Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors Using Cellulosic Surface Stabilizers"; U.S. Pat. No.6,068,858, "Methods of Making Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors Using Cellulosic Surface Stabilizers"; U.S. Pat. No.6,153,225, "Injectable Formulations of Nanoparticulate Naproxen"; U.S. Pat. No.6,165,506, "New Solid Dose Form of Nanoparticulate Naproxen"; U.S. Pat. No.6,221,400, "Methods of Treating Mammals Using Nanocrystalline Formulations of Human Immunodeficiency Virus (HIV) Protease Inhibitors"; U.S. Pat. No.6,264,922, "Nebulized Aerosols Containing Nanoparticle Dispersions"; U.S. Pat. No.6,267,989, "Methods for Preventing Crystal Growth and Particle Aggregation in Nanoparticle Compositions"; U.S. Pat. No.6,270,806, "Use of PEG-Derivatized Lipids as Surface Stabilizers for Nanoparticulate Compositions"; U.S. Pat. No.6,316,029, "Rapidly Disintegrating Solid Oral Dosage Form," U.S. Pat. No.6,375,986, "Solid Dose Nanoparticulate Compositions Comprising aSynergistic Combination of a Polymeric Surface Stabilizer and Dioctyl Sodium Sulfosuccinate"; U.S. Pat. No.6,428,814, "Bioadhesive nanoparticulate compositions having cationic surface stabilizers"; U.S. Pat. No.6,431,478, "Small Scale Mill"; and U.S. Pat. No.6,432,381, "Methods for targeting drug delivery to the upper and/or lower gastrointestinal tract," each of which is specifically incorporated by reference. Furthermore, U.S. patent application No.20020012675A1"Controlled Release Nanoparticulate Compositions," published at 31 of 1/2002, describes nanoparticle compositions and is specifically incorporated by reference.
Amorphous small particle compositions are described, for example, in U.S. patent No.4,783,484, "Particulate Composition and Use Thereof as Antimicrobial Agent"; U.S. Pat. No.4,826,689, "Method for Making Uniformly Sized Particles from Water-Insoluble Organic Compounds"; U.S. Pat. No.4,997,454, "Method for Making Uniformly-Sized Particles From Insoluble Compounds"; U.S. Pat. No.5,741,522, "Ultrasmall, non-aggregated Porous Particles of Uniform Size for Entrapping Gas Bubbles Within and Methods"; and U.S. Pat. No.5,776,496, "Ultrasmall Porous Particles for Enhancing Ultrasound Back Scatter".
Certain nanoformulations can reduce solubility problems by enhancing absorption of the drug by releasing the drug into the lumen in a controlled manner. The intestinal wall is intended to absorb nutrients and act as a barrier to pathogens and macromolecules. Small amphiphilic and lipophilic molecules can be absorbed by partitioning into lipid bilayers and by passive diffusion through intestinal epithelial cells, whereas the absorption of nano-formulations may be more complex due to the inherent nature of the intestinal wall. The first physical barrier to oral absorption of nanoparticles is the mucous barrier covering the intestinal and colonic luminal surfaces. The mucus barrier contains different layers, mainly consisting of heavily glycosylated proteins called mucins, which potentially prevent the absorption of certain nano-formulations. Modifications can be made to produce nanoformulations with enhanced mucus penetrating properties (Ensign et al, "Mucus penetrating nanoparticles: biophysical tool and method of drug and gene delivery", adv Mater 24:3887-3894 (2012)).
Once across the mucosa, transport of the nanofabric across intestinal epithelial cells can be mediated through several steps, including cell surface binding, endocytosis, intracellular trafficking, and exocytosis, resulting in transcytosis (transcellular transport), possibly involving multiple subcellular structures. Furthermore, the nanoformulation can also migrate between cells through an open tight junction, which is defined as endocytosis. The non-phagocytic pathway involves clathrin-mediated and fovea-mediated endocytosis and megaloblastic action, the most common mechanism of oral route absorption of the nano-formulation.
Non-oral administration may provide a variety of benefits, such as direct targeting of the desired site of action and prolonged drug action. Transdermal administration has been optimized for nanoformulations such as Solid Lipid Nanoparticles (SLN) and NE, which are characterized by good biocompatibility, low cytotoxicity and ideal modulation of drug release (Cappel and Kreuter, "Effect of nanoparticles on transdermal drug release.J microencapsulate 8:369-374 (1991)). Nasal administration of the nanoformulations allows them to penetrate the nasal mucosa by the transmucosal route of endocytosis or by a carrier or receptor mediated transport process (Illum, "Nanoparticulate systems for nasal delivery of drugs: a real improvement over simple systems. Pulmonary administration provides a large surface area and relatively easy access. The mucous barrier, metabolic enzymes of the tracheobronchial region and macrophages in the alveoli are often the major barriers to drug penetration. Particle size is the primary factor in determining the diffusion of the nanofabric in the bronchial tree, particles in the nanoscale region are more likely to reach the alveolar region, while particles between 1 and 5 μm in diameter are expected to deposit in the bronchioles (Musante et al, "Factors affecting the deposition of inhaled porous drug particles," JPharm Sci 91:1590-1600 (2002)). The absorption of larger particles is limited, possibly because of the inability to cross the qi-blood barrier. The particles may gradually release the drug, thereby penetrating into the blood stream, or the particles may be phagocytosed by alveolar macrophages (Bailey and Berkland, "Nanoparticle formulations in pulmonary drug delivery," med.res.rev.,29:196-212 (2009)).
Certain nano-formulations have very little permeability to biological membranes at the site of absorption, for which intravenous injection may be the preferred route to achieve effective distribution in vivo (Wacker, "Nanocarriers for intravenous injection-The long hard road to the market," int.j. Pharm.,457:50-62., 2013).
The distribution of the nanoformulations can vary greatly depending on the delivery system used, the nature of the nanoformulations, the variability between individuals, and the rate of drug loss of the nanoformulations. Certain nanoparticles, such as Solid Drug Nanoparticles (SDN), may improve drug absorption, which does not require them to reach the systemic circulation intact. Other nanoparticles survive the absorption process, thereby altering the distribution and clearance of the contained drug.
A nano-formulation of a certain size and composition may diffuse through a well-defined process, such as enhanced permeability and retention effects, in a tissue, while other nano-formulations accumulate in a specific cell population, thereby targeting a specific organ. The complex biological barrier can protect organs from exogenous compounds, while the Blood Brain Barrier (BBB) is a barrier to many therapeutic drugs. There are many different types of cells in the BBB, including endothelial cells, microglia, pericytes and astrocytes, which exhibit extremely tight junctions and highly active efflux mechanisms that limit penetration of most drugs. Transport across the BBB is generally limited to small lipophilic molecules and nutrients carried by a particular transporter. One of the most important mechanisms regulating diffusion of nanoformulations into the brain is endocytosis of brain capillary endothelial cells.
Recent studies correlate particle properties with the nanoparticle entry pathway and processing in the endothelial barrier of the human BBB, indicating that the permeation of uncoated nanoparticles into the BBB is limited, and that surface modification can affect the efficiency and mechanism of endocytosis (Lee et al, "Targeting rat anti-mouse transferrin receptor monoclonal antibodies through blood-brain barrier in mouse," J.Pharmacol. Exp. Ther.292:1048-1052 (2000)). Thus, surface-modified nanoparticles that cross the BBB and deliver one or more compounds described herein are within the scope of the invention.
Macrophages in the liver are the major source of total macrophages in the body. The Kupffer cell (Kupffer cell) in the liver possesses a large number of receptors for selectively phagocytizing opsonic particles (receptors for the crystallizable portion of the complement protein and IgG fragment). Phagocytosis may provide a mechanism (correctly.
Nanoparticles attached to polyethylene glycol (PEG) have minimal interaction with the receptor, thus inhibiting phagocytosis of the mononuclear phagocyte system (Bazile et al, "Stealth Me. PEG-PLA nanoparticles avoid uptake by the mononuclear phagocytes system," J. Pharm. Sci.84:493-498 (1995)).
Representative nanoformulations include inorganic nanoparticles, SDN, SLN, NE, liposomes, polymeric nanoparticles, and dendrimers. The compounds described herein may be contained within a nanofabricated formulation or, sometimes, attached to a surface as is the case with inorganic nanoparticles and dendrimers. Mixed nanoformulations containing elements of more than one nanoformulation class may also be used.
SDN is a lipid-free nanoparticle, which can improve the oral bioavailability and exposure of poorly water-soluble drugs (Chan, "Nanodrug particles and nanoformulations for drug delivery," Adv. Drug. Deliv. Rev.63:405 (2011)). SDN includes drugs and stabilizers and is produced using a 'top down' (high pressure homogenization and wet milling) or bottom up (solvent evaporation and precipitation) process.
SLN consists of a lipid (or lipids) that is solid at room temperature, an emulsifier, and water. Lipids utilized include, but are not limited to, triglycerides, partial glycerides, fatty acids, steroids, and waxes. SLNs are most suitable for transporting highly lipophilic drugs.
Droplets of less than 1000nm dispersed in an immiscible liquid are classified as NE. NE is used as a carrier for hydrophobic and hydrophilic agents and can be administered orally, transdermally, intravenously, intranasally, and ocularly. Chronic treatment may be preferred for oral administration, NE being effective in enhancing the oral bioavailability of small molecules, peptides and proteins.
The polymer nanoparticles are solid particles, typically about 200-800nm in size, may include synthetic and/or natural polymers, and may optionally be pegylated to minimize phagocytosis. The polymeric nanoparticles can improve the bioavailability of drugs and other substances compared to conventional formulations. Their elimination depends on several factors, including choice of polymer (including polymer size, polymer charge and targeting ligand), positively charged nanoparticles larger than 100nm are eliminated primarily by the liver (Alexis et al Factors affecting the clearance and biodistribution of polymeric nanoparticles mol Pharm 5:505-515 (2008)).
Dendrimers are tree-like nanostructured polymers, typically 10-20nm in diameter.
Liposomes are spherical vesicles that include a phospholipid bilayer. A variety of lipids can be used so that the degradation level can be controlled to some extent. In addition to oral administration, liposomes can also be administered in a variety of ways, including intravenous (mccoskill et al, 2013), transdermal (Pierre and Dos Santos Miranda Costa, 2011), intravitreal (Honda et al, 2013), and through the lung (chattopladhyy, 2013). Liposomes can be combined with synthetic polymers to form lipid-polymer hybrid nanoparticles, thereby expanding their ability to target specific sites in the body. The clearance of liposome-encapsulated drug is determined by drug release and destruction of the liposome (uptake, aggregation, pH-sensitive breakdown of liposomes by phagocytic immune cells, etc.) (Ishida et al, "Liposome clearance," Biosci Rep 22:197-224 (2002)).
One or more of these nanoparticle formulations may be used to deliver the active agents described herein to macrophages, across the blood-brain barrier, and other suitable locations.
Controlled release formulation
In a preferred embodiment, the active compound may be prepared with a carrier that prevents rapid elimination of the compound from the body, such as controlled release formulations including, but not limited to, implants and microencapsulated delivery systems. Biodegradable biocompatible polymers such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters and polylactic acid may be used. For example, enteric coating compounds may be used to prevent cracking by gastric acid. Methods of preparing such formulations will be apparent to those skilled in the art. Suitable materials are also commercially available.
Liposomal suspensions (including, but not limited to, liposomes with monoclonal antibodies to viral antigens targeted to infected cells) are also preferred as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811, incorporated by reference. For example, a liposome formulation can be prepared by: suitable lipids (e.g., stearoyl phosphatidylethanolamine, stearoyl phosphatidylcholine, arachidoyl phosphatidylcholine, and cholesterol) are dissolved in an inorganic solvent and then evaporated, leaving a dry lipid film on the surface of the container. An aqueous solution of the active compound is then introduced into the container. The vessel is then rotated by hand to release the lipid material from the sides of the vessel and disperse the lipid aggregates, thereby forming a liposome suspension.
The terminology used to describe the specific examples described herein is common and known to those skilled in the art. As used herein, the following abbreviations have the indicated meanings:
DMSO dimethyl sulfoxide
EtOAc ethyl acetate
h hours
Liq. liquid
M mole
MeOH methanol
min
RT or RT room temperature
TBAF tetrabutylammonium fluoride
THF tetrahydrofuran
General procedure for the preparation of active Compounds IX.
Methods for readily preparing active compounds are known in the art and result from selective combinations of known methods. The compounds disclosed herein may be prepared as described in detail below or by other methods known to those skilled in the art. It will be understood by those of ordinary skill in the art that changes in detail may be made without departing from the spirit of the invention and without in any way limiting the scope of the present disclosure.
For some compounds, the syntheses described herein are exemplary and may be used as starting points for preparing additional compounds of the formulae described herein. These compounds can be prepared in a variety of ways, including those synthetic schemes shown and described herein. Those skilled in the art will be able to recognize modifications of the disclosed compositions and design routes based on the disclosure herein; all such modifications and alternative routes are intended to be within the scope of the claims.
The various reaction schemes are summarized below.
Scheme 1 is a method of synthesizing nucleoside 3.
Scheme 2 is an enzymatic synthesis of compound 8.
In the schemes described herein, if the nucleobase includes functional groups that may interfere with, or be decomposed or otherwise converted during the reaction step, such functional groups may be protected using suitable protecting groups that may be removed. The protected functional group (if any) may be deprotected later.
Scheme 1: a method for synthesizing nucleoside 3.
The compound of formula 3 can be prepared from nucleoside 1 by: with, for example, acetone at H 2 SO 4 Selectively protecting 2',3' hydroxyl groups in the presence of a coupling agent such as EDC followed by coupling with the protected amino acid or in the presence of a base such as Et 3 Coupling with acid chloride, carbonate chloride or chloromethyl ester derivatives in the presence of N or NaH followed by appropriate deprotection.
Scheme 2. Enzymatic synthetic method of compound 8.
The compounds of formula 8 may also be prepared by modifying the chemistry described in ACS Omega 2021,6,15,10396-10402 and scheme 2.
The compounds of formulae a and B can also be prepared by modifying the chemistry described in the following: J.am.chem.Soc.2000,122,30,7233-7243; jmed, chem 2006,49,5,1624-1634; nucleosides, nucleosides & Nucleic Acids (2001), 20 (4-7), 743-746; european Journal of Organic Chemistry (1999), (3), 691-696; journal of Organic Chemistry (1971), 36 (1), 108-10; nucleosides & Nucleosides (1992), 11 (8), 1467-79; journal of Medicinal Chemistry (1975), 18 (8), 784-7; tetrahedron Letters (2006), 47 (4), 591-594.
The compounds of the general formulae C and F can also be prepared by modifying the chemistry described in the following: bioorganic & Medicinal Chemistry (2007), 15 (16), 5519-5528; chemistry & Biology (Oxford, united Kingdom) (2013), 20 (3), 416-423; nucleosides, nucleic Acids (2007), 26 (6-7), 573-577; WO2004096286A2; US20110288053 A1.
The compounds of formulae D and E can also be prepared by modifying the chemistry described in the following: WO 2021/159704A 1; journal of the Chemical Society, perkin Transactions 1:Organic and Bio-Organic Chemistry (1972-1999) (1991), (1), 43-8; bioorganic Chemistry (2015), 58,18-25; tetrahedron Letters (1985), 26 (37), 4467-70; journal of Medicinal Chemistry (2006), 49 (22), 6614-6620; collection of Czechoslovak Chemical Communications (1969), 34 (12), 3755-68; journal of the Chemical Society, perkin Transactions 1:Organic and Bio-Organic Chemistry (1972-1999) (1973), (7), 665-9; organic & Biomolecular Chemistry (2011), 9 (3), 676-678;
the monophosphate prodrugs of formulas a-F can also be prepared by modifying the chemistry described in the following: chem rev.2014;114 (18):9154-9218.
Deuterium incorporation:
It is expected that single or multiple replacement of hydrogen at the metabolic site in the sugar moiety of a nucleoside antiviral agent with deuterium (carbon-hydrogen bond to carbon-deuterium bond) will slow the metabolic rate. This may provide a relatively long half-life and slow body clearance. Slow metabolism of therapeutic nucleosides is expected to add additional advantages to therapeutic candidates, while other physical or biochemical properties are not affected. Intracellular hydrolysis or deuterium exchange releasable deuterium oxide (D 2 O)。
Methods for incorporating deuterium into amino acids, phenols, sugars and bases are well known to those skilled in the art. A representative process is disclosed in U.S. Pat. No.9,045,521.
Various enzymatic and chemical methods have been developed for deuterium incorporation in the sugar and nucleoside stages to provide high levels of deuterium incorporation (D/H ratio). Deuterium exchanged enzymatic methods typically incorporate lower levels. Enzymatic incorporation is of further complexity due to the cumbersome separation techniques required to separate deuterated mononucleotide blocks. Schmidt et al, ann.chem.1974,1856; schmidt et al, chem.Ber.,1968,101,590 describe 5',5' -) 2 H 2 Synthesis of adenosine prepared from 2',3' -O-isopropylidene adenosine-5 ' -carboxylic acid or from methyl-2, 3-isopropylidene- β -D-ribofuranuronic acid, dupre, m. and gaudiemer, a., tetrahedron lett.1978,2783.Kintanar et al, am.chem.Soc.1998,110,6367 reported the use of sodium or lithium aluminum borodeuteride (98 atom% 2 H incorporation) to reduce the appropriate 5' -aldehyde, a diastereomeric mixture of 5' -deuterated adenosine and 5' (R/S) -deuterated thymidine can be obtained. Berger et al, nucleoside&Nucleotides 1987,6,395 describe the presence of a polypeptide in 2 H 2 The aldehyde was heated in an O/pyridine mixture (1:1) and then NaBD was used 4 Reduction of the aldehyde converts the 5' -aldehyde derivative of 2' deoxyguanosine to 5' or 4' -deuterated-2 ' -deoxyguanosine.
Ajmer et al, labelled Compd.1986,23,963 describe obtaining 4' -deuterium labeled uridine and thymidine (98 atom% 2 H) Is a program of (a). Sinhabbu et al, J.am.chem.Soc.1985,107, 7628) demonstrate the stereoselective reduction of 1,2:5, 6-di-O-isopropylidene-beta-D-furohexa-3-ulose to 1,2:5, 6-di-O-isopropylidene-3-deutero-beta-D-hexose ribofuranose using sodium borodeuteride, followed by a further stepDuring the sugar synthesis, which is a step in nucleoside synthesis, deuterium is present in the C3' of adenosine (97 atom% 2 H) Is doped. Robin et al, org.chem.1990,55,410 reported that when 2 '-O-tert-butyldimethylsilyl (TBDMS) 3-one nucleosides were reduced with sodium borodeuteride in acetic acid, over 95 atomic% were synthesized at the C3' of adenosine 2 H is incorporated with almost complete stereoselectivity. David, S. And eustone, j., carbohyd.res.1971,16,46 and David, S. And eustone, j., carbohyd.res.1971,20,319 describe the synthesis of 2 '-deoxy-2' (S) -deutero-uridine and cytidine. Synthesis was performed using 1-methyl-2-deoxy-2' - (S) -deuterated furanoside.
Radatus et al, J.am.chem.Soc.1971,93,3086 describe chemical procedures for the synthesis of 2 '-mono-deuterated (R or S) -2' -deoxycytidine. These structures are synthesized from selective 2-mono-deutero-2-deoxy-D-ribose obtained by stereospecific reduction of 2, 3-dehydro-hexopyranose with lithium aluminum deuteride and oxidation of the resulting glyce. Wong et al J.am.chem.Soc.1978,100,3548 report on the formation of and LiAlD by involving dithioketene derivatives 4 The reaction sequence of the reduction gives deoxy-1-deutero-D-erythro-pentose, 2-deoxy-2 (S) -deuterium-D-erythro-pentose and 2-deoxy-1, 2 (S) -dideuterio-D-erythro-pentose from D-arabinose.
Pathak et al j., tetrahedron 1986,42,5427) report the status of the pass by LiAlD 4 Reduction opens the appropriate methyl 2, 3-dehydration-beta-D-ribofuranoside or beta-D-ribofuranoside to stereospecifically synthesize all eight 2' or 2' -deuterated-2 ' -deoxynucleosides. Wu et al j. Tetrahedron 1987,43,2355 describe the synthesis of all 2',2 "-dideutero-2 ' -deoxynucleosides (for deoxynucleosides and ribonucleosides), starting with C2' oxidation of the sugar, followed by NaBD 4 Or LiAlD 4 Reduced and then deoxygenated by tributyl deuterated tin. Roy et al J.am.chem.Soc.1986,108,1675 reported that 2',2' -dideutero-2 ' -deoxyguanosine and thymidine can be found in 2 H 2 O was prepared from 2-deoxyribose 5-phosphate using 2-deoxyribose 5-phosphate aldolase to about 90 atomic% deuteration. Similarly, 4',5',5' -, may be performed 2 H 3 -guanosine synthesis.
Thus, it is apparent that each position of the sugar residue can be selectively labeled.
A useful alternative to stereospecific deuteration was developed to synthesize polydextrose. The method is adopted in 2 H 2 The exchange of hydrogen with deuterium was performed in O using deuterated raney nickel catalysts at the carbon bearing the hydroxyl group (i.e., methylene and methine protons of the carbon bearing the hydroxyl group).
There are a variety of techniques available for synthesizing fully deuterated deoxyribonucleosides and ribonucleosides. Thus, in one method, deuterated Raney nickel- 2 H 2 Exchange of O with sugar produces a number of deuterated nucleosides specifically labeled at the 2',3' and 4' positions. The procedure consisted of: through Raney nickel- 2 H 2 O exchange reaction deuterated at 2',3' and 4 'positions of methyl beta-D-arabinopyranoside, and then reducing and eliminating' 2-hydroxy group by tributyl deuterated tin to obtain methyl beta-D-2 ',2',3',4' 2 H 4 2-pyrandeoxynucleosides, conversion to methyl beta-D-2 ',2',3',4' -) 2 H 4 2' -Furandeoxynucleosides and glycosylation to give various 2',2',3',4' - 2 H 4 Nucleosides (for H3 'and H4', >97 atom% 2 H incorporation.
The synthesis of deuterated phenols is described, for example, in Hoyer, h. (1950), synthesis des pan-deutero-o-nitro-phenols.chem.beer., 83:131-136. This chemistry can be used to prepare substituted phenols with deuterium labels. Deuterated phenols and substituted analogs thereof are useful, for example, in the preparation of phenoxy groups in phosphoramidate prodrugs.
The synthesis of deuterated amino acids is described, for example, in Matthews et al, biochimica et Biophysica Acta (BBA) -General Subjects, volume 497, stage 1, 1977, 3, month 29, pages 1-13. These and similar techniques can be used to prepare deuterated amino acids, which can be used to prepare phosphoramidate prodrugs of nucleosides described herein.
A method of synthesizing a deuterated analog of a compound described herein involves synthesizing a deuterated furannucleoside having a 1' -CN substitution; and ligating the nucleobase to a deuterated furannucleoside to form a deuterated nucleoside. Prodrugs, such as phosphoramidate prodrugs, may be formed by modifying the 5' -OH group on a nucleoside. When deuterated phenols and/or deuterated amino acids are used, deuterated phosphoramidate prodrugs may be prepared.
Another method involves synthesizing a furannucleoside having a 1' -CN substitution and ligating deuterated nucleobases to form a deuterated nucleoside. This method can optionally be performed using deuterated furanosides to provide additional deuteration. As with the methods described above, the nucleoside can be converted to a prodrug form, which can optionally include additional deuteration.
A third method involves synthesizing furannucleosides with 1' -CN substitutions, ligating nucleobases to form nucleosides, and converting the nucleosides to phosphoramidate prodrugs using one or both of deuterated amino acids or phenol analogs in the phosphoramidate synthesis.
Thus, using the techniques described above, one or more deuterium atoms can be provided in the sugar, base and/or prodrug moiety of the nucleoside compounds described herein.
DETAILED DESCRIPTION OF EMBODIMENT (S) OF INVENTION
Specific representative compounds described herein were prepared according to the following examples and reaction sequences; the examples and figures describing the reaction sequence are provided by way of illustration to aid in understanding the present disclosure and should not be construed to limit in any way the invention set forth in the claims that follow. The compounds of the present invention may also be used as intermediates in the subsequent examples to produce additional compounds described herein. There is no need to try to optimise the yields obtained in any of the reactions. Those skilled in the art will know how to increase such yields by conventional variations in reaction time, temperature, solvents and/or reagents.
Anhydrous solvents were purchased from Aldrich Chemical Company (Milwaukee, wis.) and EMD Chemicals (Gibbston, N.J.). Reagents were purchased from commercial sources. Unless otherwise indicated, the materials used in the examples were obtained from readily available commercial suppliers or synthesized by standard methods known to those skilled in the art of chemical synthesis. Melting points (mp) were determined on an electrothermal digital melting point meter and were uncorrected. 1 H and 13 the C NMR spectrum was taken at room temperature on a Varian Unity Plus 400 spectrometer and reported as low field ppm from the internal tetramethylsilane. Deuterium is carried outExchange, decoupling experiments or 2D-COSY to confirm proton distribution. Signal multiplexing is represented by s (singlet), d (doublet), dd (doublet), t (triplet), q (quartet), br (broad), bs (broad singlet), m (multiplet). All J values are in Hz. Mass spectra were determined on a Micromass Platform LC spectrometer using electrospray technique. Elemental analysis was performed by Atlantic Microlab company (Norcross, GA). Analytical TLC was performed on Whatman LK6F silica gel plate and preparative TLC was performed on Whatman PK5F silica gel plate. Column chromatography is performed on silica gel or by reverse phase high performance liquid chromatography.
Experiment
1- ((2R, 3R,4S, 5R) -3, 4-dihydroxy-5- (hydroxymethyl) tetrahydrothiophen-2-yl) pyrimidine-2, 4 (1H, 3H) -dione (8) and 4-amino-1- ((2R, 3R,4S, 5R) -3, 4-dihydroxy-5- (hydroxymethyl) tetrahydrothiophen-2-yl) pyrimidin-2 (1H) -one (9) are synthesized by following the procedure reported in the following: takashi Naka, noriaki Minakawa, hiroshi Abe, daisuke Kaga, and Akira Matsuda The Stereoselective Synthesis of 4' -beta-thioribonucleosides via the Pummerer Reaction J.am.chem.Soc.2000,122,30,7233-7243.
1- ((2R, 3R,4S, 5R) -3, 4-dihydroxy-5- (hydroxymethyl) tetrahydrothiophen-2-yl) pyrimidine-2, 4 (1H, 3H) -dione (8): 1 H NMR(400MHz,MeOH-d 4 )δ3.44-3.41(m,1H),3.82-3.80(m,2H),4.18(t,J=3.8Hz,1H),4.31-4.28(m,1H),5.78(d,J=8.1Hz,1H),6.07(d,J=6.3Hz,1H),8.23(d,J=8.1Hz,1H);MS(ESI):m/z[M+H] + C 9 H 13 N 2 O 5 calculated S261.3, experimental 261.0.
4-amino-1- ((2R, 3R,4S, 5R) -3, 4-dihydroxy-5- (hydroxymethyl) tetrahydrothiophen-2-yl) pyrimidin-2 (1H) -one (9): 1 H NMR(400MHz,MeOH-d 4 )δ3.84-3.43(m,1H),3.84(d,J=4.84Hz,2H),4.13-4.11(m,1H),4.23-4.21(m,1H),5.95(d,J=7.5Hz,1H),6.06(d,J=5.1Hz,1H),8.29(d,J=7.6Hz,1H)。MS(ESI):m/z[M+H] + C 9 H 14 N 3 O 4 calculated S value is 260.3, experimental value is 260.0.
Scheme 3: synthesis of Compound 13
1- ((3 aS,4R,6 aR) -6- (hydroxymethyl) -2, 2-dimethyl-tetrahydrothieno [3, 4-d)][1,3]Dioxol-4-yl) pyrimidine-2, 4 (1 h,3 h) -dione (10): to a solution of compound 8 (30 mg,0.115 mmol) in dry acetone (5 mL) was added 2, 2-dimethoxypropane (1.0 mL) and a catalytic amount of cc H 2 SO 4 . The reaction mixture was stirred at room temperature for 16 hours. Et is added to 3 N (0.5 ml) was added slowly to the mixture and the solvent was removed under reduced pressure, and the residue was purified by silica gel column chromatography (DCM/meoh=30:1-5:1) to give 10 (35 mg, 82%). 1 H NMR(400MHz,MeOH-d 4 )δ1.34(s,3H),1.57(s,3H),3.71-3.69(m,1H),3.83-3.81(m,2H),4.90-4.87(m,1H),4.96-4.94(m,1H),5.74(d,J=8.1Hz,1H),6.04(d,J=2.7Hz,1H),8.13(d,J=8.1Hz,1H)。MS(ESI):m/z[M+H] + C 12 H 17 N 2 O 5 Calculated S value is 301.3, experimental value is 301.0.
((S) - (((2R, 3S,4R, 5R) -5- (2, 4-dioxo-3, 4-dihydropyrimidin-1 (2H) -yl) -3, 4-dihydroxytetrahydrothiophen-2-yl) methoxy) (phenoxy) sulfonyl) -L-alanine isopropyl ester (13): to a stirred solution of 10 (15 mg,0.050mmol,1.00 eq.) in anhydrous DMF (1 mL) and THF (1 mL) at 0deg.C was slowly added tert-butylmagnesium chloride (200 μL,1M in THF, 4.0 eq.). After the addition, the mixture was stirred at 0 ℃ for 15 minutes. To the above mixture was added dropwise ((S) - (perfluorophenoxy) - (phenoxy) sulfonyl) -L-alanine isopropyl ester 11 (34 mg,0.075mmol,1.5 eq.) and the resulting mixture was stirred at room temperature for 24 hours, the mixture was quenched with cold water and the aqueous phase extracted with EtOAc (20 ml×3). The combined organic layers were purified by Na 2 SO 4 Drying and filtration, and removal of the solvent under reduced pressure gave crude compound 12. The crude compound 12 was dissolved in 2mL of 90% HCOOH/H 2 O (V: V) and stirred at room temperature for 4 hours. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography (DCM/meoh=30:1-10:1) to give 13 (9.0 mg, 50%). 1 H NMR(400MHz,MeOH-d 4 )δ1.27-1.24(m,6H),1.38-1.36(m,3H),3.59-3.58(m,1H),3.96-3.91(m,1H),4.18-4.17(m,1H),4.28-4.26(m,1H),4.38-4.32(m,2H),5.03-4.97(m,1H),5.72(d,J=8.1Hz,1H),6.08(d,J=6.4Hz,1H),7.30-7.23(m,3H),7.42-7.38(m,2H),8.05(d,J=8.1Hz,1H)。 31 P NMR(400MHz,MeOH-d 4 )δ3.44;MS(ESI):m/z[M+Na] + C 21 H 28 N 3 NaO 9 PS calculated 552.4, experimental 552.1.
Scheme 4: synthesis of Compound 16
4-amino-1- ((3 aS,4R,6 aR) -6- (hydroxymethyl) -2, 2-dimethyl-tetrahydrothieno [3, 4-d)][1,3]Dioxol-4-yl) pyrimidin-2 (1H) -one (14): to a solution of compound 9 (45 mg,0.174 mmol) in dry acetone (5 mL) was added 2, 2-dimethoxypropane (1.0 mL) and a catalytic amount of cc H 2 SO 4 . The reaction mixture was stirred at room temperature for 16 hours. Et is added to 3 N (0.5 ml) was slowly added to the mixture and the solvent removed under reduced pressure, and the residue was purified by silica gel column chromatography (DCM/meoh=30:1-5:1) to give 14 (34 mg, 65%). 1 H NMR(400MHz,MeOH-d 4 )δ1.34(s,3H),1.57(m,3H),3.70-3.68(m,1H),3.82-3.80(m,2H),4.90-4.88(m,1H),4.94-4.92(m,1H),5.97(d,J=7.5Hz,1H),6.09(d,J=2.3Hz,1H),8.14(d,J=7.5Hz,1H)。
((S) - (((2 r,3S,4r,5 r) -5- (4-amino-2-oxopyrimidin-1 (2H) -yl) -3, 4-dihydroxytetrahydrothiophen-2-yl) methoxy) (phenoxy) sulfonyl) -L-alanine isopropyl ester (16): to a stirred solution of nucleoside 14 (30 mg,0.100mmol,1.00 eq.) in anhydrous DMF (1 mL) and THF (2 mL) at 0deg.C was slowly added tert-butylmagnesium chloride (400 μL,0.400mmol,4.0 eq., 1M in THF). After the addition was completed, the mixture was stirred at 0 ℃ for 15 minutes. To the above mixture was added dropwise ((S) - (per fluorophenoxy) - (phenoxy) sulfonyl) -L-alanine isopropyl ester 11 (68 mg,0.150mmol,1.5 eq.) and the resulting mixture was stirred at room temperature for 16 hours. The mixture was quenched with cold water and the aqueous phase extracted with EtOAc (20 ml×3). The combined organic layers were purified by Na 2 SO 4 Drying and filtration, and removal of the solvent under reduced pressure gave crude compound 15. The crude compound 15 was dissolved in 4mL of 90% HCOOH/H 2 O (V: V) and stirred at room temperature for 4 hours. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography (DCM/meoh=30:1-10:1) to give 16 (9.1 mg, 59%). 1 H NMR(400MHz,MeOH-d 4 )δ1.26-1.24(m,6H),1.38-1.36(m,3H),3.64-3.60(m,1H),3.96-3.92(m,1H),4.16-4.14(m,1H),4.26-4.24(m,1H),4.40-4.33(m,2H),5.03-4.97(m,1H),5.97(d,J=7.5Hz,1H),6.12(d,J=5.7Hz,1H),7.30-7.21(m,3H),7.38-7.42(m,2H),8.13(d,J=8.0Hz,1H). 31 P NMR(400MHz,MeOH-d 4 )δ3.44;MS(ESI):m/z[M+H] + C 21 H 30 N 4 O 8 PS calculated 529.5, experimental 529.4.
Scheme 5: synthesis of Compound 17
Synthesis of 1- ((2R, 3S,4R, 5R) -3, 4-dihydroxy-5- (hydroxymethyl) -tetrahydrothiophen-2-yl) -4- (hydroxyamino) pyrimidin-2 (1H) -one (17) to 9 (12 mg,0.046mmol,1 eq.) in H 2 NH was added to the solution in O (2 mL) 2 HCl (64 mg,0.93mmol,20 eq.). The mixture was stirred at room temperature until TLC showed complete conversion of 9 to compound 17 (24 hours). The resulting mixture was concentrated under reduced pressure and the residue was purified using flash chromatography (DCM/MeOH system) to give 6.0mg of compound 9 in 47% yield. Colorless powder. 1 H NMR(400MHz,MeOH-d 4 )δ3.40(s,1H),3.81-3.73(m,2H),4.24-4.23(m,1H),4.32-4.30(m,1H),5.72(d,J=8.2Hz,1H),6.11(d,J=7.3Hz,1H),7.42(d,J=8.0Hz,1H)。MS(ESI):m/z[M+H] + C 9 H 14 N 3 O 5 S calculated 276.3, experimental 276.0.
Example 2
Cytotoxicity assays
As previously described, the formation was evaluated in Vero, human PBM, CEM (human lymphoblastic), MT-2 and HepG2 cellsToxicity of the compounds (see Schinazi r.f., sommadossi j. -p., saalmann v., cannon d.l., xie m. -y., hart g.c., smith g.a., and Hahn e.f., ankicrob. Agents chemther.1990, 34,1061-67). Cycloheximide (Cycloheximide) was included as a positive cytotoxicity control, while untreated cells exposed to the solvent were included as a negative control. Cytotoxic IC 50 Or CC 50 Are obtained from concentration-response curves using the median-effect methods described previously (see Chou T.—C. And Talalay P.adv. Enzyme Regul.1984,22,27-55; belen' kii M.S. and Schinazi R.F. Anti Res.1994,25, 1-11). The results are shown in table 8 below:
example 3
Mitochondrial toxicity assay in HepG2 cells:
i) Effects of compounds on cell growth and lactate production: the effect on HepG2cell growth can be determined by incubating the cells in the presence of 0 μm, 0.1 μm, 1 μm, 10 μm and 100 μm drug. Cells (5X 10 per well) 4 And) 12-well cell culture clusters were inoculated in a minimal essential medium supplemented with 10% fetal bovine serum, 1% sodium pyruvate and 1% penicillin/streptomycin containing nonessential amino acids and incubated for 4 days at 37 ℃. At the end of the incubation period, the cell number can be determined using a cytometer. Another example is Pan-Zhou X-R, cui L, zhou X-J, sommadossi J-P, darley-Usmer VM. "Differential effects of antiretroviral nucleoside analogs on mitochondrial function in HepG cells," Antimicrob. Agents chemther. 2000; 44:496-503.
To measure the effect of compounds on lactate production, hepG2cells from stock cultures can be diluted and at 2.5×10 per well 4 Individual cells were plated into 12-well plates. Different concentrations (0. Mu.M, 0.1. Mu.M, 1. Mu.M, 10. Mu.M and 100. Mu.M) of the compound can be added and the culture can be humidified at 37℃with 5% CO 2 Incubate in atmosphere for 4 days. On day 4, the number of cells per well can be determined and the medium collected. The medium may then be filtered and the lactic acid content of the medium determined using a lactic acid colorimetric assay (Sigma-Aldrich). Since lactic acid products can be considered as markers of impaired mitochondrial functionThis increase in lactate production levels detected in cells grown in the presence of the test compound is indicative of drug-induced cytotoxicity.
ii) effect of compound on mitochondrial DNA synthesis: a real-time PCR assay has been developed to accurately quantify mitochondrial DNA content (see Stuyver LJ, lostina S, adams M, mathew JS, pai BS, grier J, tharnish PM, choi Y, chong Y, choo H, chu CK, otto MJ, schinazi RF. Anti activities and cellular toxicities of modified ',3' -dideoxy-2',3' -dideoxyidine analyses, antibiotics chemther. 2002; 46:3854-60). This assay can be used in all studies described in this application to determine the effect of compounds on mitochondrial DNA content. In this assay, low passage number HepG2 cells were seeded at 5,000 cells/well in collagen coated 96-well plates. Test compounds were added to the medium to obtain final concentrations of 0. Mu.M, 0.1. Mu.M, 10. Mu.M and 100. Mu.M. On day 7 of culture, cell nucleic acids can be prepared using a commercially available column (RNeasy 96 kit; qiagen). These kits co-purify RNA and DNA, thus eluting total nucleic acid from the column. Mitochondrial cytochrome c oxidase subunit II (coxi) genes and β -actin or rRNA genes can be amplified from 5 μl of eluted nucleic acids using a multiplex Q-PCR protocol, as well as appropriate primers and probes for target and reference amplification. For coxi, the following sense, probe and antisense primers, respectively, can be used: 5'-TGCCCGCCA TCATCCTA-3', 5 '-tetrachloro-6-carboxyfluorescein-TCCTCATCGCCCTCCC ATCCC-TAMRA-3' and 5'-CGTCTGTTATGTAAAGGATGCGT-3'. For exon 3 of the β -actin gene (GenBank accession No. E01094), the sense, probe and antisense primers are 5'-GCGCGGCTACAGCTTCA-3', 5'-6-FAMCACC ACGGCCGAGCGGGATAMRA-3' and 5'-TCTCCTTAATGTCACGCAC GAT-3', respectively. Primers and probes for the rRNA gene are commercially available from Applied Biosystems. Since all genes obtained the same amplification efficiency, the comparative CT method can be used to study potential inhibition of mitochondrial DNA synthesis. The comparative CT method uses an arithmetic formula in which the amount of target (COXII gene) is normalized to the amount of endogenous reference (β -actin or rRNA gene) and to a calibrator (day 7 no drug control). The arithmetic formula of the method is given by 2- ΔΔCT, where ΔΔCT is (CT of average target test sample CT-target control) - (CT of average reference test CT-reference control) (see Johnson MR, K Wang, JB Smith, MJ Heslin, RB diasiol. Quantisation of dihydropyrimidine dehydrogenase expression by real-time reverse transcription polymerase chain reaction. Animal. Biochem.2000; 278:175-184). A decrease in mitochondrial DNA content in cells grown in the presence of the drug indicates mitochondrial toxicity.
Example 4
Mitochondrial toxicity-Glu/Gal
Scheme overview
HepG2 cells were plated on 96 or 384 well tissue culture polystyrene plates. After 24 hours, cells were given a range of concentrations of the test compound and incubated for 72 hours in galactose or glucose supplemented medium. If cells grown in galactose-containing media are more sensitive to the test compound than cells grown in glucose-containing media, the test compound is said to cause mitochondrial toxicity.
The object is: the sensitivity of HepG2 cells grown in galactose or glucose containing media to test compounds was measured.
Experimental procedure
HepG2 human hepatocellular carcinoma cells were plated on 96 or 384 well tissue culture polystyrene plates containing galactose or glucose containing medium supplemented with 10% fetal bovine serum and antibiotics and incubated overnight. Test compounds were given at increasing cell concentrations (final DMSO concentration of 0.5%; typical final test compound concentrations for the eight-point dose-response curve were 100, 30, 10, 3, 1, 0.3, 0.1, 0.03 μm; n = 3 replicates per concentration) and cells were incubated for 72 hours. The appropriate control was used as a quality control at the same time. Cell viability was measured using Hoechst staining and cell counts were performed by HCS reader.
Example 5
Mitochondrial toxicity assay in Neuro2A cells:
to estimate the potential of the compounds described herein to cause neuronal toxicity, mouse Neuro2A cells (American type culture Collection 131) can be used AS a model system (see Ray AS, hernandez-Santiago BI, mathew JS, murakami E, bozeman C, xie MY, dutschman GE, gullen E, yang Z, hurwitz S, cheng YC, chu CK, mcClure H, schinazi RF, anderson KS. Mechanical of anti-human immunodeficiency virus activity of beta-D-6-cyclopropylamino-2',3' -didehydro-2',3' -dideoxyguline. Anictirb. Agents Chemother 2005,49, 1994-2001). The concentration required to inhibit cell growth by 50% (CC 50) can be measured using a 3- (4, 5-dimethyl-thiazol-2-yl) -2, 5-diphenyltetrazolium bromide dye-based assay, as described. Cellular lactic acid and mitochondrial DNA levels can be perturbed at defined drug concentrations as described above. ddC and AZT can be used as control nucleoside analogues.
Example 6
Bone marrow cytotoxicity assay
Primary human bone marrow mononuclear cells are available from Cambrex Bioscience (Walkersville, MD). CFU-GM assays were performed using double-layered soft agar in the presence of 50 units/mL of human recombinant granulocyte/macrophage colony stimulating factor, while BFU-E assays were performed using ethylcellulose matrices containing 1 unit/mL of erythropoietin (see Sommadosis JP, carlisle R.toxity of 3' -azido-3' -deoxythymidine and- (1, 3-dihydroxy-2-pro-xymethyl) guanine for normal human hepatopoietic progenitor cells in vitro. Antimicrob. Agents chemther. 1987;31:452-454; sommadosis, JP, schinazi, RF, chu, CK, and Xie, MY. Comparon of cytotoxicity of the (-) and (+) enantiomer of 2',3' -dide-3 ' -thiacytidine in normal human bone marrow progenitor cells biochem. Pharmacol.1992; 44:1-5). Each experiment can be repeated in cells from three different donors. AZT was used as a positive control. The cells can be contacted with 5% CO at 37deg.C in the presence of the compound 2 Incubate for 14-18 days and colonies of more than 50 cells can be counted using an inverted microscope to determine IC 50 .50% Inhibitory Concentration (IC) 50 ) Can be obtained by least squares linear regression analysis of drug concentration logarithms and BFU-E survival scores. Statistical analysis can be performed using student t-test of independent unpaired samples.
Example 7
In vitro human mitochondrial RNA Polymerase (POLRMT) assay
An in vitro RNA nucleotide incorporation assay can be performed as described previously using POLRMT (INDIGO Biosciences) (Arnold et al 2012). In the short term, the term "a" is used, 32 the P radiolabeled RNA primer (5 '-UUUUGCCGCGCC) can be used with a 3 molar excess of the appropriate DNA template (5' -GGGAATGCA)NGGCGCGGC, where position N may be replaced by A, T or C). 125nM POLRMT can be combined with 500nM 5' -radioactive RNA/DNA hybrid, 10mM MgCl 2 Incubated with 100. Mu.M of the corresponding nucleoside triphosphates. For non-nucleoside analogues, 100 μm inhibitor can be added at the same time as 100 μm UTP. The incorporation can be carried out at 30℃for 2 hours and the reaction stopped by adding 10mM EDTA and formamide. The samples were visualized on a 20% denaturing polyacrylamide gel. The data may be analyzed by normalizing the product fraction of each nucleoside triphosphate analog relative to the product fraction of the corresponding natural nucleoside triphosphate.
Example 8
Effect of nucleotide analogs on DNA polymerase and exonuclease Activity of mitochondrial DNA polymerase gamma
i) Purification of human polymerase gamma: recombinant large and small subunits of polymerase gamma can be purified as described previously (see Graves SW, johnson AA, johnson KA. Expression, purification, and initial kinetic characterization of the large subunit of the human mitochondrial DNA polymerase biochemistry 1998,37,6050-8;Johnson AA,Tsai Y,Graves SW,Johnson KA.Human mitochondrial DNA polymerase holoenzyme:reconstitution and characterization.Biochemistry 2000;39:1702-8). The protein concentration can be determined spectrophotometrically at 280m, and the extinction coefficients of the polymerase gamma large subunit and small subunit are 234,420 and 71,894M-1cm-1, respectively.
ii) kinetic analysis of nucleotide incorporation: pre-steady state kinetic analysis can be performed to determine the catalytic efficiency (K/K) of DNA polymerase γ for incorporation of nucleoside-TP and natural dNTP substrates. This allows the relative ability of the enzyme to incorporate modified analogues to be determined and the toxicity predicted. Pre-homeostasis of gamma-incorporation of nucleotide analogs into DNA polymeraseKinetic analysis was performed essentially AS described previously (see Murakami E, ray AS, schinazi RF, anderson KS. Invested the effects of stereochemistry on incorporation and removal of-fluorocytidine analogs by mitochondrial DNA polymerase gamma: compactison of D-and L-D4FC-TP. Anti Res.2004,62,57-64;Feng JY,Murakami E,Zorca SM,Johnson AA,Johnson KA,Schinazi RF,Furman PA,Anderson KS.Relationship between antiviral activity and host toxicity:comparison of the incorporation efficiencies of 2',3' -dideoxy-5-fluoro-3' -thiacytidine-triphosphate analogs by human immunodeficiency virus type 1reverse transcriptase and human mitochondrial DNA polymerase.Antimicrob Agents Chemother.2004,48,1300-6). Briefly, a pre-incubation mixture of large (250 nM) and small (1.25 mM) subunits of polymerase gamma and 60nM DNA template/primer in 50mM Tris-HCl, 100mM NaCl pH 7.8 can be added to a MgCl-containing solution 2 (2.5 mM) and various concentrations of nucleotide analogs. The reaction can be quenched and analyzed as described previously. The data may be fitted to the same equation as described above.
iii) Determination of human polymerase γ3'5' exonuclease activity: human polymerase gamma exonuclease activity can be studied by measuring the rate of cleavage product formation in the absence of dntps. By mixing MgCl 2 (2.5 mM) was added to a pre-incubation mixture of polymerase gamma large subunit (40 nM), small subunit (270 nM) and 1,500nM of chain-terminating template/primer in 50mM Tris-HCl, 100mM NaCl pH 7.8 to initiate the reaction and quenched with 0.3M EDTA at the indicated time points. All reaction mixtures were analyzed on a 20% denaturing polyacrylamide sequencing gel (8M urea), imaged on a Bio-Rad GS-525 molecular imaging system, and quantified using Molecular Analyst (Bio-Rad). The product formed from the early time points will be plotted as a function of time. The data will be fitted by linear regression of Sigma Plot (Jandel Scientific). The slope of the line can be divided by the concentration of active enzyme in the reaction to calculate the kexo (see Murakami E, ray AS, schinazi RF, anderson KS. Investing the effects of stereochemistry on incorporation and removal of-flu) of exonuclease activity orocytidine analogs by mitochondrial DNA polymerase gamma:comparison of D-and L-D4FC-TP.Antiviral Res.2004;62:57-64;Feng JY,Murakami E,Zorca SM,Johnson AA,Johnson KA,Schinazi RF,Furman PA,Anderson KS.Relationship between antiviral activity and host toxicity:comparison of the incorporation efficiencies of 2’,3’-dideoxy-5-fluoro-3’-thiacytidine-triphosphate analogs by human immunodeficiency virus type 1reverse transcriptase and human mitochondrial DNA polymerase.Antimicrob Agents Chemother.2004;48:1300-6)。
Example 9
Inhibition of human DNA polymerase by NTP
Research objective
Determining whether nucleoside triphosphates analogs inhibit human DNA polymerase α, β, and γ and calculating IC 50 Values.
Materials and methods
Human DNA polymerase α -the enzyme can be purchased from Chimerx (catalog No. 1075) and assayed with some modifications according to their recommendations. 2' -Me-UTP was treated with inorganic pyrophosphatase (Sigma) to remove any pyrophosphate contamination. The final concentration of 500. Mu.M 2' -Me-UTP can be combined with 1mM DTT, 50mM Tris, 50mM NaCl, 6mM MgCl 2 And 1 unit of pyrophosphatase was incubated at 37℃for 1 hour, and then inactivated at 95℃for 10 minutes. A mixture of 0.05 units of human DNA polymerase α and 5' end radiolabeled 24nt DNA primer (5 ' -TCAGGTCCCTGTTCGGGCGCCACT) and 48nt DNA template (5 ' -CAGTGTGGAAAATCTCTAGCAGTGGCGCCCGAACAGGGACCTGAAAGC) can be mixed with compounds increasing in concentration from 0 to 100. Mu.M in 60mM Tris-HCl (pH 8.0), 5mM magnesium acetate, 0.3mg/ml bovine serum albumin, 1mM dithiothreitol, 0.1mM spermine, 0.05mM each of dCTP, dGTP, dTTP, dATP in a final reaction volume of 20. Mu.l at 37℃for 5 minutes (all concentrations represent the final concentration after mixing). The reaction can be stopped by mixing with 0.3M (final) EDTA. The product was separated on a 20% polyacrylamide gel and quantified on a Bio-Rad molecular imager FX. Experimental results may be formulated into dose-response formulas using Graphpad Prism or SynergySoftware Kaleidagraph The equation (ymin+ ((ymax) - (ymin)))/(1+ (compound concentration)/IC 50 ) Slope) to determine IC 50 Values. The data may be normalized to the control.
Human DNA polymerase β -can be purchased from Chimerx (catalog No. 1077) and assayed with some modifications according to their recommendations. A mixture of 0.1 units of human DNA polymerase beta and 5' -end radiolabeled 24nt DNA primer (5 ' -TCAGGTCCCTGTTCGGGCGCCACT) annealed to 48nt DNA template (5 ' -CAGTGTGGAAAATCTCTAGCAGTGGCGCCCGAACAGGGACCTGAAAGC) may be mixed with a compound increasing in concentration from 0 to 100. Mu.M in 50mM Tris-HCl (pH 8.7), 10mM KCl, 10mM MgCl 2 0.4mg/ml bovine serum albumin, 1mM dithiothreitol, 15% (v/v) glycerol, each 0.05mM dCTP, dGTP, dTTP, dATP, with a final reaction volume of 20. Mu.l, at 37℃for 5 minutes (all concentrations represent the final concentration after mixing). The reaction can be stopped by mixing with 0.3M (final) EDTA. The product can be separated on a 20% polyacrylamide gel and quantified on a Bio-Rad molecular imager FX. Experimental results can be fit to a dose response equation ymin+ ((ymax) - (ymin)))/(1+ (compound concentration)/IC using Graphpad Prism or SynergySoftware Kaleidagraph 50 ) Slope) to determine IC 50 Values. The data may be normalized to the control.
Human DNA polymerase γ -the enzyme can be purchased from Chimerx (catalog No. 1076) and assayed with some modifications according to their recommendations. A mixture of 0.625 units of human DNA polymerase gamma and 5' -end radiolabelled 24nt DNA primer (5 ' -TCAGGTCCCTGTTCGGGCGCCACT) annealed to 36nt DNA template (5 ' -TCTCTAGAAGTGGCGCCCGAACAGGGACCTGAAAGC) may be mixed with a compound increasing in concentration from 0 to 100. Mu.M in 50mM Tris-HCl (pH 7.8), 100mM NaCl, 5mM MgCl 2 And 0.05mM each of dCTP, dGTP, dTTP, dATP, the final reaction volume was 20. Mu.l, at 37℃for 200 minutes (all concentrations represent the final concentration after mixing). The reaction can be stopped by mixing with 0.3M (final) EDTA. The product can be separated on a 20% polyacrylamide gel and quantified on a Bio-Rad molecular imager FX. Experimental results can be fitted to a dose response equation (ymin+ ((y vero) - (y min))) using Graphpad Prism or SynergySoftware Kaleidograph(1+ (Compound concentration)/IC 50 ) Slope) to determine IC 50 Values. The data may be normalized to the control.
Cell pharmacology of HepG2 cells
HepG2 cells were obtained from the American type culture Collection (Rockville, md.) and were grown at 225cm 2 The tissue culture flasks were grown in minimal essential medium supplemented with nonessential amino acids, 1% penicillin-streptomycin. The medium was refreshed every three days and the cells were subcultured once a week. After the adherent monolayer was detached by exposing it to 30mL of trypsin-EDTA for 10 min and washing it 3 times continuously with medium, confluent HepG2 cells were plated at 2.5×10 per well 6 The density of individual cells was seeded in 6-well plates and exposed to 10. Mu.M [ for a specified period of time ] 3 H]Labeled active compound (500 dpm/pmol).
The cells maintained 5% CO at 37 ℃C 2 Under an atmosphere. At selected time points, cells were washed three times with ice-cold Phosphate Buffered Saline (PBS).
The intracellular active compounds and their respective metabolites were extracted by incubating the cell pellet with 60% methanol at-20 ℃ overnight, followed by extraction with an additional 20 pal cold methanol in an ice bath for one hour. The extracts were then combined, dried under a gentle stream of filtered air and stored at-20 ℃ until HPLC analysis.
Example 11
Cytopharmacology of Vero, calu-3 and Caco-2 cells
Vero, calu-3 and Caco-2 cells were seeded at 1X 106 per well in 12-well plates and incubated in a cell incubator at 37℃in a humidified atmosphere of 5% CO 2. The adherent cells were then exposed to 10. Mu.M RS-3995. After 4 hours, the drug-containing medium was removed and the cells were washed twice with ice-cold Phosphate Buffered Saline (PBS). Cells were resuspended in 70% ice-cold methanol containing 20nM ddATP overnight at-20 ℃. The supernatant was then dried under air flow and the dried samples were stored at-20 ℃ until analysis by LC-MS/MS. Each sample was reconstituted in 200 μl of mobile phase prior to analysis.
Chromatographic separation and detection were performed on a Vanquish Flex system (Thermo Scientific, waltham, mass.) coupled to a TSQ Quantiva triple quadrupole mass spectrometer (Thermo Scientific, waltham, mass.). Analytes were separated using a Kinetex EVO-C18 column (100 x 2.1mm,2.6 μm) (Phenomenex, torrance, CA) at a flow rate of 250 μl/min. Mobile phase a consisted of 2mM monoammonium phosphate and 3mM hexylamine in water and mobile phase B consisted of acetonitrile. The LC gradient increased from 2% to 60% of mobile phase B over 7 minutes and then returned to the initial conditions. Selected reaction monitoring (spray voltage: 3200V (positive) or 2500V (negative), sheath gas: 35Arb, assist gas: 20Arb, ion transport tube temperature: 350 ℃ C., evaporator temperature: 380 ℃ C.) in positive and negative modes was used to detect targets.
Table 1: levels of Compound 8 Monophosphate (MP), diphosphate (DP) and Triphosphate (TP) in Vero, calu-3 and Caco-2 cells after 4 hours.
BLOQ: below a quantitative level
Example 12
MERS assay
Cells and viruses:
human lung cancer cells (A-549) are useful in primary antiviral assays and are available from the American type culture Collection (ATCC, rockville, md., USA). Cells can be cultured in minimal essential medium supplemented with 10% fetal bovine serum (containing 0.15% NaCHO 3 MEM of (2); hyclone Laboratories, logan, utah, USA). When evaluating the efficacy of the compounds, the final concentration of serum can be reduced to 2%, and the medium can contain 50 μg/mL gentamicin (Sigma-Aldrich, st.louis, mo.). Since MERS-Co virus does not produce a detectable viral cytopathic effect, viral replication in a549 cells can be detected by titration of the viral supernatant of infected, compound-treated a549 cells in Vero76 cells.
Vero76 cells are available from ATCC and can be routinely introduced with 0.15% NaCHO supplemented with 5% fetal bovine serum 3 Is a MEM of (a). In evaluating compounds, one canThe final concentration of serum was reduced to 2% and supplemented with 50. Mu.g/mL gentamicin.
The middle east coronavirus strain EMC (MERS-CoV) is the original isolate of human, amplified in cell culture by Ron foundation (Erasmus Medical Center, rotterdam, the Netherlands) and obtained from the disease control center (Atlanta, ga.).
Control:
(Interferon alfacon-1, a recombinant non-naturally occurring type I interferon (Blatt, L. Et al, J. Interferon Cytokine Res. (1996) 16 (7): 489-499 and Alberti, A., biodrugs (1999) 12 (5): 343-357) can be used as a positive control drug in all antiviral assays. Intergen=0.03 ng/mL.
Antiviral assay:
the virus can be diluted in MEM to multiplicity of infection = 0.001, and each compound can be diluted in mem+2% fbs using a half-log 8 dilution series. Compounds may be first added to 96-well plates of pooled a549 cells, followed by viral addition over 5 minutes. Inhibition of each test compound dilution can be assessed in triplicate. After plating, the plates may be incubated for 4 days at 37 ℃. The plates may then be frozen at-80 ℃.
Virus yield reduction assay:
the infectious viral yield per well in an antiviral assay can be determined. Each plate of the antiviral assay can be thawed. Sample wells for each compound concentration tested can be pooled and the titer of infectious virus determined by CPE assay in Vero 76 cells. Wells can be CPE scored and virus titers calculated. The reduction in virus yield by 90% can then be calculated by regression analysis. This represents a titer of 1log compared to untreated virus control 1 0 inhibition.
Determining efficacy of Compounds against HCoV-OC43 and SARS-CoV-2 infection
Virus (virus)
HCoV-OC43 is obtained from ATCC (Manasas, va.), SARS-CoV-2 is obtained from BEIResources (NR-52281: USA-WA/2020). HCoV-OC43 and SARS-CoV-2 are propagated in appropriate cells and passed through TCID, respectively 50 Titration was performed and aliquots were then stored at-80 ℃ until further use.
Cells and medium:
for cytotoxicity and antiviral studies, the following immortalized/transformed cell lines were used: human colon epithelial cells (Caco-2;HTB-37 TM manassas, VA, USA), human bronchial epithelial cells (Calu-3; />HTB-55 TM Manassas, VA, USA), human small alveolar cells expressing the human ACE-2 receptor by lentiviral transduction (A549) hACE2 The method comprises the steps of carrying out a first treatment on the surface of the Available from dr. Susan Weiss donation (Lei et al 2021)) and african green monkey kidney cells (Vero; />CCL-81 TM Manassas, VA, USA). The culture medium consists of (1) Caco-2 and Calu-3: isagl minimum essential medium (Eagle's minimum essential medium, EMEM), 10% Fetal Bovine Serum (FBS), 100U/mL penicillin-streptomycin (pen-strep) and 2 μ M L-glutamine (L-glut), (2) A549 hACE2 And Vero: dalberg modified eagle Medium (Dulbecco' smodified eagle medium, DMEM), 10% FBS, 100U/mL pen-strep. Additional studies were performed in differentiated primary normal human bronchus/tracheas cells (NHBE) obtained from individual donors per culture (Lonza Biosciences CC-2540s, basal, switzerland) cultured in 3D via a standard gas-liquid interface (ALI; stemCell Technologies 2021) or as custom-made apical lung organoids (HBO; lee and LeCher et al, unpublished). HBTEC in customization of Pneuma-felt TM Amplified in Ex Plus medium (Stem Cell Technologies, vancouver, B.C.), and in custom Pneuma-felt supplemented with hydrocortisone and heparin sulfate TM ALI medium (ALI; stem Cell Technologies, vancouver, B.C.) or Pneuma-Cult TM OrganoidsDifferentiation in apical medium (HBO; stem Cell Technologies, vancouver, B.C.). For all experiments, the cells were at 37℃and 95% O 2 、5% CO 2 Growing in an incubator.
Antiviral screening assay:
for standard antiviral screening, cells (Caco-2, calu-3 monolayer, ace-2 h549 And Vero) in 96-well plates to confluence (1X 10) 5 Individual cells). By treating cells with 2-fold serial dilutions (0-10. Mu.M) of the compound of interest in respective basal media containing 2% heat-inactivated FBS (ΔFBS) and then with 0.1 (Vero) or 1.0 (Caco 2, calu-3, A549) hACE2 ) MOI infection with SARS-CoV2 was performed for 48 hours (Vero) or 72 hours (Caco 2, calu-3, A549) hACE2 ) A dose response curve is plotted. The cells/supernatant were collected in 150. Mu.L of RLT buffer @Hilden, germany) for downstream RNA extraction (RNeasy 96 extraction kit; />Hilden, germany) and subsequent qRT-PCR to detect viral load.
Advanced antiviral assays were also performed on lead compounds in ALI-Calu-3, ALI-NHBE and HBO-NHBE by dose response assays with the following modifications: ALI-Calu 3-will be 1.8X10 4 Individual cells were seeded into 96-well 1.0 μm well transwell inserts (Corning, USA). After 3 days, the medium was removed from the apical chamber and the cells were cultured in ALI for an additional week. ALI-NHBE-will be 1.5X10 5 Individual cells were seeded onto 24-well collagen-coated 0.4 μm-well transwell inserts (Corning, USA). After 3 days, the medium was removed from the apical chamber and the cells were cultured in ALI for an additional 3 weeks. For both ALI cultures, compounds were added to the basal outside chamber at the indicated dilutions. Cells were washed 3 times with HEPES buffer saline (HBSS) on the apical surface to remove excess mucus, then infected by adding 50. Mu.L of SARS-CoV-2 virus (MOI 1.0) to the apical chamber, adsorbed for 5 hours, then virus was removed and cells were retained in ALI for an additional 3 days. For the followingHBO culture, 3X 10 3 The individual cells are inoculated in the medium containingIn a hanging drop suspension of basement membrane matrix (Corning, USA), individual organoids were produced per well and cultured for 21 days. Serial dilutions of compound and virus (MOI 1.0) were added directly to the wells for a period of 3 days. Cultures infected with Calu3-ALI and HBO were collected in 150. Mu.L of RLT buffer (.sup.L)>Hilden, germany) and NHBE-ALI cultures were collected in 300. Mu. LTril TM RNA was extracted from the reagents by phenyl-chloroform method (ThermoFisher Scientific, USA) according to the manufacturer's protocol. All infections were performed in BSL-3 grade laboratories of the University of Ememory according to the guidelines of fifth edition Biosafety in Microbiological and Biomedical Laboratories. All experiments were performed independently in triplicate or in triplicate.
SARS-CoV-2 yield inhibition assay by qRT-PCR assay:
virus yield inhibition assays (Zandi et al 2020) were performed as described above. Briefly, viral RNA was detected by real-time PCR using 6-carboxyfluorescein (FAM) labeled probes and primers directed against SARS-CoV2 nonstructural protein 3 (nsp 3). (SARS-CoV-2FWD:AGA AGA TTG GTT AGA TGA TGA TAG T;SARS-CoV-2REV:TTC CAT CTC TAA TTG AGG TTG AAC C;SARS-CoV-2 Probe: 56-FAM/TC CTC ACT GCC GTC TTG TTG ACC A/3BHQ_1) RNA isolated from uninfected cells was used as a negative control for virus detection. RNA was added to Mastermix (qScript) TM XLT one-step RT-qPCRQuantabio, USA) and run on a StepOne Plus real-time PCR (Roche, germany) according to the manufacturer's protocol. Calculation of C from duplicate groups T The values were then used to quantify virus yield by standard curve. Median Effective Concentrations (EC) of compounds were calculated using GraphPad Prism version 7 (GraphPad Software company, san Diego, CA) 50 ) And a concentration having 90% inhibitory effect (EC 90 ) And reported as mean ± standard deviation.
Inhibition of SARS-CoV-2 production by neon green reporter gene:
virus yield inhibition assays were performed as described previously (Tao et al 2021). Briefly, cells were infected at MOI 0.1 (Vero) with or without the above compound, but using an icSARS-CoV-2-mNG infectious clone expressing neon green (Xie et al 2020). Cultures in control wells were monitored daily for neon green expression. 48 hours (Vero), 72 hours (Caco-2, calu-3, A549) hACE2 After either ALI-NHBE and HBO-NHBE) or 96 hours (ALI-NHBE and HBO-NHBE), all wells were imaged and the antiviral activity of the compounds was determined as a percentage reduction in average relative fluorescence relative to the control.
Images were obtained using one of two methods: (1) Live cells were imaged using LAX software (Leica Biosystems) on a Leica FC 7000GT microscope with pE-300 fluorescent lamp cover, images were processed with Image J software and cells were collected in RLT buffer for downstream qRT-PCR, or (2) cells were fixed directly in 4% paraformaldehyde for 30 min to be removed from BSL3, permeabilized in 1% ND-40-PBS buffer, DAPI counterstained, and imaged on a cell-imaging multimode reader of the station 5 and quantified on Gen5 software (Biotek, winooski, VT). Uninfected wells were measured as negative controls for background fluorescence.
anti-SARS-CoV-2 activity is shown in tables 2-5 below, and toxicity data is shown in Table 6 below:
TABLE 2
ND: is not determined
TABLE 3 Table 3
TABLE 4 Table 4
TABLE 4 Table 4
TABLE 5
Table 6: toxicity of Compound 8 in Caco2 and Calu3 cells
Quantitative and non-compartmental PK analysis of compound 8 in mouse plasma samples:
the method comprises the following steps:
administering compound 8 to CD-1 mice by PO (30 mg/kg,3 mice) or IV (15 mg/kg,3 mice). Plasma samples were collected at 30 minutes, 2 hours, 4 hours and 7 hours.
Sample preparation:
mix 20 μl mouse plasma with 100 μl MeOH.
Air-drying the supernatant and then reconstitution in 200 μ L H O.
-performing LC-MS analysis.
-calibration curve range: 50nM to 100. Mu.M.
LC-MS/MS conditions:
-an instrument: TSQ Quantiva, column: kineex XB-C8 (50X 2.1mm,2.6 μm)
LC buffer: a) The method comprises the following steps 0.1% formic acid, and B): acetonitrile
LC gradient: 0-0.3 min, 2% B;0.3-3 minutes, 2% -80% of B;3-3.2 minutes, 80% B;3.2-3.5 minutes, 80% -2% of B;3.5-8 min, 2% B
Compound 8 mitochondrial toxicity in HepG2 cells:
CC for MtDNA 50 >100. Mu.M (inhibition at 100. Mu.M)<1%)
-CC 50 rDNA>100. Mu.M (36.32% inhibition at 100. Mu.M)
Reference is made to:
Li,Y.,Renner,D.M.,Comar,C.E.,Whelan,J.N.,Reyes,H.M.,Cardenas-Diaz,F.L.,and Weiss,S.R.(2021).SARS-CoV-2 induces double-stranded RNA-mediated innate immune responses in respiratory epithelial-derived cells and cardiomyocytes.Proceedings of the National Academy of Sciences,118(16).
Stem Cell Technologies.Model the human airway in vitro as ALI cultures or airway organoids.
Lee,J.H.and LeCher,J.C.,et al(2021).Apical-out human bronchial organoid models for SARS-CoV-2 infaction studies.Unpublished Study.
Zandi,K.,Amblard,F.,Musall,K.,Downs-Bowen,J.,Kleinbard,R.,Oo,A.,and Schinazi,R.F.(2020).Repurposing nucleoside analogs for human coronaviruses.Antimicrobial agents and chemotherapy,65(1),e01652-20.
Tao,S.,Zandi,K.,Bassit,L.,Ong,Y.T.,Verma,K.,Liu,P.,and Schinazi,R.F.(2021).Comparison of anti-SARS-CoV-2 activity and intracellular metabolism of remdesivir and its parent nucleoside.Current Research in Pharmacology and Drug Discovery,2,100045.
Xie,X.,Mumato,A.,Lokugamage,K.G.,Narayanan,K.,Zhang,X.,Zou,J.,and Shi,P.Y.(2020).An infectious cDNA clone of SARS-CoV-2.Cell host&microbe,27(5),841-848.
evaluation in the SARS-CoV-2 infection model of the human lung epithelial and monocyte systems:
in vitro migration experiments and Virus infection H441 Club cell line was carried out on an Alvetex scaffold (ReproCELL, glasgow, UK) coated with rat tail collagen (Sigma) in 50/50DMEM/F12 with 2% v/v Ultroser G (Crescent Chemical, islandia, N.Y.) at the gas-liquid interfaceGrowing for 2 weeks. The filter was then turned over and placed in fresh medium at the bottom of the well. Virus (PR 8: A/Puerto Rico/8/1934; OC43; or NR-52281, SARS-CoV-2 isolate USA-WA 1/2020) WAs added to the medium so that the multiplicity of infection (MOI) WAs 0.1 and incubated for 24 hours. This setup requires manual flipping of the filter prior to migration, a subtle process performed under BSL3 conditions. Thus, epithelial cells must be infected when the cells are submerged and no longer in ALI, which may introduce artifacts reminiscent of pneumonia. The filters were transferred to RPMI medium containing LTB4 (100 nM) and CCL2 (250 pg/mL), whether or not additional drug was added. The final concentration of drug was 1 or 10uM. Untreated conditions contained 0.01% v/v DMSO as vehicle control. Blood mononuclear cells were purified using RosetteSep (StemCell). About 10 in total 6 Individual cells were loaded onto an Alvetex scaffold for migration for 24 hours. After migration, triPure (Roche) was added to the epithelial cells and frozen at-80 ℃.
Plasma stability:
450 μl of human, mouse or hamster plasma was exposed to 10 μΜ compound and incubated at 37 ℃. At 0 min, 5 min, 15 min, 30 min, 60 min, 90 min and 120 min, 50 μl of plasma samples were mixed with 200 μl of ice-cold methanol (70%). 50. Mu.L of the supernatant was dried and incubated at 100. Mu. L H 2 And (3) recovering in O. As positive control, propancaline bromide (Propantheline bromide) was used. The supernatants were then subjected to LC-MS analysis (LC-MS conditions: instrument: thermo TSQ quantiva. Chromatographic column: kinetex C88 (50X 2.1mm,2.6 μm.) LC buffer: A): 0.1% formic acid, and B): acetonitrile.
Cell pharmacology:
uptake and excretion of compounds was measured in cell cultures of HAE cells as well as a variety of other cells. HAE cells involved in cell culture were cultured at 0.15X10 6 Density of individual cells/well, other cells were seeded at 1×10 6 Density inoculation of individual/wells. To measure uptake, the compound was incubated in cells at a concentration of 10 μm for 4 hours. To measure the amount of compound discharged from cells, fine particles were prepared Cells were pre-treated at a concentration of 10 μm for 24 hours, at which time the medium was changed, and then harvested at 0, 2, 4, 6, 8, 12, 24 and 32 hours.
LC-MS/MS: TSQ Quantiva. Buffer a:2mM NH 3 H 2 PO 4 And 3mM hexylamine; buffer B: acetonitrile; flow rate: 250. Mu.L/min. HPLC column: kineex EVO c18×2.1mm,2.6 μm. MS detection: SRM mode.
The scope of the invention is not limited by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are also intended to fall within the scope of the appended claims.
Various publications are cited herein, the disclosures of which are incorporated by reference in their entirety.

Claims (158)

1. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering a therapeutic or preventing amount of a compound of formula (a) or (A1) to a patient in need of treatment or prevention:
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
y and R are independently selected from the group consisting of: H. OH, halo, optionally substituted O-linked amino acid, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR ', SR ', wherein each R ' is independently-C (O) -C 1-12 Alkyl, -C (O) -C 2-12 Alkenyl, -C (O) -C 2-12 Alkynyl, -C (O) -C 3-6 Cycloalkyl, -C (O) O-C 1-12 Alkyl, -C (O) O-C 2-12 Alkenyl, -C (O) O-C 2-12 Alkynyl, -C (O) O-C 3-6 Cycloalkyl, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl and C 3-6 Cycloalkyl, wherein the group may be substituted with one or more substituents selected from the group consisting of: halogen (fluorine, chlorine, bromine or iodine), hydroxy, amino, alkylamino, arylamino, alkoxy, nitro and cyano,
R 1 is and R 1A H, CH independently 3 、CH 2 F、CHF 2 Or CF (CF) 3 Wherein when R is 1 Where Me, the carbon to which it is attached may be all or part of R or S or any mixture thereof, or R 1 And R is 1A Can be combined to form C 3-7 A cycloalkyl ring;
R 2 is H, CN, N 3 、F、CH 2 Halogen, CH 2 -N 3 、O-CH 2 -P-(OH) 3 Substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl or substituted or unsubstituted C 2-8 Alkynyl;
R 3 selected from the group consisting of: H. f, N 3 Substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl, O- (C) 1-8 ) Alkyl and N 3
R 5 Is S, the number of the groups is S,
R 8 and R is 8’ Independently selected from the group consisting of: H. OH, halo, optionally substituted O-linked amino acid, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR ', SR ', wherein each R ' is independently-C (O) -C 1-12 Alkyl, -C (O) -C 2-12 Alkenyl, -C (O) -C 2-12 Alkynyl, -C (O) -C 3-6 Cycloalkyl, -C (O) O-C 1-12 Alkyl, -C (O) O-C 2-12 Alkenyl, -C (O) O-C 2-12 Alkynyl, -C (O) O-C 3-6 Cycloalkyl, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-6 Cycloalkyl, wherein the group may be substituted with one or more substituents selected from the group consisting of: halogen (fluorine, chlorine, bromine or iodine), hydroxy, amino, alkylamino, arylamino, alkoxy, nitro and cyano,
R 4 is OH, an optionally substituted O-linked amino acid, -O-C (O) -C 1-12 Alkyl, -O-C (O) -C 2-12 Alkenyl, -O-C (O) -C 2-12 Alkynyl, -O-C (O) -C 3-6 Cycloalkyl, -O-C (O) O-C 1-12 Alkyl, -O-C (O) O-C 2-12 Alkenyl, -O-C (O) O-C 2-12 Alkynyl, -O-C (O) O-C 3-6 Cycloalkyl, OC 1-6 Alkyl, OC 1-6 Haloalkyl, OC 1-6 Alkoxy, OC 2-6 Alkenyl, OC 2-6 Alkynyl, OC 3-6 Cycloalkyl, O-P (O) R 6 R 7 、O-CH 2 -P-(OH) 3 、O-CH 2 -P-(OH) 3 Or mono-, di-or triphosphate, wherein, when R 4 When chiral is present in the phosphorus center of (C), it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 and R is 7 Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl and heteroaryl groups, such as phenyl and pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, carbon chain derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl;
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl;
the base is selected from the group consisting of:
X 1 is CH, C- (C) 1-6 ) Alkyl, C- (C) 2-6 ) Alkenyl, C- (C) 2-6 ) Alkynyl, C- (C) 3-7 ) Cycloalkyl, C- (C) 1-6 ) Haloalkyl, C- (C) 1-6 ) Hydroxyalkyl, C-OR 22 、C-N(R 22 ) 2 C-halo, C-CN or N,
X 1’ is CH, C- (C) 1-6 ) Alkyl, C- (C) 2-6 ) Alkenyl, C- (C) 2-6 ) Alkynyl, C-halo, C-CN or N
R 9 And X 2 Independently H, OH, NH 2 Halo (i.e. F, cl, br or I), SH, NHOH, O (C 1-10 ) Alkyl, O (C) 2-10 ) Alkene, O (C) 2-10 ) Alkyne, O (C) 3-7 ) Cycloalkyl, -O-C (O) -C 1-12 Alkyl, -O-C (O) -C 2-12 Alkenyl, -O-C (O) -C 2-12 Alkynyl, -O-C (O) -C 3-6 Cycloalkyl, -O-C (O) O-C 1-12 Alkyl, -O-C (O) O-C 2-12 Alkenyl, -O-C (O) O-C 2-12 Alkynyl, -O-C (O) O-C 3-6 Cycloalkyl, S (C) 1-10 ) Alkyl, S (C) 2-10 ) Alkene, S (C) 2-10 ) Alkyne, S (C) 3-7 ) Cycloalkyl, optionally unsaturated NH (C) 1-10 ) Alkyl, optionally unsaturated N ((C) 1-10 ) Alkyl group 2 、NH(C 3-7 ) Cycloalkyl, optionally unsaturated NH (CO) (C 1-20 ) Alkyl, optionally unsaturated NH (CO) O (C 1-20 ) Alkyl, NHOH, optionally unsaturated NHO (CO) (C 1-20 ) Alkyl or optionally unsaturated NHO (CO) NH (C 1-20 ) Alkyl, (C) 1-3 ) An alkyl group, a hydroxyl group,
R 9’ is OH, NH 2 、SH、NHOH、-O-C(O)-C 1-12 Alkyl, -O-C (O) -C 2-12 Alkenyl, -O-C (O) -C 2-12 Alkynyl, -O-C (O) -C 3-6 Cycloalkyl, -O-C (O) O-C 1-12 Alkyl, -O-C (O) O-C 2-12 Alkenyl, -O-C (O) O-C 2-12 Alkynyl or-O-C (O) O-C 3-6 A cycloalkyl group,
R 10 is H or F, and is not limited to the above,
X 2’ is N or CH, and
w is O or S.
2. The method of claim 1, wherein R 2 Is H or substituted or unsubstituted C 2-8 Alkynyl groups.
3. The method of claim 1, wherein R 3 Is H.
4. The method of claim 1, wherein R 1 Is and R 1A Is H.
5. The method of claim 1, wherein R 8 And R is 8’ Is OH.
6. The method of claim 1, wherein R 4 Is OH or O-P (O) R 6 R 7
7. The method of claim 1, wherein the base is
8. The method of claim 8, wherein R 9’ Is OH, NH 2 Or NHOH
9. The method of claim 1, wherein the base is
10. The method of claim 10, wherein X 2 Is NH 2 OH or SH.
11. The method of claim 1, wherein the compound is
12. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering to a patient in need of treatment or prevention a therapeutic or preventing amount of a compound of formula (B) or (B1):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, Y, R, R 1 、R 1A 、R 2 、R 3 、R 5 And R is 8’ As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl, and heteroaryl groups, such as phenyl and pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 NaphtheneRadical, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
13. The method of claim 12, wherein R 2 Is H or substituted or unsubstituted C 2-8 Alkynyl groups.
14. The method of claim 12, wherein R 3 Is H.
15. The method of claim 12, wherein R 8’ Is OH.
16. The method of claim 12, wherein Y is H.
17. The method of claim 12, wherein R 1 And R is 1A Is H.
18. The method of claim 12, wherein a is O.
19. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering to a patient in need of treatment or prevention a therapeutic or preventing amount of a compound of formula (C) or (C1):
Or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R、R 1 、R 1A 、R 2 、R 3 、R 5 、R 8 、R 8’ and Y is as defined in formula A,
x is OH, NH 2 、SH、NHOH、-O-C(O)-C 1-12 Alkyl, -O-C (O) -C 2-12 Alkenyl, -O-C (O) -C 2-12 Alkynyl, -O-C (O) -C 3-6 Cycloalkyl, -O-C (O) O-C 1-12 Alkyl, -O-C (O) O-C 2-12 Alkenyl, -O-C (O) O-C 2-12 Alkynyl or-O-C (O) O-C 3-6 A cycloalkyl group,
z is H or F, and
w is O or S.
20. The method of claim 19, wherein R 2 Is H or substituted or unsubstituted C 2-8 Alkynyl groups.
21. The method of claim 19, wherein R 3 Is H.
22. The method of claim 19, wherein R 8 And R is 8’ Is OH.
23. The method of claim 19, wherein Y is H.
24. The method of claim 19, wherein R is H.
25. The method of claim 19, wherein Z is H.
26. The method of claim 19, wherein X is OH, NH 2 Or NHOH.
27. The method of claim 19, wherein W is O.
28. The method of claim 19, wherein R 1 And R is 1A Is H.
29. The method of claim 19, wherein R 4 Is OH or O-P (O) R 6 R 7
30. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering a therapeutic or preventing amount of a compound of formula (D) or (D1) to a patient in need of treatment or prevention:
Or a pharmaceutically acceptable salt or prodrug thereof, wherein R, R 1 、R 1A 、R 2 、R 3 、R 5 、R 8’ And Y is as defined in formula A, and A and D are as defined in formula C.
31. The method of claim 30, wherein R 2 Is H or substituted or unsubstituted C 2-8 Alkynyl groups.
32. The method of claim 30, wherein R 3 Is H.
33. The method of claim 30, wherein R 8’ Is OH.
34. The method of claim 30, wherein Y is H.
35. The method of claim 30, wherein R is H.
36. The method of claim 30, wherein Z is H.
37. The method of claim 30, wherein X is OH, NH 2 Or NHOH.
38. The method of claim 30, wherein W is O.
39. The method of claim 30, wherein R 1 And R is 1A Is H.
40. The method of claim 30, wherein R 4 Is OH or O-P (O) R 6 R 7
41. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering to a patient in need of treatment or prevention a therapeutic or preventing amount of a compound of formula (E) or (E1):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 1A 、R 2 、R 3 And R is 4 As defined in the formula a above,
R 30 is S, the number of the groups is S,
R 31 is O or S, and is preferably selected from the group consisting of,
when R is 30 When S is R 31 Is O, and
R 32 and R is 33 H, F, C independently 1 -C 3 Alkyl, C 2 -C 3 Alkene or C 2 -C 3 Alkynes.
42. The method of claim 41, wherein R is 31 Is O.
43. The method of claim 41, wherein R is 32 And R is 33 Independently H or F.
44. The method of claim 41, wherein R is 3 Is H.
45. The method of claim 41, wherein R is 2 Is N 3 Or substituted or unsubstituted C 2-8 Alkynyl groups.
46. The method of claim 41, wherein R is 1 And R is 1A Is H.
47. The method of claim 41, wherein R is 4 Is OH or O-P (O) R 6 R 7
48. The method of claim 41, wherein the base is
49. The method of claim 41, wherein X 1 Is N.
50. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families, comprising administering to a patient in need of treatment or prevention a therapeutic or preventing amount of a compound of formula (F) or (F1):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 1A 、R 2 、R 3 And R is 4 As defined in the formula a above,
R 34 is S, and
R 35 and R is 36 Is independently H, F or CH 3
51. The method of claim 50, wherein R is 35 And R is 36 Is H.
52. The method of claim 50, wherein R is 34 Is CH 2
53. The method of claim 50, wherein R is 4 Is OH or O-P (O) R 6 R 7
54. The method of claim 50, wherein R is 3 Is H.
55. The method of claim 50, wherein R is 2 Is H or substituted or unsubstituted C 2-8 Alkynyl groups.
56. The method of claim 50, wherein R is 1 And R is 1A Is H.
57. The method of any one of claims 1-56, wherein the compound is capable of being present in the β -D or β -L configuration.
58. The method of any one of claims 1-56, wherein the virus is a coronavirus.
59. The method of claim 58, wherein the coronavirus is SARS-CoV2, MERS, SARS or OC-43.
60. The method of claim 58, wherein the coronavirus is SARS-CoV2.
61. The method of any one of claims 1-61, wherein the compound is co-administered with one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
62. The method of claim 61, wherein the compound is administered with adefovir, N-hydroxycytidine, or pharmaceutically acceptable salts or prodrugs thereof.
63. The method of claim 61, wherein the additional active compound is a JAK inhibitor and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib, or baratinib, or a pharmaceutically acceptable salt or prodrug thereof.
64. The method of claim 61, wherein the one or more additional active agents comprise an anticoagulant or a platelet aggregation inhibitor.
65. The method of claim 61, wherein the one or more additional active agents comprise an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
66. The use of a compound according to any one of claims 1 to 56 for the manufacture of a medicament for the treatment or prophylaxis of infections of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families.
67. The use of claim 66, wherein the infection is a coronaviridae infection.
68. The use of claim 67, wherein said coronavirus is SARS-CoV2, MERS, SARS or OC-43.
69. The use of claim 67, wherein said coronavirus is SARS-CoV2.
70. The use of claim 66, wherein said medicament further comprises one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
71. The use of claim 66, wherein said medicament further comprises adefovir, N-hydroxycytidine, or pharmaceutically acceptable salts or prodrugs thereof.
72. The use of claim 66, wherein the medicament further comprises a JAK inhibitor and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib or baratinib, or a pharmaceutically acceptable salt or prodrug thereof.
73. The use of claim 66, wherein said medicament further comprises an anticoagulant or a platelet aggregation inhibitor.
74. The use of claim 66, wherein the medicament further comprises an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
75. The use of claim 66, wherein said drug is a transdermal composition or a nanoparticle composition.
76. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (a):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R 1 is H, deuterium, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 2-8 Alkynyl or N 3
R 2 And R is 2’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR 7 And SR (Surfural) 7
Each R 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R ', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R ', optionally substituted-C (NR ') OR' optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
provided that R is 2 And R is 2’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy, carboxyl, C 1-12 Acyl, aryl, heteroaryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrazine, carbamate, phosphonic acid, phosphonate, boric acid, and borate;
R 3 And R is 3’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl,OR 7 And SR (Surfural) 7 Wherein each R is 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
Wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy, carboxyl, C 1-12 Acyl, aryl, heteroaryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrazine, carbamate, phosphonic acid, phosphonate, boric acid, and borate;
provided that R is 3 And R is 3’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R 4 Selected from the group consisting of: H. deuterium, CN, halo, N 3 Substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl, substituted or unsubstituted (C 1-8 ) Haloalkyl and N 3
R 5 Is and R 5’ H, CH independently 3 、CH 2 F、CHF 2 Or CF (CF) 3 Wherein when R is 5 Is CH 3 When it is attached to carbon, it may be all or part of R or S or any mixture thereof, or R 5 And R is 5’ Can be combined to form C 3-7 A cycloalkyl ring;
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) D-amino acidsOr esters of L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group;
the base is selected from the group consisting of:
y is H or a halogen group,
x is N or CH, and the number of the N or the CH,
w is O or S, and the R is selected from the group consisting of O and S,
X 1 and X 1’ Is independently CH, C- (C) 1-6 ) Alkyl, C- (C) 2-6 ) Alkenyl, C- (C) 2-6 ) Alkynyl, C- (C) 3-7 ) Cycloalkyl, C- (C) 1-6 ) Haloalkyl, C- (C) 1-6 ) Hydroxyalkyl, C-OR 22 、C-N(R 22 ) 2 C-halo, C-CN or N,
X 2 and X 2’ Independently H, halo, OR 9’ Or NR (NR) 10 R 10’
R 9’ Is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxy Benzylic esters, optionally substituted (acyloxybenzylic) esters, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
R 10 and R is 10’ Independently are H, OH, L-amino acid amide, D-amino acid amide, (acyloxybenzyl) amine, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, PEG amide, PEG carbamate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', lipid amide, optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Or lipid carbamates wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), restriction stripsThe member is R 10 And R is 10’ And cannot be OH.
77. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (B):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 2 、R 2’ 、R 3 、R 4 、R 5 R 5’ 、R 7 And R is 8 As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, ringA heteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl group; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl group, and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
78. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (C):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
base, R 1 、R 2 、R 2’ 、R 3 And R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein when chiral at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 7 Is an L-amino acid ester, a D-amino acid ester, an N-substituted L-amino acid ester, an N-substituted D-amino acid ester, an N, N-disubstituted L-amino acid ester, an N, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, an optionally substituted bis-acyloxybenzyl ester, an optionally substituted (acyloxybenzyl) ester, an optionally substituted-C (O) -C 1- 12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 Cycloalkyl group, and
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl group,Optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl or substitutedHeteroaryl of (a); wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
79. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (D):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is selected from the group consisting of:
/>and is also provided with
X 1 、X 1’ 、X 2’ 、X 2 、R 2 、R 2’ 、R 3 、R 3’ 、R 4 、R 5 、R 5’ And R is 6 As defined in formula a.
80. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (E):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
The base is selected from the group consisting of:
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 、R 4 、R 5 and R is 5’ As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl group、C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl group, and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
81. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (F):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is selected from the group consisting of:
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 and R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein when chiral at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionallysubstituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl group:C 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
82. The process of claim 76 wherein R is 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
83. The process of claim 77 wherein R is 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
84. The process of claim 78 wherein R is 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H.
85. The method of claim 79, wherein R is 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
86. The process of claim 80 wherein R is 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
87. The process of claim 81 wherein R is 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H.
88. The process of claim 76 wherein R is 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
89. The process of claim 77 wherein R is 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
90. The process of claim 78 wherein R is 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionallysubstituted-O-C (O) -C 1-12 An alkyl group.
91. The method of claim 79, wherein R is 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
92. The process of claim 80 wherein R is 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
93. The process of claim 81 wherein R is 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
94. The method of claim 76, wherein said compound is one of the following:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt or prodrug thereof.
95. The method of claim 77, wherein said compound is one of the following:
/>
or a pharmaceutically acceptable salt or prodrug thereof.
96. The method of claim 76, wherein said compound is one of the following:
or a pharmaceutically acceptable salt or prodrug thereof.
97. The method of claim 94, wherein said compound is one of the following:
/>
or a pharmaceutically acceptable salt or prodrug thereof.
98. The method of claim 76, wherein the compound isOr pharmaceutically acceptable thereofIs a salt or prodrug of (a).
99. The method of any one of claims 76-98, wherein the compound is capable of being present in the β -D or β -L configuration.
100. The method of any one of claims 76-98, wherein the virus is a coronavirus.
101. The method of claim 100, wherein the coronavirus is human coronavirus 229E, SARS, MERS, SARS-CoV-1, OC43, or SARS-CoV-2.
102. The method of claim 100, wherein the coronavirus is SARS-CoV2.
103. The method of any one of claims 76-98, wherein the compound is co-administered with one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
104. The method of claim 103 wherein the compound is administered with adefovir, N-hydroxycytidine, mo Nuola, PF-07321332, PF-07304814, or a pharmaceutically acceptable salt or prodrug thereof.
105. The method of claim 103, wherein the additional active compound is a JAK inhibitor and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib, or baritinib, or a pharmaceutically acceptable salt or prodrug thereof.
106. The method of claim 103, wherein the one or more additional active agents comprise an anticoagulant or a platelet aggregation inhibitor.
107. The method of claim 103, wherein the one or more additional active agents comprise an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
108. The use of a compound of any one of claims 76-98 for the manufacture of a medicament for the treatment or prevention of an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families.
109. The use of claim 108, wherein the infection is a coronaviridae infection.
110. The use of claim 109, wherein the coronavirus is human coronavirus 229E, SARS, MERS, SARS-CoV-1, OC43, or SARS-CoV-2.
111. The use of claim 109, wherein the coronavirus is SARS-CoV2.
112. The use of claim 108, wherein the medicament further comprises one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
113. The use of claim 108, wherein the medicament further comprises adefovir, N-hydroxycytosine, mo Nuola, PF-07321332, PF-07304814, or a pharmaceutically acceptable salt or prodrug thereof.
114. The use of claim 108, wherein the medicament further comprises a JAK inhibitor, and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib, or baritinib, or a pharmaceutically acceptable salt or prodrug thereof.
115. The use of claim 108, wherein the medicament further comprises an anticoagulant or a platelet aggregation inhibitor.
116. The use of claim 108, wherein the medicament further comprises an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
117. The use of claim 108, wherein the medicament is a transdermal composition or a nanoparticle composition.
118. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (a):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
R 1 is H, deuterium, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 2-8 Alkynyl or N 3
R 2 And R is 2’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR 7 And SR (Surfural) 7
Each R 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
provided that R is 2 And R is 2’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy, carboxyl, C 1-12 Acyl, aryl, heteroaryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, acidAnhydrides, oximes, hydrazines, carbamates, phosphonic acids, phosphonates, boric acid and borates;
R 3 and R is 3’ Independently selected from the group consisting of: H. deuterium, OH, SH, NH 2 Halo, substituted or unsubstituted C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 3-6 Cycloalkyl, cyano, cyanoalkyl, azido, azidoalkyl, OR 7 And SR (Surfural) 7 Wherein each R is 7 Independently L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -C 1-12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 A cycloalkyl group,
wherein the optional substituents are selected from the group consisting of: halo, C 1-12 Haloalkyl, C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl, C 3-7 Cycloalkyl, hydroxy, carboxyl, C 1-12 Acyl, aryl, heteroAryl, C 1-6 Acyloxy, amino, amido, carboxyl derivatives, alkylamino, di-C 1-12 Alkylamino, arylamino, C 1-12 Alkoxy, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamoyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrazine, carbamate, phosphonic acid, phosphonate, boric acid, and borate;
provided that R is 3 And R is 3’ Can not be all OH, SH or NH 2 、OR 7 Or SR (S.J) 7
R 4 Selected from the group consisting of: H. deuterium, CN, halo, N 3 Substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl, substituted or unsubstituted (C 1-8 ) Haloalkyl and N 3
R 5 Is and R 5’ H, CH independently 3 、CH 2 F、CHF 2 Or CF (CF) 3 Wherein when R is 5 Is CH 3 When it is attached to carbon, it may be all or part of R or S or any mixture thereof, or R 5 And R is 5’ Can be combined to form C 3-7 A cycloalkyl ring;
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 And R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group; />
The base is
Y is H or a halogen group,
x is N or CH, and the number of the N or the CH,
w is O or S, and the R is selected from the group consisting of O and S,
X 1 and X 1’ Is independently CH, C- (C) 1-6 ) Alkyl, C- (C) 2-6 ) Alkenyl, C- (C) 2-6 ) Alkynyl, C- (C) 3-7 ) Cycloalkyl, C- (C) 1-6 ) Haloalkyl, C- (C) 1-6 ) Hydroxyalkyl, C-OR 22 、C-N(R 22 ) 2 C-halo, C-CN or N,
X 2 and X 2’ Independently H, halo, OR 9’ Or NR (NR) 10 R 10’
R 9’ H, L-amino acid esters, D-amino acid esters, N-substituted L-amino acid esters, N-substituted D-amino acid esters, N-disubstituted L-amino acid esters, N, N-disubstituted D-amino acid esters, (acyloxybenzyl) ethers, optionally substituted bis-acyloxybenzyl) esters, optionally substituted (acyloxybenzyl) esters, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy),
R 10 and R is 10’ Independently are H, OH, L-amino acid amide, D-amino acid amide, (acyloxybenzyl) amine, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R ', optionally substituted-C (S) S-R', optionally substituted C 1-12 Alkyl, optionally substituted C 2-12 Alkenyl, optionally substituted C 2-12 Alkynyl, optionally substituted C 3-6 Cycloalkyl, PEG amide, PEG carbamate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted -CH 2 -CH 2 -S-C (O) -R ', lipid amide, optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Or lipid carbamates wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy) with the proviso that R 10 And R is 10’ And cannot be OH.
119. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (B):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is
R 1 、R 2 、R 2’ 、R 3 、R 4 、R 5 R 5’ 、R 7 And R is 8 As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three groups Each independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl, and->
(c)Wherein R is 30 Selected from the group consisting ofGroup: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
120. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (C):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is
R 1 、R 2 、R 2’ 、R 3 And R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein when chiral at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 7 is an L-amino acid ester, a D-amino acid ester, an N-substituted L-amino acid ester, an N-substituted D-amino acid ester, an N, N-disubstituted L-amino acid ester, an N, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, an optionally substituted bis-acyloxybenzyl ester, an optionally substituted (acyloxybenzyl) ester, an optionally substituted-C (O) -C 1- 12 R ', optionally substituted-C (O) O-R', optionally substituted-C (O) S-R '; optionally substituted-C (S) S-R': optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 And optionally substituted-O-C (O) N (R') 2 PEG esters, PEG carbonates, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 S-C (O) -R', lipid esters or lipid carbonates,
wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionallySubstituted C 12-22 Alkoxy),
r' is C 1-16 Alkyl, C 2-16 Alkenyl, C 2-16 Alkynyl or C 3-7 Cycloalkyl group, and
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
121. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (D):
Or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is
X 1 、X 1’ 、X 2’ 、X 2 、R 2 、R 2’ 、R 3 、R 3’ 、R 4 、R 5 、R 5’ And R is 6 As defined in formula a.
122. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (E):
or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 、R 4 、R 5 And R is 5’ As defined in the formula a above,
a is O or S, and
d is selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, aryl such as phenyl, and heteroaryl such as pyridyl, wherein aryl and heteroaryl are optionally substituted with zero to three substituents independently selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsR 17 And R is 18 H, C independently 1-20 Alkyl, carbon chain derived from fatty alcohol or optionally substituted C 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 Alkyl group, and
(c)wherein R is 30 Selected from the group consisting of: substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted (C) 2-10 ) Alkene, substituted or unsubstituted (C 2-10 ) Alkyne, C 1-4 (alkyl) aryl, heteroaryl and C 1-6 A haloalkyl group.
123. A method for treating or preventing an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families comprising administering a therapeutic or preventing amount of a compound of formula (F):
Or a pharmaceutically acceptable salt or prodrug thereof, wherein:
the base is
X 1 、X 1’ 、X 2’ 、X 2 R 2 、R 2’ 、R 3 And R is 3’ As defined in the formula a above,
R 4’ selected from the group consisting of: H. deuterium, CN, substituted or unsubstituted (C 1-8 ) Alkyl, substituted or unsubstituted (C 2-8 ) Alkenyl, substituted or unsubstituted (C 2-8 ) Alkynyl and substituted or unsubstituted (C 1-8 ) A haloalkyl group, a halogen atom,
R 6’ selected from the group consisting of: -OR 6 、-P(O)R 7 R 8 And mono-, di-or tri-phosphates, wherein when chiral at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 6 is H, L-amino acid ester, D-amino acid ester, N-substituted L-amino acid ester, N-substituted D-amino acid ester, N-disubstituted L-amino acid ester, N-disubstituted D-amino acid ester, (acyloxybenzyl) ether, optionally substituted bis-acyloxybenzyl) ester, optionally substituted (acyloxybenzyl) ester, optionally substituted-C (O) -R ', optionally substituted-C (O) O-R', optionally substituted-C (O) SR ', optionally substituted-C (S) SR', PEG ester, PEG carbonate, optionally substituted-CH 2 -O-C (O) -R', optionally substituted-CH 2 -O-C (O) O-R', optionally substituted-CH 2 -CH 2 -S-C (O) -R ', optionally substituted-C (NR ') OR ', optionally substituted-C (NR ') SR ', optionally substituted-C (NR ') N (R ') 2 Optionally substituted-O-C (O) N (R') 2 Lipid esters, lipid carbonates (wherein the lipid is optionally substituted C 12-22 Alkyl, optionally substituted C 12-22 Alkenyl, optionally substituted C 12-22 Alkynyl or optionally substituted C 12-22 Alkoxy), O-P (O) R 8 R 8’ Or mono-, di-or triphosphate, wherein when chiral is present at the phosphorus center, it may be all or part of R p Or S p Or any mixture thereof, or a mixture of any of them,
R 8 and R is 8’ Independently selected from the group consisting of:
(a)OR 15 wherein R is 15 Selected from the group consisting of: H. li, na, K, substituted or unsubstituted C 1-20 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, optionally substituted-C (NR ') OR', optionally substituted-C (NR ') SR', optionally substituted-C (NR ') N (R') 2 Optionally substituted-O-C (O) N (R') 2 、C 1-4 (alkyl) aryl, benzyl, C 1-6 Haloalkyl, C 2-3 (alkyl) OC 1-20 Alkyl, C 2-3 (alkyl) OC 1-20 Alkene, C 2-3 (alkyl) OC 1-20 Alkynes, aryl groups such as phenyl and heteroaryl groups such as pyridyl, wherein the aryl and heteroaryl groups are optionally substituted with zero to three substituents selected from the group consisting of: (CH) 2 ) 0-6 CO 2 R 16 And (CH) 2 ) 0-6 CON(R 16 ) 2
Wherein R is 16 Independently H, substituted or unsubstituted C 1-20 Alkyl, substitutedOr unsubstituted C 1-20 Alkene, substituted or unsubstituted C 1-20 Alkynes, carbon chains derived from fatty alcohols or C substituted by 1-20 Alkyl: c (C) 1-6 Alkyl, C 1-6 Alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl, C 1-5 Alkene, C 1-5 Alkyne, C 3-7 Cycloalkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and
(b) Esters of D-amino acids or L-amino acidsWherein R is 17 And R is 18 H, C independently 1-20 Alkyl, C 1-20 Alkene, C 1-20 Alkynes, carbon chains derived from fatty alcohols or C optionally substituted 1-20 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl, cycloalkyl-C 1-6 Alkyl, cycloheteroalkyl, aryl, heteroaryl, substituted aryl, or substituted heteroaryl; wherein the substituents are C 1-5 Alkyl or C substituted by 1-5 Alkyl: c (C) 1-6 Alkyl, alkoxy, di (C) 1-6 Alkyl) -amino, fluoro, C 3-10 Cycloalkyl or cycloalkyl; and R is 17A Is H or C 1-2 An alkyl group.
124. The process of claim 118 wherein R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
125. The process of claim 119 wherein R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
126. The method as recited in claim 120, wherein R 1 Is H, R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H.
127. The method as recited in claim 121, wherein R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
128. The method as recited in claim 122, wherein R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 ,R 4 Is H, R 5 And R is 5’ Is H or Me.
129. The method as recited in claim 123, wherein R 2 Is H, R 2’ Is OH OR OR 7 ,R 3 Is H, R 3’ Is OH OR OR 7 And R is 4 Is H
130. The process of claim 118 wherein R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
131. The process of claim 119 wherein R 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
132. The method as recited in claim 120, wherein R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
133. The method as recited in claim 121, wherein R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 Alkyl and R 6 Is H, L-amino acid ester, D-amino acid ester or optionally substituted-C (O) -C 1-12 An alkyl group.
134. The process of claim 122 wherein R 2’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
135. The method as recited in claim 123, wherein R 2’ And R is 3’ Is OH, L-amino acid ester, D-amino acid ester or optionally substituted-O-C (O) -C 1-12 An alkyl group.
136. The method of any one of claims 118-135, wherein the compound is capable of being present in the β -D or β -L configuration.
137. The method of any one of claims 118-135, wherein the virus is a coronavirus.
138. The method of claim 137, wherein the coronavirus is human coronavirus 229E, SARS, MERS, SARS-CoV-1, OC43, or SARS-CoV-2.
139. The method of claim 137, wherein the coronavirus is SARS-CoV-2.
140. The method of any one of claims 118-135, wherein the compound is co-administered with one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
141. The method of claim 140 wherein the compound is administered with adefovir, N-hydroxycytidine, mo Nuola, PF-07321332, PF-07304814, or a pharmaceutically acceptable salt or prodrug thereof.
142. The method of claim 140, wherein the additional active compound is a JAK inhibitor and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib, or baratinib, or a pharmaceutically acceptable salt or prodrug thereof.
143. The method of claim 140, wherein the one or more additional active agents comprise an anticoagulant or a platelet aggregation inhibitor.
144. The method of claim 140, wherein the one or more additional active agents comprise an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
145. The use of a compound of any one of claims 118-135 for the manufacture of a medicament for the treatment or prevention of an infection of the coronaviridae, flaviviridae, picornaviridae, bunyaviridae or togaviridae families.
146. The use of claim 145, wherein the infection is a coronaviridae infection.
147. The use of claim 146, wherein the coronavirus is human coronavirus 229E, SARS, MERS, SARS-CoV-1, OC43, or SARS-CoV-2.
148. The use of claim 146, wherein the coronavirus is SARS-CoV2.
149. The use of claim 145, wherein the medicament further comprises one or more additional active compounds selected from the group consisting of: fusion inhibitors, entry inhibitors, protease inhibitors, polymerase inhibitors, antiviral nucleosides, viral entry inhibitors, viral maturation inhibitors, JAK inhibitors, angiotensin converting enzyme 2 (ACE 2) inhibitors, SARS-CoV specific human monoclonal antibodies including CR3022, agents of different or unknown mechanisms.
150. The use of claim 145, wherein the medicament further comprises adefovir, N-hydroxycytosine, mo Nuola, PF-07321332, PF-07304814, or a pharmaceutically acceptable salt or prodrug thereof.
151. The use of claim 145, wherein the medicament further comprises a JAK inhibitor, and the JAK inhibitor is strapdown scrupulously and respectfully, tofacitinib, or baratinib, or a pharmaceutically acceptable salt or prodrug thereof.
152. The use of claim 145, wherein the medicament further comprises an anticoagulant or a platelet aggregation inhibitor.
153. The use of claim 145, wherein the medicament further comprises an ACE-2 inhibitor, a CYP-450 inhibitor, or a NOX inhibitor.
154. The use of claim 145, wherein the medicament is a transdermal composition or a nanoparticle composition.
155. The method of any one of claims 1-65, 76-107, or 118-144, wherein the compound is administered in combination with an NS5A inhibitor.
156. The method of claim 155, wherein the NS5A inhibitor is dacatavir.
157. The use of any one of claims 66-75, 108-117, or 145-154, wherein the medicament further comprises an NS5A inhibitor.
158. The use of claim 157, wherein the NS5A inhibitor is dacatavir.
CN202280041407.1A 2021-04-09 2022-04-11 Thionucleosides as antiviral agents Pending CN117881403A (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US63/173,354 2021-04-09
US63/175,673 2021-04-16
US63/210,246 2021-06-14
US63/288,163 2021-12-10
US202263298836P 2022-01-12 2022-01-12
US63/298,836 2022-01-12
PCT/US2022/024290 WO2022217155A2 (en) 2021-04-09 2022-04-11 Thionucleosides as antiviral agents

Publications (1)

Publication Number Publication Date
CN117881403A true CN117881403A (en) 2024-04-12

Family

ID=90314874

Family Applications (3)

Application Number Title Priority Date Filing Date
CN202280041644.8A Pending CN117769427A (en) 2021-04-09 2022-04-11 Modified nucleosides and nucleotide analogues as antiviral agents for coronaviruses and other viruses
CN202280041352.4A Pending CN117858711A (en) 2021-04-09 2022-04-11 Nucleoside and nucleotide analogs as antiviral agents
CN202280041407.1A Pending CN117881403A (en) 2021-04-09 2022-04-11 Thionucleosides as antiviral agents

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN202280041644.8A Pending CN117769427A (en) 2021-04-09 2022-04-11 Modified nucleosides and nucleotide analogues as antiviral agents for coronaviruses and other viruses
CN202280041352.4A Pending CN117858711A (en) 2021-04-09 2022-04-11 Nucleoside and nucleotide analogs as antiviral agents

Country Status (1)

Country Link
CN (3) CN117769427A (en)

Also Published As

Publication number Publication date
CN117858711A (en) 2024-04-09
CN117769427A (en) 2024-03-26

Similar Documents

Publication Publication Date Title
US20240216413A1 (en) Thionucleosides as antiviral agents
US11207370B2 (en) Peptidomimetics for the treatment of coronavirus and picornavirus infections
US20240216414A1 (en) Antiviral Agents and Nucleoside Analogs for Treatment of Zika Virus
WO2019133712A1 (en) Combined modalities for nucleosides and/or nadph oxidase (nox) inhibitors as myeloid-specific antiviral agents
CN107709344B (en) Nucleoside analogues for the treatment of flaviviridae and cancer
US20240262857A1 (en) Novel universal anti-rna virus agents
CA2946867C (en) 2&#39;-disubstituted nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
US20170334941A1 (en) 2&#39;,2&#39;-dihalo nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
WO2007097991A2 (en) Methods and kits for dosing of antiviral agents
JP2022106920A (en) Alkyne-containing nucleotide and nucleoside therapeutic compositions and uses related thereto
JP2024111292A (en) Phospholipid compounds and uses thereof
CN117881403A (en) Thionucleosides as antiviral agents

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination