CN110317211B - Substituted polycyclic pyridone compound and prodrug thereof - Google Patents

Substituted polycyclic pyridone compound and prodrug thereof Download PDF

Info

Publication number
CN110317211B
CN110317211B CN201910675084.1A CN201910675084A CN110317211B CN 110317211 B CN110317211 B CN 110317211B CN 201910675084 A CN201910675084 A CN 201910675084A CN 110317211 B CN110317211 B CN 110317211B
Authority
CN
China
Prior art keywords
compound
deuterium
hydrogen
compounds
influenza
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.)
Active
Application number
CN201910675084.1A
Other languages
Chinese (zh)
Other versions
CN110317211A (en
Inventor
王义汉
刘志强
李焕银
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.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN110317211A publication Critical patent/CN110317211A/en
Application granted granted Critical
Publication of CN110317211B publication Critical patent/CN110317211B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Landscapes

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

Abstract

The invention provides a deuterium-substituted polycyclic pyridone compound, a prodrug thereof, a composition containing the compound and application of the composition. The compounds and compositions of the present invention exhibit cap-dependent endonuclease inhibitory activity and have superior pharmacokinetic properties.

Description

Substituted polycyclic pyridone compound and prodrug thereof
Technical Field
The invention relates to a substituted polycyclic pyridone compound, a prodrug thereof, a composition containing the compound and application of the compound. More particularly, the present invention relates to certain deuterium substituted (((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl) -6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) triazin-7-yl) oxy) methyl carbonates and their parent drugs, these deuterium substituted compounds and their compositions exhibit Cap-dependent endonuclease (Cap-dependent endonulase) inhibitory activity, and these deuterium substituted compounds have superior pharmacokinetic properties.
Background
Influenza, referred to as influenza (influenza or flu), is an acute respiratory infectious disease caused by infection with influenza virus and has become the most lethal viral infectious disease. Elderly people over 65 years old, children under 2 years old, and people with chronic respiratory and cardiovascular diseases are more susceptible to influenza complications and are therefore high risk groups for influenza. In the 20 th century, there were three large-scale influenza outbreaks, spanish influenza in 1918, asian influenza in 1957 and hong kong influenza in china in 1968. Of these, Spanish influenza (H1N1) is devastating, with deaths exceeding the first world war infecting 30% of the world population, resulting in at least 2000 million deaths. The other two influenza outbreaks were caused by influenza viruses type H2N2 and H3N2, respectively, which, although smaller in scale, also resulted in hundreds of deaths. A novel influenza A H1N1 virus appearing in 2009 causes the first influenza pandemic in the 21 st century, and researches show that the gene of the virus contains gene sequences of various influenza viruses such as human, pig, poultry and the like, and the virus is a recombinant virus. In a short period of months, the virus spreads rapidly throughout the world, causing great panic. The world health organization raises the warning from three levels for many times until the warning is the highest six levels.
Influenza viruses are classified into three types, a, b, and c (also referred to as A, B, C) according to the antigenic properties of nucleoprotein and matrix protein. The influenza A virus has the widest host range, can infect people, pigs, horses and poultry, has strong pathogenicity, can cause pandemics of influenza in the world and has the greatest harm to human beings; the influenza B virus mainly infects human and pigs, has low pathogenic capability and can cause local outbreak of influenza; influenza c only infects infants and people with low immunity, rarely causes epidemic and is relatively less harmful.
As anti-influenza drugs, there are known mainly: (1) amantadine (Amantadine) and Rimantadine (Rimantadine) are two anti-influenza virus drugs that are on the market earlier, and play an antiviral role mainly by blocking the M2 ion channel of viruses and inhibiting virus uncoating. (2) Oseltamivir Phosphate (Oseltamivir Phosphate, trade name: Tamiflu, developed by Roche) and Zanamivir (Zanamivir, trade name: Allevir (Relenza), developed by Kurarin Schke) are neuraminidase inhibitors, have high curative effect and good safety and tolerance, and are the first choice drugs for resisting influenza at present. However, there are problems of occurrence and side effects of drug-resistant strains, and concerns about a world pandemic of a novel influenza virus having high pathogenicity or lethality, and therefore development of an anti-influenza drug having a new mechanism is desired.
Regarding the cap-dependent endonuclease as an enzyme derived from influenza virus, it is considered that the cap-dependent endonuclease is suitable for a target of an anti-influenza drug because it is essential for virus propagation and has a virus-specific enzyme activity that a host does not have. The cap-dependent endonuclease of influenza virus is an endonuclease activity which generates a fragment containing 9 to 13 bases of a cap structure (bases of the cap structure are not included in the number of bases) using a host mRNA precursor as a substrate. This fragment functions as a primer for viral RNA polymerase and is used for the synthesis of mRNA encoding viral proteins. That is, it is considered that the substance which inhibits the cap-dependent endonuclease inhibits synthesis of viral protein by inhibiting synthesis of viral mRNA, and as a result, viral growth is inhibited.
Baloxavir Marboxil (also known as S-033188, chemical name (((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl) -6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) triazin-7-yl) oxy) methyl carbonate, having the following structure) is developed by japan salt seine pharmaceutical co, Ltd (shinogl & co., Ltd), a first-initiated (first-in-class), single dose experimental oral drug, aims at inhibiting cap-dependent endonuclease in influenza virus and plays a role in inhibiting virus replication. In 2016, month 2, salt fiend assigned the right to develop in japan and areas outside taiwan of china to roche. Baloxavir Marboxil has been approved by accelerated approval in japan in 2018 at 23.2 and successfully marketed in japan for the treatment of influenza a and b in adult and pediatric patients under the trade name Xofluza. In 6 months 2018, roche announced that the FDA in the united states had accepted a new drug application by Baloxavir Marboxil and granted priority vetting, and it is expected that a vetting decision will be made 24 days 12 months 2018. Clinical trials have shown that Baloxavir Marboxil, given only as a single dose (40 or 80mg each, once a day, 1 day), reduces the duration of influenza symptoms and significantly reduces viral shedding within a day, has a greater antiviral efficacy than oseltamivir (75 mg each, twice a day, 5 days), and is highly effective in reducing viral titers in more than 50% of patients (including children) to below the detection limit on the first day after treatment, in addition to being highly effective against influenza a (H5N1 or H7N 9).
Figure GDA0003592353000000021
Poor absorption, distribution, metabolism and/or excretion (ADME) properties are known to be the major cause of failure in many drug candidate clinical trials. Many drugs currently on the market also have limited their range of application due to poor ADME properties. The rapid metabolism of drugs can result in the difficulty of obtaining many drugs that are otherwise effective in treating disease due to their rapid metabolic clearance from the body. Although frequent or high dose administration may solve the problem of rapid clearance of the drug, this method may cause problems such as poor patient compliance, side effects caused by high dose administration, and increased treatment costs. In addition, rapidly metabolizing drugs may also expose patients to undesirable toxic or reactive metabolites.
Although Baloxavir Marboxil is effective in treating influenza, there is still a serious clinical unmet need in the art and finding new compounds with good oral bioavailability and druggability is a challenging task. Accordingly, there remains a need in the art to develop compounds that inhibit cap-dependent endonuclease activity and/or that are better pharmacokinetic, and the present invention provides such compounds.
Disclosure of Invention
In view of the above technical problems, the present invention discloses a novel deuterium-substituted polycyclic pyridone derivative, a prodrug thereof, a composition comprising the same, and uses thereof, which have better cap-dependent endonuclease activity inhibition, particularly better pharmacokinetic properties and/or oral bioavailability.
In contrast, the invention adopts the following technical scheme:
in a first aspect of the invention, there is provided a compound of formula (I):
Figure GDA0003592353000000031
wherein the content of the first and second substances,
p is selected from H or-C(R11R12)OC(O)OC(R13R14R15);
Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8Each independently selected from hydrogen or deuterium;
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
In another aspect, the present invention provides pharmaceutical compositions comprising a compound of the present invention, or a tautomer, stereoisomer, prodrug, pharmaceutically acceptable salt, crystal form, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient. In a specific embodiment, a compound of the present invention, or a tautomer, stereoisomer, prodrug, pharmaceutically acceptable salt, crystalline form, hydrate, or solvate thereof, is provided in an effective amount in the pharmaceutical composition. In particular embodiments, a compound of the present invention, or a tautomer, stereoisomer, prodrug, pharmaceutically acceptable salt, crystalline form, hydrate, or solvate thereof, is provided in a therapeutically effective amount. In particular embodiments, a compound of the present invention, or a tautomer, stereoisomer, prodrug, pharmaceutically acceptable salt, crystalline form, hydrate, or solvate thereof, is provided in a prophylactically effective amount.
In another aspect, the present invention provides a method for preparing a pharmaceutical composition as described above, or a tautomer, stereoisomer, prodrug, pharmaceutically acceptable salt, crystal form, hydrate, or solvate thereof, comprising the steps of: a pharmaceutically acceptable excipient is mixed with a compound of the present invention, or a pharmaceutically acceptable salt, crystal form, hydrate, or solvate thereof, to form a pharmaceutical composition.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, in combination with a neuraminidase inhibitor, an RNA-dependent RNA polymerase inhibitor, an M2 protein inhibitor, a PB2Cap binding inhibitor, an anti-HA antibody, or an immunoactive agent.
In another aspect, the present invention provides a use of a compound of the present invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, for the preparation of a medicament for the treatment and prevention of diseases caused by a virus having a cap-dependent endonuclease.
In another aspect, the present invention also relates to a method for treating and/or preventing a disease caused by a virus having a cap-dependent endonuclease in a subject, which comprises administering to the subject a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt, crystal form, hydrate or solvate compound thereof or a pharmaceutical composition thereof. In particular embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In particular embodiments, the compound is administered chronically.
The present invention further provides a therapeutic method or prophylactic method for influenza infection using the prodrug compound, and the above compound having an anti-influenza effect. The present invention further provides a parent compound of a prodrug compound. The parent compound is useful as an anti-influenza agent, or an intermediate of the prodrug compound.
The compounds of the present invention have activity to inhibit cap-dependent endonucleases. More preferred compounds are prodrugs, which become parent compounds having cap-dependent endonuclease inhibitory activity in vivo after administration, and thus are useful as therapeutic and/or prophylactic agents for influenza infection.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Definition of
Herein, "deuterated", unless otherwise specified, means that one or more hydrogens of a compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
Herein, unless otherwise specified, "non-deuterated compound" means a compound containing deuterium at an atomic ratio of deuterium not higher than the natural deuterium isotope content (0.015%).
The term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977)66: 1-19. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases.
The compounds of the present invention may be in amorphous or crystalline form. Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "crystalline form" refers to the different arrangements of chemical drug molecules, typically expressed as the presence of the drug substance in the solid state. One drug can exist in a plurality of crystal form substances, and different crystal forms of the same drug can be dissolved and absorbed in vivo differently, so that the dissolution and release of the preparation can be influenced.
The term "crystalline form" refers to the different arrangements of chemical drug molecules, typically expressed as the presence of the drug substance in the solid state. One drug can exist in a plurality of crystal form substances, and different crystal forms of the same drug can be dissolved and absorbed in vivo differently, so that the dissolution and release of the preparation can be influenced.
As used herein, the term "subject" includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In other embodiments, the subject is a non-human animal.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes the effect that occurs when a subject has a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on the following factors: for example, biological goals, pharmacokinetics of the compound, the disease being treated, mode of administration, and the age, health, and condition of the subject. An effective amount includes both therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can include an amount that improves the overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
As used herein, unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in preventing a disease, disorder, or condition. The term "prophylactically effective amount" can include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic efficacy of other prophylactic agents.
"combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously with another therapeutic agent in a single unit dosage form.
Detailed Description
Compound (I)
The term "prodrug" as used herein refers to a compound represented by the formula (II) or a pharmaceutically acceptable salt thereof in the following reaction formula, and means a compound which is converted into a compound represented by the formula (III) by a decomposition reaction caused by a drug-metabolizing enzyme, a hydrolase, gastric acid, intestinal bacteria, or the like under physiological conditions in vivo, and exhibits a cap-dependent endonuclease (CEN) inhibitory activity and/or a CPE inhibitory effect.
Figure GDA0003592353000000071
(wherein each symbol is as defined above)
More preferably, the prodrug of formula (II) is represented by: bioavailability and/or AUC (area enclosed by plasma concentration curve versus time axis) and/or C when administered in vivomax(maximum plasma concentration occurring after administration) is improved as compared with the compound represented by the formula (III).
Therefore, the prodrug is efficiently absorbed into the body in the stomach and/or intestine and the like after administration to a living body (for example, oral administration), and thereafter is converted into the compound represented by the formula (III), and thus can preferably exhibit a higher therapeutic and/or prophylactic effect than the compound represented by the formula (III).
The present invention provides compounds of formula (I), or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates, or solvate compounds thereof:
Figure GDA0003592353000000072
wherein the content of the first and second substances,
p is selected from H or-C (R)11R12)OC(O)OC(R13R14R15);
Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8Each independently selected from hydrogen or deuterium;
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that the above-mentioned compounds contain at least one deuterium atom.
As a preferred embodiment of the present invention, the deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
Specifically, Y in the present invention1、Y2、Y3、Y4、Y5、Y6、Y7、Y8、R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14And R15The deuterium isotope content in each deuterated position is at least 5%, more preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 15%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
As a preferred embodiment of the present invention, the compound of formula (I) contains at least one deuterium atom, more preferably contains two deuterium atoms, more preferably contains three deuterium atoms, more preferably contains four deuterium atoms, more preferably contains five deuterium atoms, more preferably contains six deuterium atoms, more preferably contains seven deuterium atoms, more preferably contains eight deuterium atoms, more preferably contains nine deuterium atoms, more preferably contains ten deuterium atoms, more preferably contains eleven deuterium atoms, more preferably contains twelve deuterium atoms, more preferably contains thirteen deuterium atoms, more preferably contains fourteen deuterium atoms, more preferably contains fifteen deuterium atoms, more preferably contains sixteen deuterium atoms, more preferably contains seventeen deuterium atoms, more preferably contains eighteen deuterium atoms, more preferably contains nineteen deuterium atoms, more preferably contains twenty-one deuterium atoms, more preferably twenty two deuterium atoms, and even more preferably twenty three deuterium atoms.
In a particular embodiment, "Y1、Y2、Y3、Y4、Y5、Y6、Y7And Y8Each independently selected from hydrogen or deuterium "including Y1Selected from hydrogen or deuterium, Y2Selected from hydrogen or deuterium, Y3Selected from hydrogen or deuterium, and so on, up to Y8Selected from hydrogen or deuterium. More specifically, including Y1Is hydrogen or Y1Being deuterium, Y2Is hydrogen or Y2Is deuterium, Y3Is hydrogen or Y3Deuterium, and so on, until Y8Is hydrogen or Y8Is a technical scheme of deuterium.
In another embodiment, "R" is1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14And R15Each independently selected from hydrogen or deuterium "comprising R1Selected from hydrogen or deuterium, R2Selected from hydrogen or deuterium, R3Selected from hydrogen or deuterium, and so on, up to R15Selected from hydrogen or deuterium. More specifically, includes R1Is hydrogen or R1Is deuterium, R2Is hydrogen or R2Is deuterium, R3Is hydrogen or R3Deuterium, and so on, up to R15Is hydrogen or R15Is a technical scheme of deuterium.
In a preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, and R1-R15With the proviso that said compound contains at least one deuterium atom, as defined above.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, and R1-R15As defined above, with the proviso that R7-R15At least one of them is deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof,wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1And R2Is hydrogen, and R3-R15Each independently selected from hydrogen or deuterium, with the proviso that the compound contains at least one deuterium atom.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1And R2Is hydrogen, and R3-R15Each independently selected from hydrogen or deuterium, with the proviso that R7-R15At least one of them is deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, and R3、R7-R15Each independently selected from hydrogen or deuterium, with the proviso that the compound contains at least one deuterium atom.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, and R3、R7-R15Each independently selected from hydrogen or deuterium, with the proviso that R7-R15At least one of them is deuterium.
In another more preferred embodiment, R7-R10Are the same.
In another more preferred embodiment, R7-R10Is deuterium.
In another more preferred embodiment, R7-R10Is hydrogen.
In another more preferred embodiment, R11-R12Are the same.
In another more preferred embodiment, R11-R12Is deuterium.
In another more preferred embodiment, R11-R12Is hydrogen.
In another more preferred embodiment, R13-R15Are the same.
In another more preferred embodiment, R13-R15Is deuterium.
In another more preferred embodiment, R13-R15Is hydrogen.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3Is deuterium, and R7-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3Is deuterium, and R7-R15Are independently selected fromFrom hydrogen or deuterium with the proviso that R7-R15At least one of them is deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3Is deuterium, and R7-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3Is deuterium, and R7-R15Each independently selected from hydrogen or deuterium, with the proviso that R7-R15At least one of them is deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R7-R10Is deuterium, and R3、R11-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R7-R10Is deuterium, and R3、R11-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3And R7-R10Is deuterium, and R11-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3And R7-R10Is deuterium, and R11-R15Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R13-R15Is deuterium, and R3、R7-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a process for the preparation of formula (I)A compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3And R13-R15Is deuterium, and R7-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R13-R15Is deuterium, and R3、R7-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3And R13-R15Is deuterium, and R7-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R7-R10And R13-R15Is deuterium, and R3And R11-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R7-R10And R13-R15Is deuterium, and R3And R11-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from H or-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3、R7-R10And R13-R15Is deuterium, and R11-R12Each independently selected from hydrogen or deuterium.
In another preferred embodiment, the present invention relates to a compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein P is selected from-C (R)11R12)OC(O)OC(R13R14R15),Y1-Y8Is hydrogen, R1-R2And R4-R6Is hydrogen, R3、R7-R10And R13-R15Is deuterium, and R11-R12Each independently selected from hydrogen or deuterium.
As a preferred embodiment of the present invention, the compound is of any one of the following structures, or a pharmaceutically acceptable salt thereof, but is not limited to the following structures:
Figure GDA0003592353000000121
Figure GDA0003592353000000131
the compounds of the invention may include one or more asymmetric centers and may therefore exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
One skilled in the art will appreciate that the organic compound may form a complex with the solvent in which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is combined with a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes solvates in solution and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates.
The term "hydrate" refers to a compound that is associated with an aqueous phase. In general, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, hydrates of the compounds can be used, for example, of the formula R. x H2O represents, wherein R is the compound, and x is a number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrates (x is a number greater than 0 and less than 1), e.g., hemihydrate (R0.5H)2O)) and polyhydrates (x is a number greater than 1, e.g. dihydrate (R.2H)2O) and hexahydrate (R.6H)2O))。
The compounds of the invention may be in amorphous or crystalline form (polymorphs). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may cause a crystalline form to dominate. Various polymorphs of a compound may be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those recited in formula (I), but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and36and (4) Cl. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the invention, e.g. by incorporation of radioactive isotopes (e.g. by introducing3H and14C) can be used in drug and/or substrate tissue distribution assays. Tritium, i.e.3H and carbon-14, i.e.14The C isotopes are particularly preferred because of their ease of preparation and detection. Further, by heavier isotopes, e.g. deuterium, i.e.2H, may be preferred in some cases because of the higher metabolic stability that may provide therapeutic benefits, such as increased in vivo half-life or reduced dosage requirements. Isotopically-labelled compounds of formula (I) of the present invention and prodrugs thereof can generally be prepared by substituting a readily available isotopically-labelled reagent for a non-isotopically-labelled reagent in the course of performing the procedures disclosed in the schemes and/or in the examples and preparations below.
Pharmaceutical compositions, formulations and kits
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. Pharmaceutically acceptable carriers, adjuvants, or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
The invention also includes kits (e.g., pharmaceutical packages). The provided kits can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, provided kits may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to rapidly increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient, e.g., an intramuscular or subcutaneous bolus dose results in a slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, allowing the concentration of the active ingredient in the blood to rise rapidly to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the body of the subject. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More generally, however, the compositions are provided in unit dosage form for convenient administration of the precise dosage. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable for the purpose of producing the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired form of administration.
For oral dosages, a typical regimen is one oral dose per day. Using these dosing modes, each dose provides about 0.01 to about 50mg/kg of a compound of the invention, with preferred doses each providing about 10 to about 40mg/kg, especially about 10 to about 30 mg/kg.
In order to provide a blood level similar to, or lower than, the use of the injected dose, a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring and flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with a cream base, for example of the oil-in-water type. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and compositions are included within the scope of the present invention.
The compounds of the present invention may also be administered by transdermal means. Thus, transdermal administration can be achieved using a reservoir (reservoir) or porous membrane type, or a patch of various solid matrices.
The above components of the compositions for oral, injectable or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17th edition,1985, Mack Publishing Company, Easton, Pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1, 4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitution. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Indications
As used herein, the term "compounds of the present invention" refers to compounds of formula (I). The term also includes tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate compounds of the compounds of formula (I).
The compounds of the present invention are anti-influenza virus drugs having inhibitory activity against dependent endonucleases and therefore can be used for the treatment and/or prevention of epidemic infectious diseases.
Influenza viruses are negative-sense single-stranded RNA viruses and are a member of the orthomyxoviridae family. There are currently 3 influenza viruses: influenza a, influenza B and influenza C. Influenza a virus has a host cell-derived lipid membrane that contains hemagglutinin, neuraminidase, and M2 proteins protruding from the surface of the virus. Influenza a viruses have been further classified according to hemagglutinin (H or HA) and neuraminidase (N). There are approximately 16H antigens (H1 to H16) and 9N antigens (N1 to N9). Influenza a viruses include several subtypes, including H1N1, H1N2, H2N2, H3N1, H3N2, H3N8, H5N1, H5N2, H5N3, H5N8, H5N9, H7N1, H7N2, H7N3, H7N7, H7N9, H9N2, and H10N 7. Influenza polymerase is a heterotrimer composed of 3 subunits: polymerase Acid (PA), polymerase base 1(PB1), and polymerase base 2(PB 2). In the nucleus of infected cells, this polymerase is responsible for the replication and transcription of viral RNA. The PA subunit comprises an endonuclease active site. The endonuclease activity of PA cleaves cellular mRNA, which is then used as a primer by the PB1 subunit for viral mRNA synthesis.
In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to treat and/or ameliorate influenza virus infection. In other embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to prevent influenza virus infection.
In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to inhibit replication of influenza virus. In other embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to inhibit an influenza polymerase complex. In other embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to inhibit and/or reduce the activity of cap-dependent endonuclease. In other embodiments, an effective amount of a compound of the invention or a pharmaceutical composition described herein can be used to inhibit and/or reduce the ability of an endonuclease to cleave mRNA.
In some embodiments, the influenza virus infection may be an influenza a virus infection. In other embodiments, the influenza virus infection may be an influenza B virus infection. In other embodiments, the influenza virus infection may be an influenza C virus infection. In some embodiments, the compounds of the present invention are useful for treating and/or ameliorating one or more subtypes of influenza. For example, the compounds of the invention may be used to treat H1N1 and/or H3N 2. Additionally or alternatively, the compounds of the invention may be used to treat H2N2, H5N1, and/or H7N 9. In some embodiments, the compounds of the present invention are effective against more than 1 subtype of influenza. For example, the compounds of the present invention are effective against 2, 3,4 and/or 5 or more subtypes of influenza.
In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition comprising a compound of the invention may be used to treat and/or ameliorate upper respiratory tract infections due to (direct and/or indirect) influenza virus infection. In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition comprising a compound of the invention may be used to treat and/or ameliorate a lower respiratory viral infection due to (direct and/or indirect) influenza viral infection. In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition comprising a compound of the invention can be used to treat and/or alleviate one or more symptoms of influenza virus infection (e.g., cough, sore throat, headache, nasal congestion, fever or chills, muscle or joint pain, fatigue, etc.). In some embodiments, an effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention may be used to treat and/or ameliorate bronchiolitis and/or tracheobronchitis due to influenza virus infection. In some embodiments, an effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention may be used to treat and/or ameliorate pneumonia due to influenza virus infection.
In some embodiments, an effective amount of a compound of the invention or a pharmaceutical composition comprising a compound of the invention can be used to reduce the severity of one or more symptoms of influenza infection. Examples of symptoms include, but are not limited to, fever, chills, cough, sore throat, runny nose, nasal congestion, muscle soreness, body pain, headache, fatigue, vomiting, and/or diarrhea.
In some embodiments, an effective amount of a compound of the invention is an amount effective to reduce viral titer to a lower level, for example, from about 10E4TCID50/mL (TCID ═ tissue culture infectious dose) to about 10E3TCID50/mL, or to about 100TCID50/mL, or to about 10TCID 50/mL. In some embodiments, an effective amount of a compound of the invention is: the amount of viral load is effectively reduced compared to the viral load prior to administration of the compound of formula (I). For example, wherein the viral load is measured prior to administration of the compound of formula (I) and again after the start of a treatment regimen using the compound of formula (I) (e.g., 1-2 days after the start of treatment). In some embodiments, an effective amount of a compound of the invention may be an amount effective to reduce viral load to less than about 10E4TCID 50/mL. In some embodiments, an effective amount of a compound of the invention is: an amount effective to achieve a reduction in viral titer in a nasal/pharyngeal or nasal cavity wash sample of an individual as compared to the viral load prior to administration of the compound of formula (I) in the range: about 1.5-log to about 2.5-log reduction, or about 3-log to about 4-log reduction. For example, wherein the viral load is measured prior to administration of the compound of formula (I), and prior to use of the compound of formula (I), and again after initiation of a treatment regimen using the compound of formula (I) (e.g., 1-2 days after initiation of treatment).
In some embodiments, the compounds of the invention can result in one or more overall quality of life health compared to an untreated individual, such as a greatly reduced duration of disease, reduced disease severity, reduced time to return to normal health and normal activity, and reduced time to alleviate one or more symptoms of a viral infection compared to an untreated individual. In some embodiments, the compounds of the invention may cause a reduction in the length and/or severity of one or more symptoms associated with a viral infection as compared to an untreated individual. In some embodiments, the compounds of the invention may cause a reduction in one or more secondary complications associated with viral infection, including but not limited to otitis media (ear inflammation) sinusitis, bronchitis, and pneumonia, as compared to untreated individuals.
In some embodiments, a compound of the invention can cause a reduction in viral replication by at least 1,2, 3,4, 5, 10, 15, 20, 25, 75, 100-fold or more relative to the pre-treatment level of the individual, as determined after initiation of a treatment regimen.
After a period of time, the infectious agent may develop resistance to one or more therapeutic agents. The term "drug resistance" as used herein refers to a strain of virus that exhibits a delayed, reduced, and/or zero response to one or more therapeutic agents. For example, following treatment with an antiviral agent, the viral load of an individual infected with a drug-resistant virus may be reduced to a lesser extent compared to the amount of reduction in viral load exhibited by an individual infected with a non-drug resistant strain. In some embodiments, a compound of the invention may be administered to a subject infected with an influenza virus resistant to one or more different anti-influenza agents (e.g., amantadine and/or oseltamivir). In some embodiments, a compound of the invention may be administered to an individual infected with an influenza virus that is resistant to an inhibitor of M2 protein. In some embodiments, the development of drug resistant influenza strains is delayed compared to the development of influenza strains against other influenza drugs, but when individuals are treated with the compounds of the invention.
In some embodiments, the compounds of the invention can reduce the percentage of individuals experiencing complications from influenza virus infection compared to the percentage of individuals experiencing complications treated with oseltamivir. For example, the percentage of complications experienced by subjects treated with a compound of formula (I) was 10%, 20%, 30%, 40%, 60%, 70%, 80% and 90% less subjects treated with oseltamivir.
In some embodiments, a compound of the invention can reduce the percentage of individuals experiencing complications from influenza virus infection compared to the percentage of individuals experiencing complications treated with Baloxavir marboxil. For example, the percentage of individuals treated with a compound of formula (I) experiencing complications is 10%, 20%, 30%, 40%, 60%, 70%, 80% and 90% less than individuals treated with Baloxavir marboxil.
Combination therapy
In some embodiments, a compound of the present invention, or a pharmaceutical composition comprising a compound described herein, may be used in combination with one or more additional agents. In some embodiments, the compounds of the present invention may be used in combination with one or more agents currently used in the standard of care for the treatment of influenza. For example, the additional agent may be amantadine (adamantan-1-amine, Symmetrel), rimantadine (flunadine), zanamivir (renenza), and oseltamivir (Tamiflu). For the treatment of influenza, additional agents include, but are not limited to: neuraminidase inhibitors, M2 protein inhibitors, polymerase inhibitors, PB2 inhibitors, peramivir, laninamivir, fapiravir, laninamivir octanoate, influenza enzymes (DAS181, NexBio), ADS-8902 (amantadine HCl/oseltamivir/ribavirin, Adamas Pharmaceuticals), immunomodulators (e.g., type I interferons), beraprost, ribavirin, and the like.
In some embodiments, a compound of the present invention may be administered with one or more additional agents in a single pharmaceutical composition. In some embodiments, the compounds of the present invention may be administered with one or more additional agents as two or more separate pharmaceutical compositions. For example, a compound of the invention can be administered in one pharmaceutical composition and at least one additional agent can be administered in a second pharmaceutical composition. If there are at least two additional pharmaceutical agents, one or more of the additional pharmaceutical agents may be present in a first pharmaceutical composition comprising a compound of the present invention, and at least one of the other additional pharmaceutical agents may be present in a second pharmaceutical composition. The order of administration of the compounds of the present invention and one or more additional agents may vary. In some embodiments, a compound of the invention may be administered prior to all additional agents. In other embodiments, a compound of the invention may be administered prior to at least one additional agent. In still other embodiments, the compounds of the present invention may be administered simultaneously with one or more additional agents. In still other embodiments, the compounds of the present invention may be administered after the administration of at least one additional agent. In some embodiments, a compound of the invention may be administered after all additional agents are administered.
In some embodiments, the combination of a compound of the invention in combination with one or more additional agents may result in an additive effect. In some embodiments, the combination of a compound of the invention in combination with one or more additional agents may result in a synergistic effect. In some embodiments, the combination of a compound of the invention in combination with one or more additional agents may result in a strong synergistic effect. In some embodiments, the combination of a compound of the invention in combination with one or more additional agents is not antagonistic.
As used herein, the term "antagonistic" means: the activity of a combination of compounds is lower when the activity of each compound is determined separately (i.e. as a single compound) compared to the sum of the activities of the compounds in the combination. As used herein, the term "synergistic effect" means: when the activity of each compound is determined individually, the activity of the combination of compounds is greater than the sum of the individual activities of the compounds in the combination. As used herein, the term "additive effect" means: when the activity of each compound is measured individually, the activity of the combination of compounds is approximately equal to the sum of the individual activities of the compounds in the combination.
Potential advantages of using a compound of the invention in combination with one or more additional agents as described above (including pharmaceutically acceptable salts and prodrugs thereof) may be: the amount of one or more additional agents (including pharmaceutically acceptable salts and prodrugs thereof) required to effectively treat a disease state disclosed herein (e.g., influenza) is reduced compared to the amount required to achieve the same therapeutic result when the one or more additional agents (including pharmaceutically acceptable salts and prodrugs thereof) are administered in the absence of the compound of formula (I) or a pharmaceutically acceptable salt thereof. For example, the amount of additional agents (including pharmaceutically acceptable salts and prodrugs thereof) as described above when administered in combination with a compound of the invention may be less than the amount of additional agents (including pharmaceutically acceptable salts and prodrugs thereof) required to achieve the same viral load reduction when administered in monotherapy. Another potential advantage of utilizing the compounds of the present invention in combination with one or more additional agents (including pharmaceutically acceptable salts and prodrugs thereof) as described above is: the use of two or more compounds with different mechanisms of action enables the establishment of a higher barrier to the formation of drug resistant viral strains than when the compounds are administered as monotherapy.
Process for preparing the Compounds of the invention
The compounds of the invention (including salts thereof) may be prepared using known organic synthesis techniques and may be synthesized according to any of a number of possible synthetic routes, such as those in the schemes below. The reaction for preparing the compounds of the present invention may be carried out in a suitable solvent, which can be easily selected by one skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperatures at which the reaction is carried out (e.g., temperatures in the range of solvent freezing temperatures to solvent boiling temperatures). A given reaction may be carried out in one solvent or a mixture of more than one solvent. The skilled person can select the solvent for a particular reaction step depending on the particular reaction step.
The preparation of the compounds of the invention may involve the protection and deprotection of different chemical groups. The skilled person can easily decide whether protection and deprotection is required and the choice of appropriate protecting groups. The chemistry of protecting Groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4 th edition, John Wiley & Sons: new Jersey, (2006), which is incorporated herein by reference in its entirety.
The compound of the present invention can be prepared by reacting a racemic mixture of the compounds with an optically active resolving agent to form a pair of diastereomeric compounds, separating the diastereomers and recovering the optically pure enantiomers thereof, into their individual stereoisomers. Enantiomeric resolution can be carried out using diastereomeric derivatives of the compounds of the invention, preferentially dissociative complexes (e.g., crystalline diastereomeric salts). Diastereomers have significantly different physical properties (e.g., melting points, boiling points, solubilities, reactivities, etc.) and can be readily separated by virtue of these dissimilarities. Diastereomers may be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that does not racemize. A more detailed description of techniques suitable for resolution of stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, Andre Collet, Samue1 H.Wilen, "Enantiomers, Racemates And resolution" ("Enantiomers, racemes And resolution"), John Wiley And Sons, Inc., 1981.
The reaction may be monitored by any suitable method known in the art. For example, it can be determined by spectroscopic means, such as Nuclear Magnetic Resonance (NMR) spectroscopy (e.g.1H or13C) Infrared (IR) spectroscopy, spectrophotometry (e.g., UV-visible light), Mass Spectrometry (MS)), or by chromatographic methods such as High Performance Liquid Chromatography (HPLC) or Thin Layer Chromatography (TLC).
In the general synthetic methods and examples, the meanings of the abbreviations are as follows.
Boc: tert-butoxycarbonyl group
DBU: diazabicycloundecenes
DMA: n, N-dimethyl acetamide
DMF: n, N-dimethylformamide
NMP: n-methyl pyrrolidone
THF: tetrahydrofuran (THF)
And (3) OBn: benzyloxy radical
T3P: propyl phosphonic anhydride
PPT: pyridinium p-toluenesulfonate
MSA: methanesulfonic acid
ACN: acetonitrile
MTBE: methyl tert-butyl ether
The compounds of formula (I) according to the invention can be prepared by following scheme 1:
reaction scheme 1
Figure GDA0003592353000000231
Wherein R is3、R7-R15Each independently of the otherIs selected from hydrogen or deuterium, and R3、R7-R15Not all hydrogen.
The compound of formula (C) can be prepared by reacting the compound of formula (a) and the compound of formula (B) in the presence of a suitable condensing agent (e.g., propylphosphoric anhydride (T3P), methanesulfonic acid, or p-toluenesulfonic acid), a suitable solvent (e.g., a solvent such as DMF, DMA, NMP, THF, ethyl acetate, butyl acetate, dioxane, or the like, or a mixed solvent of these) and a suitable base (e.g., sodium carbonate, potassium carbonate, cesium carbonate, or the like). The reaction is carried out at a temperature ranging from about 10 ℃ to 80 ℃ and may take from about 1 to 24 hours to complete. The compound of formula (I-1) can be synthesized by leaving a protecting group Bn from The compound of formula (C) (for example, it can be carried out by a general method described in Protective Groups in Organic Synthesis, Theodora W Green (John Wiley & Sons et al.) The compound of formula (I-2) can be obtained by a general method of converting a compound of formula (I-1) and a compound of formula (D) into an ether group by a hydroxyl group in The presence of an appropriate base (for example, it can be obtained by a method described in Protective Groups in Organic Synthesis, Theodora W Green (John Wiley & Sons), prog. Med.5: 2157-2161(1985), and Supplied by The chemical fiber-The word's Knoedge et al).
The compound of formula (A-1) can be prepared by the following reaction scheme 2:
reaction scheme 2
Figure GDA0003592353000000241
Preparation routes and synthetic methods for the compounds of formula (A-1) and formula (E-1) have been disclosed in WO2017/221869A 1.
The compound of formula (A-2) can be prepared by the following reaction scheme 3:
reaction scheme 3
Figure GDA0003592353000000242
The preparation route and the synthesis method of the compound of the formula (A-2) are the same as those of the compound of the formula (A-1), except that the compound F-1 is replaced by a deuterated compound F-2.
The compounds of formula (B-1) and (B-2) can be prepared by the following reaction scheme 4:
reaction scheme 4
Figure GDA0003592353000000251
Preparation routes and synthetic methods for compounds of formula (B-1) have been disclosed in WO2017/221869A 1. The process of the contract for the compound of formula (B-2) differs from the compound of formula (B-1) in that the last step replaces NaBH with a deuterated metal reducing agent4
Preparation of intermediate compound 9
Figure GDA0003592353000000252
The synthetic route is as follows:
Figure GDA0003592353000000253
step 1 Synthesis of Compound 3
Compound 1(2.0g, 1.05mmol) and compound 2(2.3g, 1.7mmol) were added to a DMA (20ml) solution, the solution was heated to 40 ℃, then sodium tert-butoxide (1.6g, 1.6mmol) was slowly added, the reaction was stirred at 40 ℃ for 3 hours, after completion of the reaction, cooled to room temperature, then acetic acid was slowly added dropwise to adjust the pH to about 7, 10ml of water was added, extraction was performed with dichloromethane (50ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v): 10:1) to give 2.1g of a white solid in 72% yield.
Step 2 Synthesis of Compound 4
Adding compound 3(2.0g, 7.2mmol) into ethanol (10ml) and water (10ml), heating the solution to 60 ℃, then adding hydrazine hydrate (0.7g, 14.3mmol), stirring at 60 ℃ for reaction for 4 hours, after the reaction is completed, slowly adding 20% sodium hydroxide aqueous solution to adjust the pH to be about 13, extracting with dichloromethane (100ml × 3), combining organic phases, drying with anhydrous sodium sulfate, concentrating, and directly feeding the concentrated solution into the next step.
Step 3 Synthesis of Compound 6
Compound 5(5.0g, 20.5mmol), methyl iodide (3.5g, 24.4mmol), DBU (6.3g, 40.7mmol) were added to DMF (20ml) in this order, the reaction was reacted at room temperature for 20h, then 80ml of water was added, followed by extraction with ethyl acetate (60ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 5:1) to give 3.6g of a yellow solid in 69% yield.
Step 4 Synthesis of Compound 7
Compound 6(0.5g, 1.8mmol), pyridinium p-toluenesulfonate (1.38g, 5.5mmol), and NH were successively added2NHBoc (0.36g, 2.7mmol) was added to DMA (10ml) solution and reacted at 60 ℃ for 15h, then 20ml water was added and extracted with ethyl acetate (30ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 4:1) to give 0.6g of a yellow solid in 80% yield.
Step 5 Synthesis of Compound 8
Compound 7(1.0g, 2.7mmol) and compound 4(0.8g, 5.3mmol) were added to THF (20ml) solution in this order, then 2 drops of DBU were added dropwise, the solution was reacted at 60 ℃ for 20 hours, then the reaction solution was spin-dried, 20ml of water was added, followed by extraction with ethyl acetate (30ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 4:1) to obtain 0.7g of a solid, yield 54%.
Step 6 Synthesis of Compound 9
Compound 8(1.5g, 3.1mmol), methanesulfonic acid (0.43g, 4.5mmol) were added to acetonitrile (20ml) solution in this order, the solution was reacted at 60 ℃ for 5 hours, then the reaction solution was spin-dried, 20ml of water was added, extraction was performed with ethyl acetate (30ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 1:2) to obtain 0.5g of a solid, yield 51%.
Preparation of intermediate Compound 17
Figure GDA0003592353000000271
The synthetic route is as follows:
Figure GDA0003592353000000272
step 1 Synthesis of Compound 11
At 0 deg.C, mixing LiAlD4(7.2g, 170.9mmol) was slowly added to a solution of Compound 10(10.0g, 85.6mmol) in tetrahydrofuran (150ml), and after the addition was complete, the reaction was warmed to reflux for 10 h. After the reaction is completed, the temperature is reduced to 0 ℃, then water is added to quench the reaction, diatomite is used for filtration, and the filtrate is directly dried by spinning to obtain 4.4g of liquid which is directly put into the next step.
Step 2 Synthesis of Compound 13
Compound 12(5.0g, 34.0mmol), compound 11(4.4g, 68.0mmol), iron nitrate nonahydrate (1.3g, 3.4mmol) were added to a toluene (40ml) solution in this order, the reaction solution was reacted at 115 ℃ for 15 hours, then 20ml of water was added, followed by extraction with ethyl acetate (50ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 5:1) to give 3.9g of a white solid in 59% yield.
Step 3 Synthesis of Compound 14
Compound 13(4.0g, 21.0mmol) and compound 2(4.6g, 34mmol) were added to a DMA (20ml) solution, the solution was heated to 40 ℃, then sodium tert-butoxide (3.2g, 3.2mmol) was slowly added, the reaction was stirred at 40 ℃ for 3 hours, after completion of the reaction, acetic acid was slowly added dropwise to adjust the pH to about 7, 10ml of water was added, dichloromethane (50ml × 3) was used for extraction, the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 10:1) to give 3.7g of a white solid in 63% yield.
Step 4 Synthesis of Compound 15
Adding compound 14(2.0g, 7.2mmol) into ethanol (10ml) and water (10ml), heating the solution to 60 ℃, then adding hydrazine hydrate (0.7g, 14.3mmol), stirring at 60 ℃ for reaction for 4 hours, after the reaction is completed, slowly adding 20% sodium hydroxide aqueous solution to adjust the pH value to be about 13, extracting with dichloromethane (100ml × 3), combining organic phases, drying with anhydrous sodium sulfate, concentrating, and directly feeding the concentrated solution to the next step.
Step 5 Synthesis of Compound 16
Compound 7(1.0g, 2.7mmol) and compound 15(0.8g, 5.3mmol) were added sequentially to a THF (20ml) solution, then 2 drops of DBU were added dropwise, the solution was reacted at 60 ℃ for 20h, then the reaction solution was spin-dried, 20ml of water was added, extraction was performed with ethyl acetate (30ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 4:1) to give 0.8g of a solid, yield 60%.
Step 6 Synthesis of Compound 17
Compound 16(1.0g, 2.1mmol) and methanesulfonic acid (0.3g, 3.0mmol) were added successively to a solution of acetonitrile (20ml), the solution was reacted at 60 ℃ for 5 hours, then the reaction solution was spin-dried, 20ml of water was added, extraction was performed with ethyl acetate (30ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: petroleum ether/ethyl acetate (v/v) ═ 1:2) to give 0.45g of a solid in 68% yield.
Preparation of intermediate compound 23
Figure GDA0003592353000000281
The synthetic route is as follows:
Figure GDA0003592353000000291
step 1 Synthesis of Compound 19
Under the protection of nitrogen, the compound LDA (35ml, 70mmol) is added into anhydrous THF (50ml), the solution is cooled to-40 ℃, then compound 18(5g, 31.6mmol) is added dropwise into tetrahydrofuran solution, reaction is continued for 1.5h at the temperature after the addition, then DMF (5.74g, 78.5mmol) is slowly added into the reaction solution, hydrochloric acid (34ml,6mol/L) is added after the addition, stirring is carried out for ten minutes, extraction is carried out by using ethyl acetate (50ml multiplied by 3), organic phases are combined, anhydrous sodium sulfate is dried, and yellow liquid 6g is obtained after concentration.
Step 2 Synthesis of Compound 20
Adding thiophenol (3.9g,35.4mmol) and d-camphorsulfonic acid (1.6g,5.0mmol) into the toluene solution of the above compound 6, stirring at 60 ℃ for reaction for 4h, after the reaction is completed, cooling to 0 ℃, then adding sodium hydroxide (10ml,2mol/L), then extracting with toluene (20 ml. times.3), washing with water, combining the organic phases, drying with anhydrous sodium sulfate, concentrating, and directly feeding the concentrated solution to the next step.
Step 3 Synthesis of Compound 21
Adding aluminum trichloride (5.52g, 41.4mmol) into toluene (30ml), cooling the solution to 0 ℃, then slowly dripping tetramethyldisiloxane (5.56g, 41.4mmol) into the solution, then slowly adding compound 20 into the solution at room temperature, reacting for 3h at room temperature after the addition is finished, slowly adding 15% sulfuric acid (35ml) after the reaction is finished, then extracting with ethyl acetate (50ml × 3), combining organic phases, drying with anhydrous sodium sulfate, concentrating to dryness, and pulping with petroleum ether to obtain 7g of white solid, 79%.
Step 4 Synthesis of Compound 22
Compound 21(7.0g, 25.0mmol) was added to polyphosphoric acid (30ml), the solution was warmed to 120 ℃ and reacted at this temperature for 3h, after completion of the reaction was slowly poured into ice water, then extracted with ethyl acetate (60ml x 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated to dryness and slurried with petroleum ether to give 5g, 77% of a white solid.
Step 5 Synthesis of Compound 23
At 40 deg.C, adding NaBH4(0.4g, 11.5mmol) was added to a solution of compound 22(2.0g, 7.6mmol) in isopropanol alcohol (20ml) and the reaction was continued for 1 h. The reaction was then quenched with 1M hydrochloric acid, dichloromethaneExtraction with an alkane (30 ml. times.2) and combination of the organic phases, drying over anhydrous sodium sulfate, filtration and spin-drying gave 1.8g of product as a grey solid.
Synthesis of intermediate Compound 24
Figure GDA0003592353000000301
The following synthetic route is adopted:
Figure GDA0003592353000000302
under the condition of 40 ℃, NaBD is added4(0.5g, 11.5mmol) was added to a solution of compound 22(2.0g, 7.6mmol) in isopropanol alcohol (20ml) and the reaction was continued for 1 h. The reaction was then quenched with 1M hydrochloric acid, extracted with dichloromethane (30 ml. times.2), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered and spun dry to give the product as a grey solid, 1.8 g.
Example 1(((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl- 11-d) -6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) Preparation of triazin-7-yl) oxy) methyl methylcarbonate (Compound T-1)
Figure GDA0003592353000000303
The synthetic route is as follows
Figure GDA0003592353000000311
Step 1 Synthesis of Compound 25
Compound 9(0.5g, 1.5mmol), compound 24(0.45g, 1.5mmol) and T3P (0.73g, 2.3mmol) were added to ethyl acetate (6ml) in this order, followed by addition of methanesulfonic acid (0.29g, 3.0mmol), reaction of the reaction mixture at 70 ℃ for 6h, addition of 10ml of water, extraction with ethyl acetate (20ml × 3), combination of the organic phases, drying over anhydrous sodium sulfate, and column separation of the concentrate (eluent: petroleum ether/ethyl acetate (v/v) ═ 1:2) to give 0.6g of a white solid in 69% yield.
Step 2 Synthesis of Compound 26
Compound 25(0.6g, 1.0mmol) and lithium chloride (0.23g, 5.0mmol) were added to DMA (10ml), the solution was warmed to 80 ℃ and reacted at that temperature for 3h, after completion of the reaction was slowly poured into ice water, followed by extraction with ethyl acetate (20ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated to dryness and slurried with MTBE to give 0.4g of a white solid in 81% yield.
Step 3 Synthesis of Compound 27
Compound 26(0.3g, 0.61mmol), methylchloromethyl carbonate (0.15g, 1.3mmol), potassium carbonate (0.2g, 1.22mmol), potassium iodide (0.1g, 0.61mmol) were added to DMA (6ml) solution in this order, the reaction solution was reacted at 50 ℃ for 6h, then 10ml of water was added, followed by extraction with ethyl acetate (20ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: DCM/MeOH (v/v) ═ 20:1) to obtain 0.21g of a white solid.
Step 4 Synthesis of Compound T-1
0.21g of Compound 27 was dissolved in 20mL of methanol and prepared using the following conditions (apparatus: SFC-80 (rear, Waters); chiral column: OJ-H20 × 250mm,10um (Daicel); column temperature: 35 ℃; fluidity: CO)2Methanol (0.2% ammonia in methanol) 87/13; flow rate: 80 g/min; column pressure: 100 bar; detection wavelength: 214 nm; cycle time: 6.0 min; volume of each sample injection: 1.0mL) to yield four isomers, and chiral analysis was performed using the following conditions (instrument: agilent 1200; chiral column: IG 4.6X 250mm,5um (Decial); column temperature: 25 ℃; mobile phase: n-hexane (containing 0.1 diethylamine): ethanol (containing 0.1% diethylamine) 60: 40; flow rate: 1.0 mL/min; detection wavelength: 214 nm; sample introduction amount: 15uL) with a retention time of 35.65min, compound T-1, 45mg of white compound. LC-MS M/z 573.2(M +1)+,UV 214;1H NMR(500MHz,CDCl3)δ/ppm:7.14-7.08(m,4H),7.03-7.00(m,1H),6.89-6.87(m,1H),6.83-6.81(m,1H),5.99(s,2H),5.87(d,J=7.5Hz,1H),5.27(dd,J=14.0Hz,J=2.5Hz,1H),4.63(dd,J=14.0Hz,J=2.5Hz,1H),4.51(dd,J=10.0Hz,J=2.5Hz,1H),4.06(d,J=14.0Hz,1H),3.93(dd,J=11.0Hz,J=3.0Hz,1H),3.86(s,3H),3.76(dd,J=11.0Hz,J=3.0Hz,1H),3.55(t,J=10.5Hz,1H),3.46-3.41(m,1H),2.96-2.91(m,1H).
Example 2(((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl- 11-d) -6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) Triazin-7-yl-3, 3,4,4-d4 ) Preparation of oxy) methyl methylcarbonate (Compound T-2)
Figure GDA0003592353000000321
The synthetic route is as follows
Figure GDA0003592353000000331
Step 1 Synthesis of Compound 28
Compound 17(0.5g, 1.5mmol), compound 24(0.45g, 1.5mmol) and T3P (0.73g, 2.3mmol) were added to a solution of ethyl acetate (6ml) in this order, followed by addition of methanesulfonic acid (0.29g, 3.0mmol), reaction of the reaction mixture at 70 ℃ for 6h, addition of 10ml of water, extraction with ethyl acetate (20ml × 3), combination of the organic phases, drying over anhydrous sodium sulfate, and column separation of the concentrate (eluent: petroleum ether/ethyl acetate (v/v) ═ 1:2) to give 0.55g of a white solid in 60% yield.
Step 2 Synthesis of Compound 29
Compound 28(0.5g, 0.86mmol) and lithium chloride (0.2g, 4.3mmol) were added to DMA (10ml), the solution was warmed to 80 ℃ for reaction at that temperature for 3h, after completion of the reaction was poured slowly into ice water and then extracted with ethyl acetate (20ml x 3), the organic phases were combined, dried over anhydrous sodium sulphate, concentrated to dryness and slurried with MTBE to give a white solid 0.33g, 78%.
Step 3 Synthesis of Compound 30
Compound 29(0.33g, 0.67mmol), methylchloromethyl carbonate (0.20g, 1.4mmol), potassium carbonate (0.25g, 1.4mmol) and potassium iodide (0.1g, 0.67mmol) were added to a DMA (6ml) solution in this order, the reaction solution was reacted at 50 ℃ for 6h, then 10ml of water was added, followed by extraction with ethyl acetate (20ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: DCM/MeOH (v/v) ═ 20:1) to give 0.25g of a white solid.
Step 4 Synthesis of Compound T-2
0.25g of Compound 30 was dissolved in 25mL of methanol and prepared using the following conditions (apparatus: SFC-80 (rear, Waters); chiral column: OJ-H20 × 250mm,10um (Daicel); column temperature: 35 ℃; fluidity: CO)2Methanol (0.2% ammonia in methanol) 87/13; flow rate: 80 g/min; column pressure: 100 bar; detection wavelength: 214 nm; cycle time: 6.0 min; volume of each injection: 1.0mL) to yield four isomers, and chiral analysis was performed using the following conditions (instrument: agilent 1200; chiral column: IG 4.6X 250mm,5um (Decial); column temperature: 25 ℃; mobile phase: n-hexane (containing 0.1 diethylamine): ethanol (containing 0.1% diethylamine) 60: 40; flow rate: 1.0 mL/min; detection wavelength: 214 nm; sample introduction amount: 15uL) with a retention time of 36.53min, compound T-2, white compound 50 mg. LC-MS M/z 577.3(M +1)+,UV 214;1H NMR(500MHz,CDCl3)δ/ppm:7.14-7.08(m,4H),7.03-7.00(m,1H),6.89-6.87(m,1H),6.83-6.81(m,1H),5.99(s,2H),5.87(d,J=7.5Hz,1H),5.27(dd,J=14.0Hz,J=2.5Hz,1H),4.51(dd,J=10.0Hz,J=2.5Hz,1H),4.06(d,J=14.0Hz,1H),3.93(dd,J=11.0Hz,J=3.0Hz,1H),3.86(s,3H),3.55(t,J=10.5Hz,1H),3.12-3.08(m,1H).
Example 3(((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl) - 6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) triazine-7- Yl) oxy) methyl carbonic acid (methyl-d3 ) Preparation of ester (Compound T-3)
Figure GDA0003592353000000341
The synthetic route is as follows
Figure GDA0003592353000000351
Step 1 Compound Carbonic acid (methyl-d)3) Synthesis of chloromethyl esters
Deuterated methanol (1.0g, 28.7mmol) and triethylamine (3.4g, 34.2mmol) were added to dichloromethane (30ml), the solution was cooled to 0 ℃, then chloromethyl chloroformate (3.7g, 28.7mmol) was slowly added dropwise to the above solution, followed by reaction at room temperature for 3h, quenching with water (10ml) after completion of the reaction, extraction with DCM (20ml), combining the organic phases, drying over anhydrous sodium sulfate, and concentration to dryness to give 2.8g of a liquid in 78% yield.
Step 2 Synthesis of Compound 31
Compound 9(0.8g, 2.45mmol), compound 23(0.65g, 2.45mmol) and T3P (1.2g, 3.7mmol) were added to a solution of ethyl acetate (6ml) in this order, followed by addition of methanesulfonic acid (0.45g, 4.9mmol), reaction of the reaction mixture at 70 ℃ for 6h, addition of 10ml of water, extraction with ethyl acetate (20ml × 3), combination of the organic phases, drying over anhydrous sodium sulfate, column separation of the concentrate (eluent: petroleum ether/ethyl acetate (v/v) ═ 1:2) to give 0.9g of a white solid in 64% yield.
Step 3 Synthesis of Compound 32
Compound 31(0.9g, 1.57mmol) and lithium chloride (0.33g, 7.85mmol) were added to DMA (10ml), the solution was warmed to 80 ℃ for reaction at that temperature for 3h, after completion of the reaction was slowly poured into ice water, followed by extraction with ethyl acetate (20 ml. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated to dryness and slurried with MTBE to give 0.6g, 79% of a white solid.
Step 4 Synthesis of Compound 33
Compound 32(0.6g, 1.2mmol), carbonic acid (methyl-d) were sequentially added3) Chloromethyl ester (0.3g, 2.4mmol), potassium carbonate (0.4g, 2.4mmol) and potassium iodide (0.2g, 1.2mmol) was added to DMA (6ml), the reaction was reacted at 50 ℃ for 6h, then 10ml water was added, followed by extraction with ethyl acetate (20ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: DCM/MeOH (v/v) ═ 20:1) to give 0.55g of white solid.
Step 5 Synthesis of Compound T-3
0.55g of Compound 33 was dissolved in 50mL of methanol and prepared using the following conditions (apparatus: SFC-80 (rear, Waters); chiral column: OJ-H20 × 250mm,10um (Daicel); column temperature: 35 ℃; fluidity: CO)2Methanol (0.2% ammonia in methanol) 87/13; flow rate: 80 g/min; column pressure: 100 bar; detection wavelength: 214 nm; cycle time: 6.0 min; volume of each sample injection: 1.0mL) to yield four isomers, and chiral analysis was performed using the following conditions (instrument: agilent 1200; chiral column: IG 4.6X 250mm,5um (Decial); column temperature: 25 ℃; mobile phase: n-hexane (containing 0.1 diethylamine): ethanol (containing 0.1% diethylamine) 60: 40; flow rate: 1.0 mL/min; detection wavelength: 214 nm; sample injection amount: 15uL) was added, wherein the retention time was 37.24min, which was compound T-3, which was 100mg of a white solid. LC-MS: M/z 575.0(M +1)+,UV 214;1H NMR(500MHz,CDCl3)δ/ppm:7.14-7.08(m,4H),7.03-7.00(m,1H),6.89-6.87(m,1H),6.83-6.81(m,1H),5.99(s,2H),5.87(d,J=7.5Hz,1H),5.33(s,1H),5.27(dd,J=14.0Hz,J=2.5Hz,1H),4.63(dd,J=14.0Hz,J=2.5Hz,1H),4.51(dd,J=10.0Hz,J=2.5Hz,1H),4.06(d,J=14.0Hz,1H),3.93(dd,J=11.0Hz,J=3.0Hz,1H),3.76(dd,J=11.0Hz,J=3.0Hz,1H),3.55(t,J=10.5Hz,1H),3.46-3.41(m,1H),2.96-2.91(m,1H).
Example 4(((12aR) -12- ((11S) -7, 8-difluoro-6, 11-dihydrodibenzo (B, E) thiepin-11-yl- 11-d) -6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-1H- (1,4) oxazine (3,4-C) pyrido (2,1-F) (1,2,4) Triazin-7-yl) oxy) methylcarbonate (methyl-d3 ) Preparation of ester (Compound T-4)
Figure GDA0003592353000000361
The synthetic route is as follows
Figure GDA0003592353000000371
Step 1 Synthesis of Compound 34
Compound 26(0.5g, 1.01mmol), carbonic acid (methyl-d) were sequentially reacted3) Chloromethyl ester (0.26g, 2.06mmol), potassium carbonate (0.28g, 2.06mmol) and potassium iodide (0.1g, 1.01mmol) were added to a DMA (6ml) solution, the reaction solution was reacted at 50 ℃ for 6 hours, then 10ml of water was added, extraction was performed with ethyl acetate (20ml × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and the concentrate was subjected to column separation (eluent: DCM/MeOH (v/v) ═ 20:1) to give 0.4g of white solid.
Step 2 Synthesis of Compound T-4
0.4g of Compound 34 was dissolved in 40mL of methanol and prepared using the following conditions (apparatus: SFC-80 (rear, Waters); chiral column: OJ-H20 × 250mm,10um (Daicel); column temperature: 35 ℃; fluidity: CO)2Methanol (0.2% ammonia in methanol) 87/13; flow rate: 80 g/min; column pressure: 100 bar; detection wavelength: 214 nm; cycle time: 6.0 min; volume of each sample injection: 1.0mL) to yield four isomers, and chiral analysis was performed using the following conditions (instrument: agilent 1200; chiral column: IG 4.6X 250mm,5um (Decial); column temperature: 25 ℃; mobile phase: n-hexane (containing 0.1 diethylamine): ethanol (containing 0.1% diethylamine) 60: 40; flow rate: 1.0 mL/min; detection wavelength: 214 nm; sample introduction amount: 15uL) was added, wherein the retention time was 33.77min, which was compound T-4, 80mg of white solid. LC-MS M/z 576.3(M +1)+,UV 214;1H NMR(500MHz,CDCl3)δ/ppm:7.14-7.08(m,4H),7.03-7.00(m,1H),6.89-6.87(m,1H),6.83-6.81(m,1H),5.99(s,2H),5.87(d,J=7.5Hz,1H),5.27(dd,J=14.0Hz,J=2.5Hz,1H),4.63(dd,J=14.0Hz,J=2.5Hz,1H),4.51(dd,J=10.0Hz,J=2.5Hz,1H),4.06(d,J=14.0Hz,1H),3.93(dd,J=11.0Hz,J=3.0Hz,1H),3.76(dd,J=11.0Hz,J=3.0Hz,1H),3.55(t,J=10.5Hz,1H),3.46-3.41(m,1H),2.96-2.91(m,1H).
And (4) testing the biological activity.
(1) Metabolic stability evaluation
Microsome experiment: human liver microsomes (Human liver microsomes, HLM): 0.5mg/mL, Xenotech; rat liver microsomes (Rat liver microsomes, RLM): 0.5mg/mL, Xenotech; coenzyme (NADPH/NADH): 1mM, Sigma Life Science; magnesium chloride: 5mM, 100mM phosphate buffer (pH 7.4).
Preparing a stock solution: an amount of each of the powders of the example compound and the control compound was precisely weighed and dissolved in DMSO to 5mM, respectively.
Preparation of phosphate buffer (100mM, pH 7.4): 150mL of 0.5M potassium dihydrogenphosphate and 700mL of 0.5M dipotassium hydrogenphosphate solution prepared in advance were mixed, the pH of the mixture was adjusted to 7.4 with the 0.5M dipotassium hydrogenphosphate solution, diluted 5-fold with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer solution (100mM) containing 100mM potassium phosphate and 3.3mM magnesium chloride at a pH of 7.4.
NADPH regenerating system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U/mL G-6-P D, 3.3mM magnesium chloride) was prepared and placed on wet ice before use.
Preparing a stop solution: acetonitrile solution containing 50ng/mL propranolol hydrochloride and 200ng/mL tolbutamide (internal standard). 25057.5 mu L of phosphate buffer solution (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of human liver microsome is respectively added and mixed evenly, and liver microsome dilution liquid with the protein concentration of 0.625mg/mL is obtained. 25057.5 mu L of phosphate buffer (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of SD rat liver microsome is respectively added, and the mixture is mixed evenly to obtain liver microsome dilution with the protein concentration of 0.625 mg/mL.
Incubation of the sample: the stock solutions of the corresponding compounds were diluted to 0.25mM each with an aqueous solution containing 70% acetonitrile, and used as working solutions. 398. mu.L of human liver microsome or rat liver microsome dilutions were added to a 96-well plate (N2), 2. mu.L of 0.25mM working solution was added, and mixed well.
Determination of metabolic stability: 300. mu.L of pre-cooled stop solution was added to each well of a 96-well deep-well plate and placed on ice as a stop plate. The 96-well incubation plate and the NADPH regeneration system are placed in a 37 ℃ water bath box, shaken at 100 rpm and pre-incubated for 5 min. 80. mu.L of the incubation solution was taken out of each well of the incubation plate, added to the stop plate, mixed well, and supplemented with 20. mu.L of NADPH regenerating system solution as a 0min sample. Then 80. mu.L of NADPH regenerating system solution was added to each well of the incubation plate, the reaction was started, and the timer was started. The reaction concentration of the corresponding compound was 1. mu.M, and the protein concentration was 0.5 mg/mL. When the reaction was carried out for 10min, 30 min and 90min, 100. mu.L of each reaction solution was added to the stop plate and vortexed for 3min to terminate the reaction. The stop plates were centrifuged at 5000 Xg for 10min at 4 ℃. And (3) taking 100 mu L of supernatant to a 96-well plate in which 100 mu L of distilled water is added in advance, mixing uniformly, and performing sample analysis by adopting LC-MS/MS.
And (3) data analysis: and detecting peak areas of the corresponding compound and the internal standard through an LC-MS/MS system, and calculating the peak area ratio of the compound to the internal standard. The slope is determined by plotting the natural logarithm of the percentage of compound remaining against time and calculating t according to the following formula1/2And CLintWhere V/M is equal to 1/protein concentration.
Figure GDA0003592353000000381
t1/2(min);CLint(μL/min/mg)。
The compounds of the invention and compounds without deuteration were tested simultaneously and compared to evaluate their metabolic stability in human and rat liver microsomes. The non-deuterated compound Baloxavir marboxil was used as a control. In human and rat liver microsome experiments, the compound of the invention can obviously improve the metabolic stability by comparing with the non-deuterated compound Baloxavir marboxil. The results of the liver microsome experiments for representative example compounds are shown in table 1 below.
Table 1:
Figure GDA0003592353000000391
(2) pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 per group and compared for pharmacokinetic differences by intravenous or oral administration of a single dose of compound (10 mg/kg oral).
Rats were fed with standard feed and given water. Fasting began 16 hours prior to the experiment. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L of blood was collected from the orbit into a test tube. In the test tube there was 30. mu.L of 1% heparin salt solution. Before use, the tubes were dried overnight at 60 ℃. After completion of blood collection at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood collection, the tubes were gently inverted at least 5 times to ensure mixing and then placed on ice. The blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate the plasma from the erythrocytes. Pipette 100 μ L of plasma into a clean plastic centrifuge tube, designating the name of the compound and the time point. Plasma was stored at-80 ℃ before analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
Experiments show that the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and treatment effect.
(3) In vitro anti-influenza virus active virus strains: influenza virus a/WSN/33(H1N1), cell: MDCK;
the antiviral activity of the test compounds against influenza strains was tested using the Cytopathic (CPE) method, while the cytotoxicity was determined. Compounds were tested at 8 concentrations, double-well. Cell viability was measured using CCK-8 reagent. The starting concentration of compound was 500nM, the final concentration was 0.229nM, and the dilution was done in 3-fold gradient.
MDCK cells were seeded at 2000/well in 384-well plates at 37 ℃ in 5% CO2The culture was carried out overnight. The next day compounds and virus were added, and cells (no virus infection) and virus infection controls were set. Cell culture DMSO final concentration was 0.5%. Cells at 37 ℃ and 5% CO2Culturing for 5 days until the cell morbidity of the virus control wells reaches 80-95%. CellsToxicity experiments were identical to antiviral experiments, but without viral infection. Cell viability was measured using CCK-8 reagent and raw data was used for compound antiviral activity and cytotoxicity calculations. Compound dose response curves were analyzed and EC calculated using GraphPad Prism software50And CC50The value is obtained.
The compound of the invention and the compound without deuteration are tested and compared at the same time, and the antiviral activity and the cytotoxicity are evaluated. The results show that compared with the non-deuterated compound Baloxavir marboxil, the compound of the invention has higher CPE (CPE) inhibition effect, so that the compound of the invention can inhibit cap-dependent endonuclease (CDE) in influenza virus more effectively, thereby playing a role in inhibiting virus replication. The results of antiviral activity and cytotoxicity of representative example compounds are shown in table 2 below.
TABLE 2
EXAMPLES Compounds EC50(nM) CC50(nM)
Baloxavir marboxil 10.11 >500
T-1 10.05 >500
T-2 3.54 >500
T-3 6.11 >500
T-4 5.94 >500
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (4)

1. A compound selected from the group consisting of compounds of the formula:
Figure FDA0003567436900000011
or a pharmaceutically acceptable salt thereof.
2. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of claim 1 or a pharmaceutically acceptable salt thereof.
3. Use of the compound of claim 1 or a pharmaceutically acceptable salt thereof or the pharmaceutical composition of claim 2 for the manufacture of a medicament for the treatment and/or prevention of diseases caused by a virus having a cap-dependent endonuclease.
4. The use according to claim 3, wherein the disease caused by a virus having a cap-dependent endonuclease is selected from influenza A, influenza B or influenza C.
CN201910675084.1A 2018-07-27 2019-07-25 Substituted polycyclic pyridone compound and prodrug thereof Active CN110317211B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810841388 2018-07-27
CN2018108413886 2018-07-27

Publications (2)

Publication Number Publication Date
CN110317211A CN110317211A (en) 2019-10-11
CN110317211B true CN110317211B (en) 2022-05-13

Family

ID=68124543

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910675084.1A Active CN110317211B (en) 2018-07-27 2019-07-25 Substituted polycyclic pyridone compound and prodrug thereof

Country Status (2)

Country Link
CN (1) CN110317211B (en)
WO (1) WO2020020267A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108440564B (en) * 2018-04-11 2019-08-06 安帝康(无锡)生物科技有限公司 Substituted polycyclic carbamoylpyridone derivative and its prodrug
CN111825699B (en) * 2019-04-15 2023-02-21 上海复星星泰医药科技有限公司 Sulfur-containing heterocyclic compounds and process for producing the same
CN112707862B (en) * 2019-10-24 2022-09-27 广东东阳光药业有限公司 Preparation method of polycyclic pyridone compound
CN111057070A (en) * 2019-10-30 2020-04-24 浙江工业大学 Synthesis method of baroxavir key intermediate
CN115175913B (en) * 2019-11-13 2023-11-28 南京知和医药科技有限公司 Substituted bistricycles and pharmaceutical compositions and uses thereof
CN111233891B (en) * 2020-03-04 2021-05-04 江苏柯菲平医药股份有限公司 Fused ring pyridone derivative and preparation method and application thereof
CN111808069B (en) * 2020-07-21 2022-05-17 中国药科大学 Preparation method of baroxavir key intermediate and intermediate thereof
CN112409379B (en) * 2020-09-28 2023-07-28 长沙晶易医药科技股份有限公司 Deuterated dihydrodibenzothiazepine compound and pharmaceutical composition containing same
CN112898312B (en) * 2021-01-29 2021-11-12 湖南南新制药股份有限公司 Fused polycyclic pyridone derivative and application thereof
CN113527119A (en) * 2021-06-30 2021-10-22 和鼎(南京)医药技术有限公司 Preparation method of Barosavir intermediate
CN113717199B (en) * 2021-08-13 2022-04-01 北京北朋科技有限公司 Deuterated derivative of baroxavir and application of deuterated derivative in anti-influenza virus
CN113549088B (en) * 2021-08-25 2022-10-04 北京理工大学 Preparation method of baroxavir key intermediate
CN116284048B (en) * 2023-05-18 2023-08-15 长沙晶易医药科技股份有限公司 Compound and preparation method, pharmaceutical composition and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TR201810736T4 (en) * 2010-09-24 2018-08-27 Shionogi & Co Substituted polycyclic carbamoyl pyridone derivative prodrug.
EP4219508B1 (en) * 2015-04-28 2024-06-05 Shionogi & Co., Ltd Substituted polycyclic pyridone derivative and prodrug thereof
CN110494141A (en) * 2016-08-10 2019-11-22 盐野义制药株式会社 Pharmaceutical composition containing substituted polycyclic Pyridione derivatives and its prodrug
CN109503625A (en) * 2018-01-19 2019-03-22 赵蕾 A kind of polycyclic pyridines ketone compound and its pharmaceutical composition and purposes
CN108440564B (en) * 2018-04-11 2019-08-06 安帝康(无锡)生物科技有限公司 Substituted polycyclic carbamoylpyridone derivative and its prodrug

Also Published As

Publication number Publication date
CN110317211A (en) 2019-10-11
WO2020020267A1 (en) 2020-01-30

Similar Documents

Publication Publication Date Title
CN110317211B (en) Substituted polycyclic pyridone compound and prodrug thereof
AU2021331214B2 (en) Compounds and methods for treatment of viral infections
JP6757294B2 (en) Methods for treating Filoviridae viral infections
WO2022143473A1 (en) Nucleoside compound and use thereof
JP2021165280A (en) N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US9771361B2 (en) Inhibitors of influenza viruses replication
US20210308168A1 (en) 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
ES2821394T3 (en) 7,8-dihydro-3H-pyrazino [1,2-b] pyridazin-3,5 (6H) -dione compounds and uses thereof
CA3139977A1 (en) Peptidomimetics for the treatment of coronavirus and picornavirus infections
JP6411492B2 (en) Polycyclic carbamoylpyridone compounds and their use for treating HIV infection
KR20180041748A (en) PARP inhibitors, processes for preparing crystalline forms and uses thereof
EA034868B1 (en) Pyridazinone compounds and uses thereof
WO2022174194A1 (en) 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
WO2022174179A1 (en) 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
CN111909174B (en) Crystal form of pyridone derivative, preparation method and application
CN116568688A (en) Compounds and methods for treating viral infections
CN115677698B (en) High-efficiency antiviral compound and application thereof
EP4353724A1 (en) Compound as cdk kinase inhibitor and use thereof
US20240180948A1 (en) 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
ES2829254T3 (en) Pyrrolopyrimidine nucleosides and analogs thereof useful as antiviral agents
WO2024086592A2 (en) 4'-halogen containing nucleotide and nucleoside therapeutic compositions and uses related thereto
WO2023085432A1 (en) Anti-sars-cov-2 drug
JP2023519870A (en) Tricyclic inhibitors of influenza virus endonuclease

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
GR01 Patent grant
GR01 Patent grant