CN114073702A - Application of quinolone compounds in treating or preventing hepatitis B - Google Patents

Application of quinolone compounds in treating or preventing hepatitis B Download PDF

Info

Publication number
CN114073702A
CN114073702A CN202110245747.3A CN202110245747A CN114073702A CN 114073702 A CN114073702 A CN 114073702A CN 202110245747 A CN202110245747 A CN 202110245747A CN 114073702 A CN114073702 A CN 114073702A
Authority
CN
China
Prior art keywords
formula
compound
hepatitis
medicament
hbsag
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.)
Granted
Application number
CN202110245747.3A
Other languages
Chinese (zh)
Other versions
CN114073702B (en
Inventor
李瑛颖
陈明键
仇思念
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.)
China Israel Hyde Artificial Intelligence Drug Research And Development Co ltd
Original Assignee
China Israel Hyde Artificial Intelligence Drug Research And Development Co ltd
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 China Israel Hyde Artificial Intelligence Drug Research And Development Co ltd filed Critical China Israel Hyde Artificial Intelligence Drug Research And Development Co ltd
Priority to CN202110245747.3A priority Critical patent/CN114073702B/en
Publication of CN114073702A publication Critical patent/CN114073702A/en
Application granted granted Critical
Publication of CN114073702B publication Critical patent/CN114073702B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P31/20Antivirals for DNA viruses

Abstract

The application of quinolone compounds in treating or preventing hepatitis B. The application provides an application of a compound of a general formula 1, a deutero-compound or a pharmaceutically acceptable salt thereof in preparing a medicament for treating or preventing hepatitis B. The present application also provides a pharmaceutical composition for treating or preventing hepatitis b, comprising a compound of formula 1, a deutero-compound thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier/excipient.

Description

Application of quinolone compounds in treating or preventing hepatitis B
Technical Field
The application relates to the field of anti-hepatitis B drugs, in particular to application of quinolone compounds in treating or preventing hepatitis B.
Background
Human Hepatitis B Virus (HBV) infection is a major public health problem worldwide. The hepatitis B transmission pathway is mainly through vertical transmission and horizontal transmission. Vertical transmission refers to mother-to-baby transmission; horizontal transmission is primarily through blood.
The treatment of hepatitis B is also a long-term process, and the treatment aims to inhibit or eliminate HBV to the maximum extent, relieve inflammation and necrosis of liver cells and liver fibrosis, delay and stop the progress of diseases, reduce and prevent liver decompensation, liver cirrhosis, hepatocellular carcinoma and complications thereof, thereby improving the quality of life and prolonging the survival time.
There are many hepatitis b therapeutic drugs on the market today, mainly by antiviral treatment using interferon or nucleoside analogues. In the case of interferon, recombinant DNA leukocyte interferon (IFN-. alpha.) inhibits the replication of HBV. However, interferon is often associated with strong adverse reactions including myelosuppression, effects on thyroid function and depression when used for treating hepatitis B.
Nucleoside analogues inhibit HBV production primarily by inhibiting reverse transcriptase activity during HBV replication, and clinically useful drugs include: lamivudine, famciclovir, acyclovir, Adefovir, Entecavir, Tenofovir, foscarnet and the like, and the medicaments have certain HBV inhibiting effect.
Although these reverse transcriptase inhibitors can effectively reduce HBV DNA level and make patients control HBV level, they have no direct effect on the clearance of HBV cccDNA and HBsAg because their target of action is the process of RNA reverse transcription into DNA. Therefore, the seroconversion probability of HBsAg in single-drug treatment of the nucleoside analogue is very low, hepatitis B cannot be really cured, and patients need to take the drugs for a long time or even for life.
The problems of drug resistance, high medical costs, serious side effects of the drugs, etc. which are caused by long-term administration of the above drugs, are a heavy burden on patients with hepatitis B. The key point is that at present, no medicine can completely eliminate viruses to achieve the functional cure of hepatitis B. Therefore, the urgent need in the art is to provide a new drug for treating hepatitis b, which can eliminate HBsAg and achieve a functional cure.
Disclosure of Invention
The application provides application of quinolone compounds of general formula 1 in preventing or treating hepatitis B, the compounds can reduce Hepatitis B Virus (HBV) load, HBsAg level and/or HBeAg level, even can eliminate HBsAg and HBeAg, are expected to functionally cure hepatitis B and eliminate hepatitis B virus.
In one aspect, the present application provides the use of a compound of formula 1, a deutero-derivative thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating or preventing hepatitis b:
Figure BDA0002964027430000021
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5.
In one embodiment, formula 1 above bears a substituent (R)nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure BDA0002964027430000031
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; denotes the site of binding to the rest of formula 1.
In one embodiment, the compound of formula 1 is the following compound:
Figure BDA0002964027430000032
in one embodiment, the compound of formula 1 is a deuterated compound.
In one embodiment, the medicament is for reducing Hepatitis B Virus (HBV) load, HBsAg levels, and/or HBeAg levels.
In one embodiment, the medicament is for simultaneously reducing Hepatitis B Virus (HBV) load, HBsAg levels and HBeAg levels.
In one embodiment, the medicament is for reducing the level of HBsAg and HBeAg simultaneously.
In one embodiment, the medicament further comprises one or more additional therapeutic or prophylactic agents.
In one embodiment, the additional therapeutic or prophylactic agent is selected from at least one of an interferon, a PEGylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog,
formula A
Figure BDA0002964027430000041
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In one embodiment, the medicament is administered by a route selected from the group consisting of: oral, rectal, nasal, pulmonary, topical, buccal and sublingual, vaginal, parenteral, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural.
In one embodiment, the medicament is an oral formulation.
In one embodiment, the medicament is in the form of a tablet or capsule.
In another aspect, the present application also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1, a deutero-derivative thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier/excipient:
Figure BDA0002964027430000042
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5,
wherein the additional therapeutic or prophylactic agent is selected from at least one of an interferon, a pegylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog
Figure BDA0002964027430000051
Wherein the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate and tenofovir alafenamide.
In one embodiment, with substituent (R) in formula 1nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure BDA0002964027430000052
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; denotes the site of binding to the rest of formula 1.
In one embodiment, the compound of formula 1 is aripiprazole having the following structure:
Figure BDA0002964027430000053
in the present application, the pharmaceutically acceptable salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, thiocyanate, tosylate, undecanoate, sodium, calcium, potassium, ammonium, tetraethylammonium, methylammonium, dimethylammonium and ethanolamine salts.
In another embodiment, the compound is substituted with deuterium or is isotopically labeled. Deuterium substituted compounds are capable of increasing the half-life of the compound, delaying or slowing the metabolic process of the compound while replicating the activity of the original compound.
In one embodiment, the pharmaceutical composition is for treating or preventing hepatitis b.
In one embodiment, the pharmaceutical composition is capable of reducing Hepatitis B Virus (HBV) load, HBsAg levels, and/or HBeAg levels.
In one embodiment, the pharmaceutical composition is capable of reducing Hepatitis B Virus (HBV) load, HBsAg levels, and HBeAg levels.
The technical scheme of this application has following beneficial effect:
1. the compound of the general formula 1, a deuteron or a pharmaceutically acceptable salt thereof is applied to treating or preventing hepatitis B, thereby providing a novel hepatitis B treatment option.
2. The compound of the general formula 1, the deuteride thereof or the pharmaceutically acceptable salt thereof can simultaneously and effectively reduce the load of Hepatitis B Virus (HBV), the HBsAg level and/or the HBeAg level, even can eliminate the HBsAg and the HBeAg, and is expected to achieve the effect of functionally curing hepatitis B.
3. The compound of the general formula 1, the deuteron or the pharmaceutically acceptable salt thereof is expected to eliminate hepatitis B virus, cure hepatitis B and avoid the pain of lifelong medicine taking under the condition of being combined with the existing nucleoside analogue medicine.
4. As a marketed drug, the aripiprazole in the compound of the general formula 1, a deuteron thereof or a pharmaceutically acceptable salt thereof has excellent clinical safety and pharmacokinetic properties and has good druggability.
5. The compound of formula 1, a deuterio thereof or a pharmaceutically acceptable salt thereof can be combined with one or more additional therapeutic or prophylactic agents, particularly drugs that can reduce viral titer but cannot completely clear the virus, cannot reduce HBsAg and/or HBeAg levels, clear the hepatitis b virus from different aspects, and have the potential for synergy, thereby providing a broad idea for subsequent combination drug design.
Drawings
FIG. 1 shows the results of the inhibition of HBV DNA by the combination of aripiprazole compound of formula 1, 0.1nM Entecavir (ETV), 20. mu.M aripiprazole and 0.1nM ETV at different concentrations.
FIG. 2 shows the results of HBsAg inhibition by combinations of aripiprazole of formula 1, 0.1nM ETV, 20. mu.M aripiprazole and 0.1nM ETV at various concentrations.
FIG. 3 shows the results of HBeAg inhibition by combinations of aripiprazole of formula 1, 0.1nM ETV, 20. mu.M aripiprazole and 0.1nM ETV at various concentrations.
Detailed Description
The inventors of the present application have unexpectedly found that the compound of formula 1, its deuteride or its pharmaceutically acceptable salt described herein has potential activity of inhibiting hepatitis b virus, especially has effects of reducing Hepatitis B Virus (HBV) load, HBsAg level and/or HBeAg level, even eliminating HBsAg and HBeAg, and can be used for preventing and treating hepatitis b.
A compound of formula 1, a deuteron thereof or a pharmaceutically acceptable salt thereof
The inventors of the present application have found a group of quinolone compounds, deuterated compounds thereof, or pharmaceutically acceptable salts thereof, which can be used for treating or preventing hepatitis b, the quinolone compounds having the following general formula 1:
Figure BDA0002964027430000071
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5.
In one embodiment, with substituent (R) in formula 1nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure BDA0002964027430000081
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; denotes the site of binding to the rest of formula 1.
In one embodiment, the compound of formula 1 is aripiprazole having the following structural formula:
Figure BDA0002964027430000082
aripiprazole is known as an atypical anti-schizophrenia drug, has a bidirectional regulatory effect on the DA-ergic nervous system, and is a stabilizer of DA transmitter. Has high affinity with D2, D3, 5-HT1A and 5-HT2A receptors. The anti-schizophrenia effect is produced by partial agonism on D2 and 5-HT1A receptors and antagonism on 5-HT2A receptors. After the aripiprazole is orally taken, the peak time of the blood concentration is 3-5 hours, and the half-life period is 48-68 hours. Dehydroaripiprazole is the major active metabolite. Aripiprazole is used to treat various types of schizophrenia. The foreign clinical test shows that the aripiprazole has obvious curative effect on positive and negative symptoms of schizophrenia, can also improve accompanying emotional symptoms and reduce the recurrence rate of schizophrenia. At present, no report on the application of the traditional Chinese medicine in treating hepatitis B exists.
In one embodiment, the compound of formula 1 is a deuterated compound. The deuterated compound can replicate the activity of the original compound, improve the half-life of the compound and delay or slow down the metabolic process of the compound.
The compounds of formula 1 may be deuterated at any desired atomic position. Deuterated compounds can be prepared by methods known in the art, for example, by exchanging hydrogen for deuterium, or by synthesizing the compounds from deuterated starting materials or intermediates.
In another embodiment, the compound of formula 1 is isotopically labeled. Isotopes that can be used in the compounds of the present application include various isotopes of H, C, N, O, P, F, S, such as2H、3H、13C、14C、15N、18O、17O、31P、32P、35S、18F and36S。
in one embodiment, examples of the pharmaceutically acceptable salt of the compound of formula 1 or a deuteron thereof include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, thiocyanate, toluenesulfonate, undecanoate, sodium salt, calcium salt, potassium salt, ammonium salt, tetraethylammonium salt, methylammonium salt, dimethylammonium salt, and ethanolamine salt.
Use of compound of general formula 1, deuteron or pharmaceutically acceptable salt thereof
The application provides the use of a compound of formula 1 as defined above, a deutero-derivative thereof or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prevention of hepatitis b.
In one embodiment, the present application provides the use of a compound of general formula 1 as defined above, a deuterate thereof or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for reducing Hepatitis B Virus (HBV) load, HBsAg levels and/or HBeAg levels.
In one embodiment, the present application provides the use of a compound of formula 1 as defined above, a deuterate thereof or a pharmaceutically acceptable salt thereof in the preparation of a medicament for simultaneously reducing the Hepatitis B Virus (HBV) load, the level of HBsAg and the level of HBeAg.
In one embodiment, the present application provides the use of a compound of general formula 1 as defined above, a deuterate thereof or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for reducing HBsAg levels and/or HBeAg levels.
In one embodiment, the present application provides the use of a compound of formula 1 as defined above, a deuterate thereof or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for simultaneously reducing HBsAg levels and HBeAg levels.
In one embodiment, the present application provides the use of a compound of general formula 1 as defined above, a deuterogen thereof, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the clearance of HBsAg and/or HBeAg.
In one embodiment, the medicament further comprises one or more additional therapeutic or prophylactic agents. The one or more additional therapeutic or prophylactic agents can be any one or more additional therapeutic or prophylactic agents as described in the "additional therapeutic or prophylactic agents" section below.
In one embodiment, the additional therapeutic or prophylactic agent is selected from at least one of an interferon, a PEGylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog,
formula A
Figure BDA0002964027430000101
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, such as compounds of formula I:
Figure BDA0002964027430000111
wherein R is1Is hydroxy or C1-C3An alkanoyloxy group; r2To R5Is H; r6Is CF3(ii) a X is N, W is S, and Y is CH.
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In one embodiment, the medicament is administered by a route selected from the group consisting of: oral, rectal, nasal, pulmonary, topical, buccal and sublingual, vaginal, parenteral, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural, and the like. In one embodiment, the medicament is administered orally. In one embodiment, the medicament is administered by intravenous injection.
In one embodiment, the medicament is an oral formulation. In one embodiment, the medicament is in the form of a tablet or capsule.
In the medicament of the present application, the compound of formula 1, a deuterated compound thereof or a pharmaceutically acceptable salt thereof and another therapeutic or prophylactic agent may be formulated into one dosage form, or may be formulated into separate dosage forms, as a combination product for sequential or simultaneous administration.
Pharmaceutical composition
The present application also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1, a deutero-derivative thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier/excipient,
Figure BDA0002964027430000112
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5.
In one embodiment, with substituent (R) in formula 1nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure BDA0002964027430000121
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; denotes the site of binding to the rest of formula 1.
In one embodiment, the compound of formula 1 is aripiprazole having the following structure:
Figure BDA0002964027430000122
in one embodiment, the compound of formula 1 is a deuterated compound. The deuterated compound can replicate the activity of the original compound, improve the half-life of the compound and delay or slow down the metabolic process of the compound.
In one embodiment, examples of the pharmaceutically acceptable salt of the compound of formula 1 or a deuterate thereof described herein include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, thiocyanate, tosylate, undecanoate, sodium salt, calcium salt, potassium salt, ammonium salt, tetraethylammonium salt, methylammonium salt, dimethylammonium salt, and ethanolamine salt.
In one embodiment, the additional therapeutic or prophylactic agent that can be used in the pharmaceutical compositions described herein can be any one or more of the additional therapeutic or prophylactic agents described below in the section "additional therapeutic or prophylactic agents".
In one embodiment, the additional therapeutic or prophylactic agent that may be used in the pharmaceutical compositions described herein may be selected from at least one of an interferon, a pegylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog,
formula A
Figure BDA0002964027430000131
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, such as compounds of formula I as described below:
Figure BDA0002964027430000132
wherein R is1Is hydroxy or C1-C3An alkanoyloxy group; r2To R5Is H; r6Is CF3(ii) a X is N, W is S, and Y is CH.
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In one embodiment, the present application provides a pharmaceutical composition comprising aripiprazole and a pharmaceutically acceptable carrier.
In one embodiment, the present application provides a pharmaceutical composition comprising aripiprazole and a nucleoside analog, and a pharmaceutically acceptable carrier.
In one embodiment, the present application provides a pharmaceutical composition comprising aripiprazole, one or more nucleoside analogs selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide, and a pharmaceutically acceptable carrier.
In one embodiment, the present application provides a pharmaceutical composition comprising aripiprazole and entecavir, and a pharmaceutically acceptable carrier.
In one embodiment, the pharmaceutical composition described herein can be used for the treatment or prevention of hepatitis b.
In one embodiment, the pharmaceutical composition described herein is capable of reducing Hepatitis B Virus (HBV) load, HBsAg levels, and/or HBeAg levels.
In one embodiment, the pharmaceutical composition described herein is capable of simultaneously reducing Hepatitis B Virus (HBV) load, HBsAg levels, and HBeAg levels.
In one embodiment, the pharmaceutical composition described herein is capable of reducing HBsAg levels and/or HBeAg levels.
In one embodiment, the pharmaceutical composition described herein is capable of reducing HBsAg levels and HBeAg levels simultaneously.
In one embodiment, the pharmaceutical composition described herein is capable of clearing HBsAg and/or HBeAg.
In one embodiment, the pharmaceutical composition described herein is capable of clearing HBsAg and HBeAg simultaneously.
Therefore, the application also provides the application of the pharmaceutical composition in preparing a medicament for treating or preventing hepatitis B. In one embodiment, the present application provides the use of the pharmaceutical composition in the manufacture of a medicament for reducing Hepatitis B Virus (HBV) load, HBsAg levels, and/or HBeAg levels. In one embodiment, the present application provides the use of the pharmaceutical composition in the preparation of a medicament for simultaneously reducing the Hepatitis B Virus (HBV) load, the level of HBsAg and the level of HBeAg. In one embodiment, the present application provides the use of the pharmaceutical composition for the manufacture of a medicament for simultaneously reducing the level of HBsAg and the level of HBeAg. In one embodiment, the present application provides the use of the pharmaceutical composition in the manufacture of a medicament for the clearance of HBsAg and/or HBeAg. In one embodiment, the present application provides the use of the pharmaceutical composition for the manufacture of a medicament for simultaneous clearance of HBsAg and HBeAg.
In the pharmaceutical composition of the present application, the compound of formula 1, a deuterated compound thereof or a pharmaceutically acceptable salt thereof and another therapeutic or prophylactic agent may be formulated into one dosage form, or may be formulated into separate dosage forms, as a combination product for sequential or simultaneous administration.
Viral hepatitis
The etiological typing of viral hepatitis is currently recognized by five hepatitis viruses, namely hepatitis A virus, hepatitis B virus, hepatitis C virus, hepatitis D virus and hepatitis E virus, which are respectively written as HAV, HBV, HCV, HDV and HEV, and except the hepatitis B virus which is a DNA virus, the rest are RNA viruses.
Hepatitis b is an infectious disease mainly caused by hepatitis b virus, and is a liver disease. Clinically, it is manifested as anorexia, nausea, epigastric discomfort, liver pain, and asthenia. Some patients may have jaundice fever and hepatomegaly with impaired liver function. Some patients can become chronic, even develop cirrhosis of the liver, and a few can develop liver cancer.
The etiological agent of hepatitis b is hepatitis b virus, abbreviated as HBV, which is a DNA virus. The genome is a double-stranded, circular, incompletely closed DNA. The outermost layer of the virus is the outer membrane or coat membrane of the virus, the inner layer is the core part, and the nucleoprotein is the core antigen (HBcAg) and cannot be detected in the serum. Serum from HBsAg positive persons showed 3 particles visible under electron microscope: round and filamentous particles with a diameter of 22nm, and also less spherical particles with a diameter of 42 angstroms, also known as Dane's particles, are intact HBV particles.
The markers for hepatitis b were detected as follows: (ii) HBsAg and anti-HBs: HBsAg positive indicates that HBV is currently in the stage of infection, and anti-HBs positive for immunoprotective antibodies indicates that immunity to HBV has developed. The diagnosis basis of the chronic HBsAg carrier is that the chronic HBsAg carrier has no clinical symptoms and physical signs, the liver function is normal, and the HBsAg is continuously positive for more than 6 months. (vii) HBeAg and anti-HBe: HBeAg positive is an index of HBV active replication and strong infectivity, and the change of the detected serum from HBeAg positive to anti-HBe positive indicates that the disease is relieved and the infectivity is weakened. ③ HBcAg and anti-HBc: HBcAg positive indicates that there is a direct reaction of complete HBV particles, and the active replication of HBV is less clinically used due to the complex detection method. anti-HBc is a marker of HBV infection, and anti-HBc IgM positive indicates that in early stage of infection, there is virus replication in vivo. HBsAg, HBeAg and anti-HBc are all positive in chronic mild hepatitis B and HBsAg carriers, and have high infectivity index and are difficult to turn negative.
Additional therapeutic or prophylactic agents
In one embodiment, a pharmaceutical composition comprising a compound of formula 1, a deutero-compound thereof, or a pharmaceutically acceptable drug or pharmaceutical composition thereof described herein may further comprise one or more additional therapeutic or prophylactic agents.
In one embodiment, the additional therapeutic or prophylactic agent may be selected from at least one of an interferon, a pegylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog,
formula A
Figure BDA0002964027430000161
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, such as compounds of formula I as described below:
Figure BDA0002964027430000162
wherein R is1Is hydroxy or C1-C3An alkanoyloxy group; r2To R5Is H; r6Is CF3(ii) a X is N, W is S, and Y is CH.
In one embodiment, the additional therapeutic or prophylactic agent is selected from an interferon or a nucleoside analog.
In one embodiment, the additional therapeutic or prophylactic agent is selected from a nucleoside analog.
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In some embodiments, the additional therapeutic or prophylactic agent is selected from one or more of entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide, for example one selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide or at least two selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
Entecavir (Entecavir, ETV) is chemically known as 2-amino-1, 9-dihydro-9- [ (1S,3R,4S) -4-hydroxy-3- (hydroxymethyl) -2-methylenecyclopentane ] -6H-purin-6-one and has the following structural formula:
Figure BDA0002964027430000171
US patent US5206244A discloses entecavir and its use for the treatment of hepatitis b virus; a novel synthesis method of entecavir is disclosed in WO9809964a 1; WO0164421a1 discloses low dose entecavir solid formulations.
Entecavir is a highly effective antiviral agent, developed by schrobo corporation in the 20 th century for 90 generations, and has a strong anti-HBV effect. It can be phosphorylated to active triphosphate, which has a half-life in cells of 15 h. Entecavir triphosphate inhibits all three activities of the viral polymerase (reverse transcriptase) by competing with deoxyguanosine triphosphate, the natural substrate of HBV polymerase: (1) the start of HBV polymerase; (2) formation of a reverse transcribed negative strand of a pregenomic mRNA; (3) synthesis of HBV DNA plus strand.
The chemical name of the Tenofovir Disoproxil Fumarate (TDF) is (R) - [ [2- (6-amino-9H-purin-9-yl) -1-methylethoxy ] methyl ] phosphonic acid diisopropoxycarbonyl methyl ester fumarate, which is an ester precursor of Tenofovir, belongs to a novel nucleotide reverse transcriptase inhibitor and has the activity of inhibiting HBV virus.
TDF is another novel open-ring nucleoside phosphonate successfully developed by Gilidard company in the United states after Adefovir dipivoxil, is firstly marketed in the United states in 10 months in 2001, and is currently marketed in countries such as Europe, Australia and Canada.
TDF inhibits viral polymerase in vivo by competitively binding to a natural deoxyribose substrate and terminates DNA strand synthesis by insertion into DNA. The main action mechanism is that the tenofovir is hydrolyzed into tenofovir after being orally taken, the tenofovir is phosphorylated by cell kinase to generate a metabolite tenofovir diphosphate with pharmacological activity, the tenofovir diphosphate competes with 5 '-triphosphate deoxyadenosine monophosphate to participate in the synthesis of virus DNA, and after entering the virus DNA, the DNA is prevented from being prolonged due to the lack of 3' -OH groups, so that the replication of the virus is blocked. Clinical application shows that TDF has obvious curative effect on HBV virus and less toxic side effect, so that TDF has wide clinical application foreground.
Tenofovir Alafenamide (Tenofovir Alafenamide), a precursor drug of the new Nucleoside Reverse Transcriptase Inhibitor (NRTI) Tenofovir (Tenofovir) developed by Gilidard scientific, USA. Compared with the prior generation of similar anti-hepatitis B medicine tenofovir disoproxil TDF, the antiviral activity of tenofovir alafenamide is 10 times, the stability in blood plasma is 200 times, and the half-decay period is improved by 225 times. Compared with TDF, the tenofovir alafenamide only needs one tenth of TDF administration dosage to achieve the same antiviral curative effect as TDF. Therefore, the tenofovir alafenamide is used for preventing or/and treating Hepatitis B Virus (HBV) infection and has better curative effect, higher safety and lower drug resistance.
In some embodiments, the additional therapeutic or prophylactic agents that may be used in the medicaments or pharmaceutical compositions described herein may also include one or more additional other drugs for treating HBV infection, such as, but not limited to, 3-dioxygenase (IDO) inhibitors, antisense oligonucleotides targeted to viral mRNA, apolipoprotein a1 modulators, arginase inhibitors, B-and T-lymphocyte detoxification agent inhibitors, Bruton's Tyrosine Kinase (BTK) inhibitors, CCR2 chemokine antagonists, CD137 inhibitors, CD160 inhibitors, CD305 inhibitors, CD4 agonists and modulators, compounds targeted to HBcAg, compounds targeted to hepatitis B core antigen (HBcAg), covalently closed loop dna cccdna (cccdna) inhibitors, cyclophilin inhibitors, cytokines, cytotoxic T-lymphocyte-associated protein 4(ipi4) inhibitors, DNA polymerase inhibitors, endonuclease modulators, epigenetic modifiers, farnesoid X receptor agonists, gene modifiers or editors, HBsAg inhibitors, HBsAg secretion or assembly inhibitors, HBV antibodies, HBV DNA polymerase inhibitors, HBV replication inhibitors, HBV RNAse inhibitors, HBV vaccines, HBV virus entry inhibitors, HBx inhibitors, hepatitis B large envelope protein modulators, hepatitis B large envelope protein stimulators, hepatitis B structural protein modulators, hepatitis B surface antigen (HBsAg) inhibitors, hepatitis B surface antigen (HBsAg) secretion or assembly inhibitors, hepatitis B virus E antigen inhibitors, hepatitis B virus replication inhibitors, hepatitis virus structural protein inhibitors, HIV-1 reverse transcriptase inhibitors, hyaluronidase inhibitors, IAP inhibitors, IL-2 agonists, IL-7 agonists, immunoglobulin G modulators, immunomodulators, indoleamine-2, ribonucleotide reductase inhibitors, interferon agonists, interferon alpha 1 ligands, interferon alpha 2 ligands, interferon alpha 5 ligand modulators, interferon alpha ligands, interferon alpha ligand modulators, interferon alpha receptor ligands, interferon beta ligands, interferon receptor modulators, interleukin-2 ligands, ipi4 inhibitors, lysine demethylase inhibitors, histone demethylase inhibitors, KDM5 inhibitors, KDM1 inhibitors, killer lectin-like receptor subfamily G member 1 inhibitionAgents, lymphocyte activation gene 3 inhibitors, lymphotoxin beta receptor activators, microRNA (miRNA) gene therapy agents, Axl modulators, B7-H3 modulators, B7-H4 modulators, CD160 modulators, CD161 modulators, CD27 modulators, CD47 modulators, CD70 modulators, GITR modulators, HEVEM modulators, ICOS modulators, Mer modulators, NKG2A modulators, NKG2D modulators, OX40 modulators, SIRP alpha modulators, TIGIIT modulators, Tim-4 modulators, Tyro modulators, Na modulators, miRNA modulators, and methods of using the same+-taurate cotransporter polypeptide (NTCP) inhibitors, natural killer cell receptor 2B4 inhibitors, NOD2 gene stimulators, nucleoprotein inhibitors, nucleoprotein modulators, PD-1 inhibitors, PD-L1 inhibitors, PEG-interferon lambda, peptidyl-prolyl isomerase inhibitors, phosphatidylinositol-3 kinase (PI3K) inhibitors, recombinant Scavenger Receptor A (SRA) proteins, recombinant thymosin alpha-1, retinoic acid inducible gene 1 stimulators, reverse transcriptase inhibitors, ribonuclease inhibitors, RNA DNA polymerase inhibitors, short interfering RNA (siRNA), short synthetic hairpin RNA (sshRNA), SLC10A1 gene inhibitors, SMAC mimetics, Src tyrosine kinase inhibitors, interferon gene Stimulators (STING) agonists, NOD1 stimulators, T cell surface glycoprotein CD28 inhibitors, t cell surface glycoprotein CD8 modulators, thymosin agonists, thymosin alpha 1 ligands, Tim-3 inhibitors, TLR-3 agonists, TLR-7 agonists, TLR-9 agonists, TLR9 gene stimulators, toll-like receptor (TLR) modulators, viral ribonucleotide reductase inhibitors, zinc finger nucleases or synthetic nucleases (TALEN), and combinations thereof.
Route of administration
The medicament or pharmaceutical composition of the present application may be administered by any route suitable for the condition to be treated. Suitable routes include oral, rectal, nasal, pulmonary, topical, buccal and sublingual, vaginal and parenteral, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural and the like. One skilled in the art will appreciate that the particular route of administration may vary depending on, for example, the pharmaceutical dosage form, the condition of the recipient, etc.
In one embodiment, the medicament or pharmaceutical composition of the present application may be administered by intravenous injection.
One advantage of the medicaments or pharmaceutical compositions of the present application is that they are orally bioavailable and can be administered orally. Thus, in one embodiment, the medicament or pharmaceutical composition of the present application may be administered orally. In one embodiment, the medicament or pharmaceutical composition of the present application may be administered orally in the form of a tablet or capsule.
Pharmaceutical compositions or formulations and formulations of drugs
In certain embodiments, the compound of formula 1, a deuterate thereof or a pharmaceutically acceptable salt thereof is administered in a pharmaceutical composition. The pharmaceutical compositions of the present application may be formulated with conventional carriers and excipients, which will be selected in accordance with common practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form and, when used for delivery by non-oral administration, are generally isotonic. The Pharmaceutical compositions or medicaments or all formulations thereof described herein will optionally contain Excipients such as the vehicles described in the Handbook of Pharmaceutical Excipients (1986). Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextran, hydroxyalkyl cellulose, hydroxyalkyl methyl cellulose, stearic acid, and the like. The pH of the formulation ranges from about 3 to about 11, but is typically from about 7 to 10. In some embodiments, the pH of the formulation ranges from about 2 to about 5, but typically from about 3 to 4.
The formulations include those suitable for the aforementioned routes of administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations are commonly found in Remington's Pharmaceutical Sciences (Mack Publishing co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then shaping the product as necessary.
Formulations of the present application suitable for oral administration may exist as follows: discrete units, such as capsules or tablets, each containing a predetermined amount of active ingredient; a powder or granules; solutions or suspensions in aqueous or non-aqueous liquids; or an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
Tablets are made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by: the active ingredient in a free-flowing form such as a powder or granules is compressed in a suitable machine, optionally mixed with a binder, lubricant, inert diluent, preservative, surfactant or dispersing agent. Molded tablets may be prepared by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally formulated so as to provide sustained or controlled release of the active ingredient therefrom.
Formulations for oral administration may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
The pharmaceutical compositions or medicaments of the present application may also be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. The suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol, or as a lyophilized powder. Acceptable carriers and solvents that may be employed include water, ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Acceptable carriers and solvents that may be employed include water, ringer's solution, isotonic sodium chloride solution and hypertonic sodium chloride solution.
In the medicament or pharmaceutical composition of the present application, the compound of formula 1, its deuteroide or its pharmaceutically acceptable salt and another therapeutic or prophylactic agent may be formulated into one dosage form, or may be separately formulated into separate dosage forms, and administered sequentially or simultaneously as a combination product.
Definition of
As used herein, "C1-C3Alkyl "refers to a straight or branched chain aliphatic saturated hydrocarbon monovalent radical having 1 to 3 carbon atoms, and non-limiting examples thereof include methyl, ethyl, propyl, isopropyl, and the like.
As used herein, "C1-C3Alkoxy "refers to the group RO-where R is C, formed by the attachment of an alkyl group to the oxygen1-C3An alkyl group. C1-C3Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, and the like.
"halogen" as used herein refers to at least one of fluorine, chlorine, bromine and iodine.
As used herein, "therapeutically effective amount" or "effective amount" refers to an amount that is effective at a dose and for a period of time required to achieve the desired therapeutic result. A therapeutically effective amount will depend on the nature and severity of hepatitis b or its symptoms, the particular therapeutic agent, the overall condition of the recipient (height, weight, age, and physical condition), and like factors, and can be determined by standard clinical techniques known to those skilled in the art.
As used herein, "treating" may refer to, for example, alleviating symptoms, prolonging survival, improving quality of life, and the like. Treatment need not be a "cure". Treatment may also refer to functional cure and clearance of hepatitis b virus.
As used herein, "reducing the Hepatitis B Virus (HBV) load" refers to reducing the amount of hepatitis B virus DNA in the blood of a detectable patient.
As used herein, "reducing the level of HBsAg and/or HBeAg" refers to reducing the amount of hepatitis B virus HBsAg and/or HBeAg in the blood of a detectable patient. The amount of HBsAg and/or HBeAg is often closely related to a curative effect on hepatitis B function.
As used herein, "pharmaceutically acceptable" refers to a substance that does not affect the biological activity or properties of the compounds of the present application and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components included in the composition.
As used herein, "carrier" refers to a relatively non-toxic substance that facilitates the introduction of a compound of the present application into a cell or tissue.
Detailed Description
The following examples are for illustrative purposes only and are not intended to limit the present application. Additional objects, advantages and novel features of the present application will become apparent to one of ordinary skill in the art upon examination of the following examples.
Examples
Example 1: evaluation of in vitro anti-HBV Activity of Aripiprazole Using HepG2-NTCP cells
The compound preparation method comprises the following steps:
in the case of preparation of 20mM concentration, the volume of the solvent DMSO (. mu.l) is the mass of the sample (mg). times.purity ÷ molecular weight ÷ 20X 106
Aripiprazole is available from Shanghai ceramic Biotechnology, Inc. The control compound was entecavir (ETV, lot: P1214012; 99.0% purity) purchased from Shanghai Tantake Technology, Inc. The mother liquors of the above compounds were all at 20mM concentration and stored at-20 ℃.
HepG2-NTCP cells were purchased from Shanghai medicine Mingkude New drug development, Inc. The cell culture medium was DMEM medium (Gibco cat. No. 11960051) containing 10% fetal bovine serum (FBS, ExCell cat. No. FSP500), 500. mu.g/ml G418, 1% glutamine, 1% NEAA (non-essential amino acids), 1mM sodium pyruvate, 1% penicillin-streptavidin, and was mainly used for subculturing cells. The cell plating medium was DMEM medium (Gibco cat. No. 11960051) containing 2% fetal bovine serum (FBS, ExCell cat. No. FSP500), 500. mu.g/ml G418, 1% glutamine, 1% NEAA (non-essential amino acids), 1mM sodium pyruvate, 1% penicillin-streptomycin, and was mainly used for cell plating and drug exchange.
The main other reagents and viruses used in this example are shown in Table 1.
TABLE 1 Main Agents and viruses
Figure BDA0002964027430000231
Experimental protocol
Plating cells and compound treatment
On day 0, HepG2-NTCP in cell plating medium was seeded into 48-well plates (7.5X 10)4Individual cells/well).
On day 1, the medium was changed to cell plating medium containing 2% DMSO.
On day 2, cells were pretreated with a culture solution containing a predetermined concentration of the compound for 2 hours, and then HepG2-NTCP cells were infected with the D-type HBV virus, and a culture solution containing a predetermined concentration of the compound was added simultaneously with the infection. The test compound aripiprazole was set at 2 single drug concentrations: 20 and 10 μ M, and 1 combination concentration: 20 μ M +0.1nM ETV. Control compound ETV was set at 1 single drug concentration: 0.1 nM. At the same time, a blank containing only DMSO and no compound was set. And 2, performing a duplicate hole test.
Fresh cell plating medium containing compounds containing 2% DMSO was replaced once on days 3, 5, and 7.
On day 9, supernatants were collected and the collected cell supernatants were assayed for HBeAg and HBsAg levels by ELISA and HBV DNA levels by qPCR. See table 2 for experimental procedures.
Table 2: experimental procedure
Number of days Cell processing
0 Cell plating
2 Adding medicine for pretreatment for 2 hours, infecting virus (simultaneously adding medicine)
3 Treatment of compounds
5 Treatment of compounds
7 Treatment of compounds
9 Collecting cells, detecting HBV DNA, HBeAg and HBsAg
Sample detection
1) qPCR method for detecting HBV DNA content in cell culture supernatant
DNA was extracted from the cell culture supernatant according to the QIAamp 96DNA Blood Kit instructions. The content of HBV DNA was detected by qPCR method. And (3) PCR reaction: at 95 ℃ for 10 min; 95 ℃ for 15 sec; 60 ℃, 1min, 40 cycles.
2) ELISA method for detecting content of HBeAg and HBsAg in cell culture supernatant
The method refers to the kit specification, and the method is briefly described as follows: respectively adding 50 mu l of standard substance, sample and reference substance into a detection plate, then adding 50 mu l of enzyme conjugate into each hole, incubating for 60 minutes at 37 ℃, washing the plate with washing liquor, then sucking dry, then adding 50 mu l of premixed luminescent substrate, incubating for 10 minutes at room temperature in a dark place, and finally measuring the luminescent value by an enzyme-linked immunosorbent assay.
Data computation
HBV DNA inhibition (%) was (1-HBV copy number of sample of compound group/HBV copy number of DMSO control group) × 100%
HBsAg inhibition (%) was (1-HBsAg value of sample/DMSO control HBsAg value) × 100%
HBeAg inhibition (%) was (1-HBeAg value of sample/DMSO control HBeAg value). times.100%
Analysis of results
The results of the measurements are shown in tables 3-5 and FIGS. 1-3.
TABLE 3 HBV DNA inhibition by test Compounds
Figure BDA0002964027430000251
TABLE 4 HBsAg inhibition Rate of test Compounds
Figure BDA0002964027430000252
TABLE 5 HBeAg inhibition of test compounds
Figure BDA0002964027430000253
As shown in table 3 and fig. 1, aripiprazole has a significant inhibitory effect on HBV DNA. The aripiprazole of 10 μ M and 20 μ M shows obvious inhibition on HBV DNA in HepG2-NTCP cells, the inhibition rates respectively reach 49.63% and 45%, and the inhibition rate is obviously higher than 36.67% of the control compound ETV of 0.1 nM.
As shown in table 4 and fig. 2, aripiprazole has a significant inhibitory effect on HBsAg. The inhibition rates of 10 mu M and 20 mu M aripiprazole on HBsAg in HepG2-NTCP cells reach 65.89% and 41.25%, respectively; the inhibition rate of 20 mu M aripiprazole and 0.1nM ETV on HBsAg is 70.06%. Whereas the control compound ETV at 0.1nM inhibited HBsAg only 4.71%.
Compared with the condition that ETV has no obvious inhibition effect on the HBsAg, the aripiprazole has obvious inhibition effect on the HBsAg. Notably, the inhibitory effect of aripiprazole at 10 μ M on HBsAg was better than that of the high dose group (20 μ M); when the aripiprazole is used in combination with the ETV, the inhibition rate of the HBsAg is obviously improved compared with that when the aripiprazole and the ETV are used singly, and the synergistic effect is shown.
As shown in table 5 and fig. 3, aripiprazole has a significant inhibitory effect on HBeAg. The inhibition rates of 10 mu M and 20 mu M aripiprazole on HBeAg in HepG2-NTCP cells respectively reach 59.21% and 42.5%; the inhibition rate of 20 mu M aripiprazole and 0.1nM ETV on HBeAg reaches 66.88%. Whereas the 0.1nM control compound ETV showed no inhibitory effect on HBeAg.
Compared with the non-inhibition effect of ETV on HBeAg, the aripiprazole has a remarkable inhibition effect on HBeAg. Notably, the inhibitory effect of aripiprazole at 10 μ M on HBeAg was better than that of the high dose group (20 μ M); when the aripiprazole is used together with the ETV, the inhibition rate of the HBeAg is obviously improved compared with that when the aripiprazole and the ETV are used singly, and the synergistic effect is shown.
The test results show that aripiprazole can effectively reduce the HBV viral load, and can simultaneously reduce HBsAg and HBeAg to 65.89% and 59.21% under the condition of reducing the HBV DNA to 49.63%. While the control compound Entecavir (ETV) is only able to reduce HBV DNA as reported in the literature, and has little effect on reducing HBeAg and HBsAg.
It is worth noting that when the aripiprazole and the entecavir are jointly administered, the effects of reducing the HBsAg and the HBeAg are higher, the effects reach 70.06% and 66.88% respectively, the inhibition rate is obviously higher than that when the aripiprazole and the entecavir are singly used, and the synergistic effect is shown.
Therefore, compared with entecavir, aripiprazole can effectively reduce the HBV viral load, HBeAg and HBsAg, thereby hopefully eliminating hepatitis B virus and achieving functional cure. Particularly, when the aripiprazole is combined with a nucleoside analogue in the prior art, such as entecavir, which can reduce the HBV titer but can not reduce the HBsAg and HBeAg levels, the effect of eliminating hepatitis B virus can be better shown.
While the present application has been described with reference to particular embodiments, those skilled in the art will recognize that changes or modifications can be made to the described embodiments without departing from the spirit and scope of the present application, which is defined by the appended claims.

Claims (10)

1. Use of a compound of formula 1, a deutero-compound thereof, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating or preventing hepatitis b:
Figure FDA0002964027420000011
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5.
2. The use as claimed in claim 1, wherein in the general formula 1, there is a substituent (R)nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure FDA0002964027420000012
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; denotes the site of binding to the rest of formula 1.
3. The use according to claim 1, wherein the compound of formula 1 is the following compound:
Figure FDA0002964027420000021
4. the use of any one of claims 1-3, wherein the medicament is for reducing the hepatitis B viral load, HBsAg levels, and/or HBeAg levels.
5. The use of claim 4, wherein the medicament is for simultaneously reducing the hepatitis B viral load, HBsAg levels and HBeAg levels.
6. The use of any one of claims 1-5, wherein the medicament further comprises one or more additional therapeutic or prophylactic agents.
7. The use of claim 6, wherein the additional therapeutic or prophylactic agent is selected from at least one of an interferon, a PEGylated interferon, nitazoxanide or an analog thereof, a compound of formula A, or a nucleoside analog,
formula A
Figure FDA0002964027420000022
Preferably, the nucleoside analogue is selected from entecavir, tenofovir disoproxil fumarate and tenofovir alafenamide.
8. The use of any one of claims 1-7, wherein the medicament is administered by a route selected from the group consisting of: oral, rectal, nasal, pulmonary, topical, buccal and sublingual, vaginal, parenteral, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural, preferably the medicament is in an oral formulation, more preferably in the form of a tablet or capsule.
9. A pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1, a deuteride thereof, or a pharmaceutically acceptable salt thereof, one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier/excipient:
Figure FDA0002964027420000031
wherein each R is independently selected from halogen, deuterium, C1-C3Alkoxy radical, C1-C3Alkyl or nitro radicals, and
n is an integer of 0 to 5,
wherein the additional therapeutic or prophylactic agent is preferably selected from at least one of an interferon, a PEGylated interferon, nitazoxanide or an analogue thereof, a compound of formula A or a nucleoside analogue,
formula A
Figure FDA0002964027420000032
Wherein the nucleoside analogue is selected from entecavir, tenofovir disoproxil fumarate and tenofovir alafenamide.
10. The pharmaceutical composition of claim 9, wherein the compound of formula 1 has a substituent (R)nThe benzene ring group of (a) is selected from the group represented by the following structural formula:
Figure FDA0002964027420000033
wherein R is1Is selected from C1-C3An alkoxy group; r2And R3One of them is Cl and the other is Br; r4Is H or Cl; r5Is Cl or Br; r6Is methyl; represents a site of binding to the rest of formula 1, preferably said compound of formula 1 is:
Figure FDA0002964027420000041
CN202110245747.3A 2021-03-05 2021-03-05 Use of quinolones for treating or preventing hepatitis B Active CN114073702B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110245747.3A CN114073702B (en) 2021-03-05 2021-03-05 Use of quinolones for treating or preventing hepatitis B

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110245747.3A CN114073702B (en) 2021-03-05 2021-03-05 Use of quinolones for treating or preventing hepatitis B

Publications (2)

Publication Number Publication Date
CN114073702A true CN114073702A (en) 2022-02-22
CN114073702B CN114073702B (en) 2023-12-12

Family

ID=80283057

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110245747.3A Active CN114073702B (en) 2021-03-05 2021-03-05 Use of quinolones for treating or preventing hepatitis B

Country Status (1)

Country Link
CN (1) CN114073702B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012011917A1 (en) * 2010-07-23 2012-01-26 Activbiotics Pharma, Llc Administration of rifalazil to immunocompromised patients
CN104350142A (en) * 2012-04-18 2015-02-11 海莫希尔有限责任公司 In vitro model for pathological or physiologic conditions
WO2017023047A1 (en) * 2015-08-03 2017-02-09 성균관대학교산학협력단 Composition for preventing or treating inflammatory disease or cancer containing aripiprazole as an active ingredient
CN107921144A (en) * 2015-06-20 2018-04-17 杭州多禧生物科技有限公司 The auspicious statin analog of Australia and its conjugation conjugate with cell-binding molecules
WO2020092528A1 (en) * 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012011917A1 (en) * 2010-07-23 2012-01-26 Activbiotics Pharma, Llc Administration of rifalazil to immunocompromised patients
CN104350142A (en) * 2012-04-18 2015-02-11 海莫希尔有限责任公司 In vitro model for pathological or physiologic conditions
CN107921144A (en) * 2015-06-20 2018-04-17 杭州多禧生物科技有限公司 The auspicious statin analog of Australia and its conjugation conjugate with cell-binding molecules
WO2017023047A1 (en) * 2015-08-03 2017-02-09 성균관대학교산학협력단 Composition for preventing or treating inflammatory disease or cancer containing aripiprazole as an active ingredient
WO2020092528A1 (en) * 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity

Also Published As

Publication number Publication date
CN114073702B (en) 2023-12-12

Similar Documents

Publication Publication Date Title
CN112933085B (en) Application of compound in preparation of medicine for treating or preventing viral hepatitis
CN114073699A (en) Use of rapamycin for treating or preventing hepatitis B
CN114832023B (en) Application of herba Erodii seu Geranii effective component in treating or preventing viral hepatitis
CN114423423B (en) Pharmaceutical composition for treating or preventing viral hepatitis and application thereof
CN115944629A (en) Use of rapamycin for treating or preventing hepatitis B
CN114949216A (en) Use of PI3K and mTOR inhibitors for treating or preventing viral hepatitis
CN114073702B (en) Use of quinolones for treating or preventing hepatitis B
CN114099517B (en) Application of benzimidazole compound in treating or preventing hepatitis B
CN114903891B (en) Application of saquinavir in treatment or prevention of hepatitis B
CN114224888B (en) Application of lansoprazole in preparing medicine for treating or preventing viral hepatitis
CN114259492A (en) Application of nitazoxanide in treating hepatitis B
CN115212191A (en) Use of 3,7, 11-trimethyl-2, 6, 10-dodecatrien-1-ol for treating viral hepatitis
CN114053293A (en) Use of heterocyclic compounds for the treatment or prophylaxis of viral hepatitis
CN114209706A (en) Use of vitamin D analogues for treating or preventing viral hepatitis
WO2022179575A1 (en) Application of rapamycin in treatment or prevention of hepatitis b
CN114681439A (en) Application of fenretinide in preparation of medicine for treating or preventing viral hepatitis
CN114948949A (en) Use of HSP90 inhibitors for the treatment or prevention of viral hepatitis
CN113440504A (en) Application of dimercaptobutanedioic acid sodium in preparing medicine for treating or preventing viral hepatitis
CN115245569A (en) A pharmaceutical composition for the treatment of viral hepatitis
CN115337310A (en) Application of anti-inflammatory agent in preparation of medicine for treating or preventing viral hepatitis
CN114681614A (en) Application of benzoxazinone compounds in treating or preventing viral hepatitis
CN116173219A (en) Pharmaceutical composition for treating viral hepatitis
CN115429808A (en) Use of cyclooxygenase-2 inhibitor for treating or preventing viral hepatitis
CN114533743A (en) A pharmaceutical composition for treating or preventing viral hepatitis
CN115429789A (en) Use of phenolic compounds for treating or preventing viral hepatitis

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