CN114099517B - Application of benzimidazole compound in treating or preventing hepatitis B - Google Patents

Application of benzimidazole compound in treating or preventing hepatitis B Download PDF

Info

Publication number
CN114099517B
CN114099517B CN202110247078.3A CN202110247078A CN114099517B CN 114099517 B CN114099517 B CN 114099517B CN 202110247078 A CN202110247078 A CN 202110247078A CN 114099517 B CN114099517 B CN 114099517B
Authority
CN
China
Prior art keywords
compound
hepatitis
hbsag
hbv
formula
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
CN202110247078.3A
Other languages
Chinese (zh)
Other versions
CN114099517A (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 CN202110247078.3A priority Critical patent/CN114099517B/en
Publication of CN114099517A publication Critical patent/CN114099517A/en
Application granted granted Critical
Publication of CN114099517B publication Critical patent/CN114099517B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Abstract

The application provides application of benzimidazole compounds in treating or preventing hepatitis B. The application also provides application of the compound shown in the general formula 1, deuterated compound or pharmaceutically acceptable salt thereof in preparing medicaments 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 deuterated compound thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier.

Description

Application of benzimidazole compound in treating or preventing hepatitis B
Technical Field
The application relates to the field of anti-hepatitis B medicaments, in particular to application of benzimidazole compounds, deuterated compounds or pharmaceutically acceptable salts thereof in medicaments for treating or preventing hepatitis B, especially reducing the load of Hepatitis B Virus (HBV) and/or the level of HBsAg and/or the level of HBeAg.
Background
Human Hepatitis B Virus (HBV) infection is a significant public health problem worldwide. After acute hepatitis B virus infection, about 8% of the hepatitis B virus infection still develops into chronic hepatitis B infection, and persistent HBV infection can lead to liver cirrhosis and even liver cancer. The hepatitis B transmission path mainly comprises vertical transmission and horizontal transmission. Vertical transmission refers to maternal and infant transmission; horizontal transmission is primarily through the blood.
Hepatitis B is also a long-term treatment process, and the aim of the treatment is to inhibit or eliminate HBV to the greatest extent, reduce inflammation necrosis of liver cells and hepatic fibrosis, delay and prevent disease progression, reduce and prevent liver decompensation, cirrhosis, hepatocellular carcinoma and complications thereof, thereby improving quality of life and prolonging survival time.
There are many hepatitis b therapeutic drugs on the market at present, and antiviral treatment is mainly performed by using interferon or nucleoside analogues. For interferon, recombinant DNA leukocyte interferon (IFN-. Alpha.) can inhibit HBV replication. However, when interferon is used for treating hepatitis B, serious adverse reactions, including myelosuppression, influence on thyroid function, depression and the like are often accompanied.
Nucleoside analogs inhibit HBV production primarily by inhibiting reverse transcriptase activity during HBV replication, clinically useful drugs include: lamivudine, famciclovir, acyclovir, adefovir, entecavir, tenofovir, sodium phosphonoformate and the like, which have certain HBV inhibiting effect.
Although these reverse transcriptase inhibitors can effectively reduce HBV DNA level and control HBV level, they have no direct effect on HBV cccDNA and HBsAg clearance because the action target is RNA reverse transcription to DNA. Therefore, the single-drug treatment of nucleoside analogues has extremely low probability of seroconversion of HBsAg, and can not truly cure hepatitis B, and patients need to take medicines for a long time or even for a whole life.
Under the condition of taking the medicines for a long time, the problems of drug resistance, huge medical cost, serious side effects of the medicines and the like are heavy burden for hepatitis B patients. The key point is that at present, no medicine can completely remove viruses to cure hepatitis B. Therefore, there is an urgent need in the art to provide a new drug for treating hepatitis b, capable of eliminating HBsAg, and achieving a functional cure.
Disclosure of Invention
The application provides application of benzimidazole compounds shown in a general formula 1, deuterated compounds or pharmaceutically acceptable salts thereof in preventing or treating hepatitis B, wherein the compounds can reduce the load of Hepatitis B Virus (HBV), the level of HBsAg and/or the level of HBeAg, even can remove the HBsAg and the HBeAg, are expected to cure hepatitis B functionally and remove the hepatitis B virus.
In one aspect, the present application provides the use of a compound of formula 1, a deuterate thereof or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prophylaxis of hepatitis B, in particular for the reduction of Hepatitis B Virus (HBV) load and/or HBsAg and/or HBeAg levels, or even for the clearance of HBsAg and HBeAg,
wherein:
x represents a nitrogen atom or CH and,
R 1 Represents CH n F 3-n Wherein n is 1 or 2; hydroxy C 1 -C 6 Alkyl or NHR 6 Wherein R is 6 Is a hydrogen atom or COR, R is a hydrogen atom or C 1 -C 6 An alkyl group; c (C) 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group, an amino group,
R 2 represents optionally 1 to 4C 1 -C 6 Alkyl substituted morpholinyl, thiomorpholinyl and piperidinyl; optionally by 1-4 hydroxy groups C 1 -C 6 Alkyl substituted pyrrolidinyl; optionally by 1-2C 1 -C 6 Alkyl-substituted oxazolidinyl and tetrahydro-1, 4-thiazin-1-oxy-2-yl,
R 3 and R is 4 Each independently represents a hydrogen atom or C 1 -C 6 Alkyl group, and
R 5 represents a hydrogen atom, an amino group or a hydroxyl group.
In one embodiment, in formula 1R 1 Is difluoromethyl, hydroxymethyl, amino, formylamino or acetylamino, R 2 Morpholinyl or morpholinyl substituted with 1-3 methyl groups.
In one embodiment, the compound of formula 1 is ZSTK474:
in one embodiment, the deuterated of the compound of formula 1 is selected from:
in one embodiment, the pharmaceutically acceptable salts of the compounds of formula 1 or deuterated thereof include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane propionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinic acid, oxalate, thiocyanate, toluenesulfonate, undecanoate, sodium, calcium, potassium, ammonium, tetraethylammonium, methylammonium, dimethylammonium and ethanolamine salts.
In one embodiment, the medicament is for simultaneously reducing Hepatitis B Virus (HBV) loading, HBsAg, and HBeAg levels.
In one embodiment, the medicament is for simultaneously reducing HBsAg and HBeAg levels.
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,
a is a kind of
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 administered orally.
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 application also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier:
wherein:
x represents a nitrogen atom or CH and,
R 1 represents CH n F 3-n Wherein n is 1 or 2; hydroxy C 1 -C 6 Alkyl or NHR 6 Wherein R is 6 Is a hydrogen atom or COR, R is a hydrogen atom or C 1 -C 6 An alkyl group; c (C) 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group, an amino group,
R 2 represents optionally 1 to 4C 1 -C 6 Alkyl substituted morpholinyl, thiomorpholinyl and piperidinyl; optionally 1 to 4Hydroxy C 1 -C 6 Alkyl substituted pyrrolidinyl; optionally by 1-2C 1 -C 6 Alkyl-substituted oxazolidinyl and tetrahydro-1, 4-thiazin-1-oxy-2-yl,
R 3 and R is 4 Each independently represents a hydrogen atom or C 1 -C 6 Alkyl group, and
R 5 represents a hydrogen atom, an amino group or a hydroxyl group.
In one embodiment, the compound of formula 1 is ZSTK474:
in one embodiment, wherein the deuterated of the compound of formula 1 is selected from the group consisting of:
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:
A is a kind of
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In one embodiment, the application provides the use of a compound of formula 1, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing HBsAg and/or HBeAg levels or scavenging HBsAg and/or HBeAg.
In one embodiment, the pharmaceutically acceptable salts of the compounds of formula 1 or deuterated thereof include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane propionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinic acid, oxalate, thiocyanate, toluenesulfonate, undecanoate, sodium, calcium, potassium, ammonium, tetraethylammonium, methylammonium, dimethylammonium and ethanolamine salts.
In another embodiment, the compound of formula 1 is a deuterated compound. Deuterated compounds can replicate the activity of the original compound while increasing the half-life of the compound, delaying or slowing the metabolic process of the compound.
In another embodiment, the compound of formula 1 is isotopically labeled.
The technical scheme of the invention has the following beneficial effects:
1. the use of a compound of formula 1, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof for the treatment or prophylaxis of hepatitis b provides a novel hepatitis b treatment option.
2. The compound of the general formula 1, deuterated matters or pharmaceutically acceptable salts thereof can effectively reduce the capacity of Hepatitis B Virus (HBV), the HBsAg level and/or the HBeAg level, even remove the HBsAg and the HBeAg, and are expected to achieve the effect of curing the hepatitis B in a functional way.
3. The compound of the general formula 1, deuterated compound or pharmaceutically acceptable salt thereof is expected to clear hepatitis B virus, cure hepatitis B and avoid the pain of taking medicine for life under the condition of being combined with the existing nucleoside analogue medicine.
4. The compound of formula 1, its deuterated or pharmaceutically acceptable salt thereof can be combined with one or more additional therapeutic or prophylactic agents, in particular with agents that reduce viral titres but do not completely eliminate the virus, do not reduce HBsAg and/or HBeAg levels, eliminate hepatitis b virus from different aspects and have the potential for synergy, thus providing a broad line of thought for subsequent combination dosing designs.
Drawings
FIG. 1 shows the inhibition results of HBV DNA by co-administration of ZSTK474, entecavir (ETV) at different concentrations of 0.1nM, ZSTK474 at 10. Mu.M, and ETV at 0.1 nM.
FIG. 2 shows the inhibition results of HBsAg by various concentrations of ZSTK474, 0.1nM ETV, 10. Mu.M ZSTK474, and 0.1nM ETV co-administration.
FIG. 3 shows the inhibition results of HBeAg by various concentrations of ZSTK474, 0.1nM ETV, 10. Mu.M ZSTK474, and 0.1nM ETV co-administration.
Detailed Description
The inventors of the present application have unexpectedly found that the compound of formula 1, its deuterated product or its pharmaceutically acceptable salt according to the present application has potential activity in inhibiting hepatitis b virus, in particular has the effect of reducing the load of Hepatitis B Virus (HBV), HBsAg level and/or HBeAg level, even the effect of scavenging HBsAg and HBeAg, and can be used for preventing and treating hepatitis b.
A compound of formula 1, deuterated or pharmaceutically acceptable salt thereof
The inventors of the present application have found a group of benzimidazole compounds, their deuterates or pharmaceutically acceptable salts thereof, useful for the treatment or prevention of hepatitis b, in particular for the reduction of Hepatitis B Virus (HBV) load, HBsAg level and/or HBeAg level, in particular for the reduction of HBsAg and/or HBeAg level, said benzimidazole compounds having the following general formula 1:
Wherein:
x represents a nitrogen atom or CH and,
R 1 represents CH n F 3-n Wherein n is 1 or 2; hydroxy C 1 -C 6 Alkyl or NHR 6 Wherein R is 6 Is a hydrogen atomOr COR, R is a hydrogen atom or C 1 -C 6 An alkyl group; c (C) 1 -C 6 Alkyl or C 1 -C 6 An alkoxy group, an amino group,
R 2 represents optionally 1 to 4C 1 -C 6 Alkyl substituted morpholinyl, thiomorpholinyl and piperidinyl; optionally by 1-4 hydroxy groups C 1 -C 6 Alkyl substituted pyrrolidinyl; optionally by 1-2C 1 -C 6 Alkyl-substituted oxazolidinyl and tetrahydro-1, 4-thiazin-1-oxy-2-yl,
R 3 and R is 4 Each independently represents a hydrogen atom or C 1 -C 6 An alkyl group, a hydroxyl group,
and R is 5 Represents a hydrogen atom, an amino group or a hydroxyl group.
In one embodiment, R in formula 1 1 Is difluoromethyl, hydroxymethyl, amino, formylamino or acetylamino, R 2 Morpholinyl or morpholinyl substituted with 1-3 methyl groups.
In one embodiment, wherein the compound of formula 1 is ZSTK474:
ZSTK474 is known as a PI3K delta inhibitor. There is no report of its use in the treatment of hepatitis b.
In one embodiment, the compound of formula 1 is a deuterated compound. Deuterated 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.
The compounds of formula 1 may be deuterated at any desired atomic position. For example, the compounds of formula 1 may be on the benzene ring of the benzimidazole core and/or on the substituent R 1 、R 2 、R 3 、R 4 And R is 5 Deuterated at one, two or more or even all positions. In one embodiment, in the compound of formula 1, R 1 Can be deuterated C 1 -C 6 An alkyl group. In one embodiment, in the compound of formula 1, R 2 Can be optionally substituted with 1 to 4C 1 -C 6 Alkyl substituted deuterated morpholinyl, deuterated thiomorpholinyl, and deuterated piperidinyl; optionally by 1-4 hydroxy groups C 1 -C 6 Alkyl substituted deuterated pyrrolidinyl; optionally by 1-2C 1 -C 6 Alkyl substituted deuterated oxazolidinyl and deuterated tetrahydro-1, 4-thiazin-1-oxy-2-yl. In one embodiment, in the compound of formula 1, R 3 And R is 4 Each independently may be a deuterium atom. In one embodiment, in the compound of formula 1, R 5 And may be a deuterium atom. In one embodiment, in the compound of formula 1, R 1 、R 2 、R 3 、R 4 And R is 5 May be deuterated.
In one embodiment, the deuterated of the compound of formula 1 is selected from the following structural formulas:
deuterated compounds may be prepared by methods known in the art, for example, by exchanging hydrogen with deuterium, or by synthesizing the compound 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 application include various isotopes of H, C, N, O, P, F, S, such as 2 H、 3 H、 13 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and F 36 S。
In one embodiment, the pharmaceutically acceptable salts of the compounds of formula 1 or deuterated thereof include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane propionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinic acid, oxalate, thiocyanate, toluenesulfonate, undecanoate, sodium, calcium, potassium, ammonium, tetraethylammonium, methylammonium, dimethylammonium and ethanolamine salts.
Use of a compound of formula 1, deuterated or pharmaceutically acceptable salt thereof
The present application provides the use of a compound of formula 1, as defined above, a deuterated compound thereof or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment or prophylaxis of hepatitis b.
In one embodiment, the present application provides the use of a compound of formula 1, as defined above, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the Hepatitis B Virus (HBV) load, HBsAg level, and/or HBeAg level.
In one embodiment, the present application provides the use of a compound of formula 1, as defined above, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for simultaneously reducing the Hepatitis B Virus (HBV) load, HBsAg level, and HBeAg level.
In one embodiment, the present application provides the use of a compound of formula 1, as defined above, a deuterated compound 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 deuterated compound 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 formula 1, as defined above, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the clearance of HBsAg and/or HBeAg.
In one embodiment, the present application provides the use of a compound of formula 1, as defined above, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for simultaneous clearance of HBsAg and 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 may be any one or more additional therapeutic or prophylactic agents as described in the "additional therapeutic or prophylactic agent" 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,
a is a kind of
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, e.g., compounds of formula I:
wherein R is 1 Is hydroxy or C 1 -C 3 Alkanoyloxy; r is R 2 To R 5 Is H; r is R 6 Is CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the X is N, W is S, 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. 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 medicine of the present application, the compound of formula 1, its deuterated compound or its pharmaceutically acceptable salt and another therapeutic agent or prophylactic agent may be formulated into one dosage form, or may be formulated into separate dosage forms, respectively, for sequential or simultaneous administration as a combined product.
Pharmaceutical composition
The present application also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1 as defined above, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof, optionally one or more additional therapeutic or prophylactic agents, and a pharmaceutically acceptable carrier.
In one embodiment, R in formula 1 1 Is difluoromethyl, hydroxymethyl, amino, formylamino or acetylamino, R 2 Morpholinyl or morpholinyl substituted with 1-3 methyl groups.
In one embodiment, the compound of formula 1 is ZSTK474:
in one embodiment, the compound of formula 1 is a deuterated compound. Deuterated 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 deuterated of the compound of formula 1 is selected from:
in one embodiment, the compound of formula 1 may be isotopically labeled. Isotopes that can be used in the compounds of the applicationIncluding various isotopes of H, C, N, O, P, F, S, e.g 2 H、 3 H、 13 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and F 36 S。
In one embodiment, the pharmaceutically acceptable salts of the compounds of formula 1 or deuterated thereof include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentane propionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinic acid, oxalate, thiocyanate, toluenesulfonate, undecanoate, sodium, calcium, potassium, ammonium, tetraethylammonium, methylammonium, dimethylammonium and ethanolamine salts.
In one embodiment, the additional therapeutic or prophylactic agent may be any one or more additional therapeutic or prophylactic agents as described in the "additional therapeutic or prophylactic agent" section below.
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:
a is a kind of
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, e.g., compounds of formula I:
wherein R is 1 Is hydroxy or C 1 -C 3 Alkanoyloxy; r is R 2 To R 5 Is H; r is R 6 Is CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the X is N, W is S, Y is CH.
In one embodiment, the nucleoside analog is selected from entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
In one embodiment, the application provides a pharmaceutical composition comprising ZSTK474, its deuterates or pharmaceutically acceptable salts and nucleoside analogues thereof, and a pharmaceutically acceptable carrier.
In one embodiment, the present application provides a pharmaceutical composition comprising ZSTK474, its deuterates or pharmaceutically acceptable salts thereof and a nucleoside analog selected from entecavir, tenofovir disoproxil fumarate and tenofovir alafenamide, and a pharmaceutically acceptable carrier.
In the pharmaceutical composition of the present application, the compound of formula 1, its deuterated compound or its pharmaceutically acceptable salt and another therapeutic or prophylactic agent may be formulated into one dosage form, or may be formulated into separate dosage forms, respectively, for sequential or simultaneous administration as a combined product.
In one embodiment, the pharmaceutical composition of the present application can be used for treating or preventing hepatitis b.
In one embodiment, the pharmaceutical composition of the application is capable of reducing Hepatitis B Virus (HBV) load, HBsAg levels, and/or HBeAg levels.
In one embodiment, the pharmaceutical composition of the application is capable of simultaneously reducing Hepatitis B Virus (HBV) load, HBsAg levels, and HBeAg levels.
In one embodiment, the pharmaceutical composition of the application is capable of reducing HBsAg levels and/or HBeAg levels.
In one embodiment, the pharmaceutical composition of the application is capable of simultaneously reducing HBsAg levels and HBeAg levels.
In one embodiment, the pharmaceutical composition of the application is capable of scavenging HBsAg and/or HBeAg.
In one embodiment, the pharmaceutical composition of the application is capable of simultaneously scavenging HBsAg and HBeAg.
Therefore, the application also provides the application of the pharmaceutical composition in preparing medicines for treating or preventing hepatitis B. In one embodiment, the application provides the use of a pharmaceutical composition of the application in the manufacture of a medicament for reducing Hepatitis B Virus (HBV) load, HBsAg level and/or HBeAg level. In one embodiment, the application provides the use of a pharmaceutical composition of the application in the manufacture of a medicament for simultaneously reducing Hepatitis B Virus (HBV) load, HBsAg levels and HBeAg levels. In one embodiment, the application provides the use of a pharmaceutical composition according to the application for the manufacture of a medicament for simultaneously reducing HBsAg levels and HBeAg levels. In one embodiment, the application provides the use of a pharmaceutical composition according to the application for the manufacture of a medicament for the clearance of HBsAg and/or HBeAg. In one embodiment, the application provides the use of a pharmaceutical composition according to the application for the preparation of a medicament for simultaneous clearance of HBsAg and HBeAg.
Viral hepatitis
The etiology of viral hepatitis is typed, five hepatitis viruses, namely A, B, C, D and E, are currently recognized, and are respectively written as HAV, HBV, HCV, HDV, HEV, and the rest are RNA viruses except that the hepatitis B virus is DNA virus.
Hepatitis b is an infectious disease caused by hepatitis b virus and is mainly a liver lesion. Clinically, it is mainly manifested by anorexia, nausea, epigastric discomfort, pain in liver region and hypodynamia. Some patients may have jaundice fever and liver large with liver function impairment. Some patients may become chronicized, even develop cirrhosis, and a few may develop liver cancer.
The pathogen of viral hepatitis B is hepatitis B virus, abbreviated as HBV, and hepatitis B virus is DNA virus. The genome is double-stranded, circular, incompletely closed DNA. The outermost layer of the virus is the outer membrane or coating of the virus, the inner layer of which is the core, the nucleoprotein is the core antigen (HBcAg) and cannot be detected in serum. Serum from HBsAg positive subjects was visualized under electron microscopy for 3 particles: round and filiform particles of diameter 22nm, and also less spherical particles of diameter 42 angstroms, also known as Dane's particles, are intact HBV particles.
The markers for hepatitis b were detected as follows: (1) HBsAg and anti-HBs: HBsAg positive indicates that HBV is currently in the infectious stage, 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 signs and normal liver function, and the HBsAg is continuously positive for more than 6 months. (2) HBeAg with anti-HBe: HBeAg positive is an index of active replication and strong infectivity of HBV, and the change of the tested serum from HBeAg positive to anti-HBe positive indicates that the disease is relieved and the infectivity is weakened. (3) HBcAg with anti-HBc: HBcAg positive suggests that there is a direct reaction of intact HBV particles and that HBV active replication is rarely used clinically due to the complex detection method. anti-HBc is a marker of HBV infection, and positive IgM for anti-HBc suggests that there is viral replication in the body at the early stage of infection. Three positive HBsAg, HBeAg and anti-HBc in chronic mild hepatitis B and HBsAg carriers have high infectious index that is difficult to turn negative.
Additional therapeutic or prophylactic agents
In one embodiment, a pharmaceutical composition or a pharmaceutical composition comprising a compound of formula 1, a deuterated compound thereof, or a pharmaceutically acceptable salt thereof according to the application 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 represented by formula A or a nucleoside analog,
a is a kind of
In one embodiment, analogs of nitazoxanide include, but are not limited to, those disclosed in CN102803203B, such as the compounds of formula I as described below:
/>
wherein R is 1 Is hydroxy or C 1 -C 3 Alkanoyloxy; r is R 2 To R 5 Is H; r is R 6 Is CF (CF) 3 The method comprises the steps of carrying out a first treatment on the surface of the X is N, W is S, Y is CH.
In one embodiment, the additional therapeutic or prophylactic agent is selected from an interferon or a nucleoside analogue.
In one embodiment, the additional therapeutic or prophylactic agent is selected from nucleoside analogs.
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, from one of entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide, or from at least two of entecavir, tenofovir disoproxil fumarate, and tenofovir alafenamide.
Entecavir (ETV) is known under the chemical name 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:
U.S. patent No. 5206244a discloses entecavir and its use for the treatment of hepatitis b virus; a novel entecavir synthesis method is disclosed in WO9809964 A1; WO0164421A1 discloses low dose entecavir solid formulations.
Entecavir is a highly potent antiviral agent, developed by the american schdule company in the 90 th century, with a potent anti-HBV effect. It can be phosphorylated to an active triphosphate, which has a half-life of 15h in the cell. Entecavir triphosphates inhibit all three activities of the viral polymerase (reverse transcriptase) by competing with deoxyguanosine triphosphate, the natural substrate of HBV polymerase: (1) initiation of HBV polymerase; (2) formation of negative strand of pregenomic mRNA reverse transcription; (3) Synthesis of HBV DNA plus strand.
The chemical name of tenofovir disoproxil fumarate (Tenofovir disoproxil fumarate, TDF) is (R) - [ [2- (6-amino-9H-purin-9-yl) -1-methylethoxy ] methyl ] phosphonic acid diisopropyl oxycarbonyl methyl ester fumarate, is an ester precursor of tenofovir, belongs to a novel nucleotide reverse transcriptase inhibitor, and has the effect of inhibiting HBV virus activity.
TDF is another novel ring-opened nucleoside phosphonate that was successfully developed by the us gilid company following adefovir dipivoxil, and was first marketed in the us 10 months in 2001, and has been marketed in countries such as europe, australia and canada.
TDF inhibits viral polymerase in vivo by competitively binding to natural deoxyribose substrates and terminates DNA strand synthesis by insertion into DNA. The main action mechanism is that the tenofovir is hydrolyzed into tenofovir after oral administration, the tenofovir is phosphorylated by cell kinase, and a metabolite tenofovir diphosphate with pharmacological activity is generated, the metabolite tenofovir diphosphate competes with 5 '-deoxyadenosine triphosphate to participate in the synthesis of virus DNA, and after entering the virus DNA, the DNA is prolonged and blocked due to the lack of 3' -OH groups, so that the replication of the virus is blocked. Clinical application shows that TDF has obvious anti-HBV virus curative effect and small toxic side effect, thus having great clinical application prospect.
Tenofovir alafenamide (Tenofovir Alafenamide), a prodrug of Tenofovir (Tenofovir), a novel Nucleoside Reverse Transcriptase Inhibitor (NRTI) developed by the american gilid science company. Compared with the previous generation of anti-hepatitis B similar drug tenofovir disoproxil TDF, the antiviral activity of tenofovir alafenamide is 10 times of that of tenofovir alafenamide, the stability in blood plasma is 200 times of that of tenofovir alafenamide, and the half life is 225 times higher than that of tenofovir alafenamide. Compared with TDF, tenofovir alafenamide only needs one tenth of TDF administration dose, and can achieve the same antiviral 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, additional therapeutic or prophylactic agents that may be used in the medicaments or pharmaceutical compositions described herein may also include one or more additional agents for treating HBV infection, such as, but not limited to, 3-dioxygenase (IDO) inhibitors, antisense oligonucleotides targeting viral mRNA, apolipoprotein A1 modulators, arginase inhibitors, B-and T-lymphocyte antidote inhibitors, bruton Tyrosine Kinase (BTK) inhibitors, CCR2 chemokine antagonists, CD137 inhibitors, CD160 inhibitors, CD305 inhibitors, CD4 agonists and modulators, compounds targeting HBcAg, compounds targeting hepatitis B core antigen (HBcAg), covalently closed circular DNA (cccDNA) inhibitors, cyclophilin inhibitors, cytokines, cytotoxic T lymphocyte-associated protein 4 (ipi 4) inhibitor, DNA polymerase inhibitor, endonuclease modulator, epigenetic modifier, farnesoid X receptor agonist, gene modifier or edit, HBsAg inhibitor, HBsAg secretion or assembly inhibitor, HBV antibody, HBV DNA polymerase inhibitor, HBV replication inhibitor, HBV RNase inhibitor, HBV vaccine, HBV viral entry inhibitor, HBx inhibitor, hepatitis B large envelope protein modulator, hepatitis B large envelope protein stimulator, hepatitis B structural protein modulator, hepatitis B surface antigen (HBsAg) inhibitor, hepatitis B surface antigen (HBsAg) secretion or assembly inhibitor, hepatitis B virus E antigen inhibitor, hepatitis B virus replication inhibitor, hepatitis virus structural protein inhibitor, HIV-1 reverse transcriptase inhibitor, hyaluronidase inhibitor, IAP inhibitor, IL-2 agonist, IL-7 agonist, immunoglobulin G modulator, immunomodulator, indoleamine-2, ribonucleotide reductase inhibitor, interferon agonist, interferon alpha 1 ligand, interferon alpha 2 ligand, interferon alpha 5 ligand modulator, interferon alpha ligand modulator, interferon alpha receptor ligand Interferon beta ligand, interferon receptor modulator, interleukin-2 ligand, ipi4 inhibitor, lysine demethylase inhibitor, histone demethylase inhibitor, KDM5 inhibitor, KDM1 inhibitor, killer cell lectin-like receptor subfamily G member 1 inhibitor, lymphocyte activation gene 3 inhibitor, lymphotoxin beta receptor activator, microRNA (miRNA) gene therapeutic agent, axl modulator, B7-H3 modulator, B7-H4 modulator, CD160 modulator, CD161 modulator, CD27 modulator, CD47 modulator, CD70 modulator, GITR modulator, HEVEM modulator, ICOS modulator, mer modulator, NKG2A modulator, NKG2D modulator, OX40 modulator, SIRP alpha modulator, TIGIT modulator, tim-4 modulator, tyro modulator, na + -a taurine co-transporter polypeptide (NTCP) inhibitor, a natural killer cell receptor 2B4 inhibitor, a NOD2 gene stimulator, a nucleoprotein inhibitor, a nucleoprotein modulator, a PD-1 inhibitor, a PD-L1 inhibitor, a PEG-interferon lambda, a peptidyl prolyl isomerase inhibitor, a phosphatidylinositol-3 kinase (PI 3K) inhibitor, a recombinant Scavenger Receptor A (SRA) protein, a recombinant thymosin alpha-1, a retinoic acid inducible gene 1 stimulator, a reverse transcriptase inhibitor, a ribonuclease inhibitor, an RNA DNA polymerase inhibitor, a short interfering RNA (siRNA), a short synthetic hairpin RNA (sshRNA), a SLC10A1 gene inhibitor, a SMAC mimetic, a Src tyrosine kinase inhibitor, an interferon gene Stimulator (STING) agonist, a NOD1 stimulator, a T cell surface glycoprotein CD28 inhibitor, a T cell surface glycoprotein CD8 modulator, a thymosin agonist, a thymosin alpha 1 ligand, a Tim-3 inhibitor, a TLR-3 agonist, a TLR-7 agonist, a TLR-9 gene agonist, a toll 9 gene stimulator, a TLR-like receptor (TLR-like) or a nucleic acid inhibitor, and a nucleic acid-like combination thereof.
Route of administration
The medicament or pharmaceutical composition of the 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. Those skilled in the art will appreciate that the particular route of administration may vary depending upon, for example, the pharmaceutical dosage form, the condition of the recipient.
In one embodiment, the medicament or pharmaceutical composition of the 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 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 composition or formulation and formulation of a drug
In certain embodiments, the compound of formula 1, deuterated 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. The tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form and are generally isotonic when used for delivery by non-oral administration. The pharmaceutical compositions or medicaments of the application or all formulations thereof will optionally contain excipients, for example as described in "Handbook of Pharmaceutical Excipients" (1986). Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextran, hydroxyalkyl celluloses, hydroxyalkyl methylcellulose, 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 is typically from about 3 to 4.
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 generally 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 is composed of one or more accessory ingredients. In general, 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: discrete units, such as capsules or tablets, each containing a predetermined amount of the active ingredient; 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 free-flowing form, such as a powder or granules, is pressed in a suitable machine, optionally mixed with a binder, lubricant, inert diluent, preservative, surfactant or dispersant. Molded tablets may be made 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 slow or controlled release of the active ingredient therein.
Formulations for oral use 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, for example as a sterile injectable aqueous or oleaginous suspension. The suspensions may be formulated according to known techniques 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 can be used 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 can be used include water, ringer's solution, isotonic sodium chloride solution, and hypertonic sodium chloride solution.
Definition of the definition
"alkyl" as used herein refers to a straight or branched chain aliphatic saturated hydrocarbon monovalent radical having from 1 to 6 carbon atoms, and non-limiting examples thereof include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, and the like.
As used herein, "alkoxy" refers to the group RO-where R is C, formed by the linking of an alkyl group to oxygen 1 -C 6 An alkyl group. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy, tert-butoxy, pentyloxy, isopentyloxy, neopentyloxy, hexyloxy, and the like.
As used herein, "hydroxy" refers to an-OH group. Which may be protected by a protecting group, for example by an ester linkage, to form an alkanoyloxy group, for example an acetoxy protecting group. The protecting group can be removed under the action of in vivo esterase to form active hydroxyl.
As used herein, "halogen" refers to at least one of fluorine, chlorine, bromine, iodine.
As used herein, "amino" refers to substituted or unsubstituted-NH 2 A group. It can be protected by the formation of amide bonds and functions during metabolism by the formation of active amino groups by degradation of in vivo enzymes.
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 a desired therapeutic result. The therapeutically effective amount will depend on factors such as the nature and severity of the hepatitis b or symptoms thereof, the particular therapeutic agent, the general condition of the recipient (e.g., height, weight, age, and physical condition), and the like, and can be determined by standard clinical techniques known to those skilled in the art.
As used herein, "treatment" may refer to, for example, alleviation of symptoms, prolongation of survival, improvement of quality of life, and the like. The treatment need not be a "cure". Treatment may also refer to functional cure and clearance of hepatitis b virus.
As used herein, "reducing Hepatitis B Virus (HBV) load" refers to reducing the amount of HBV DNA in the blood of a patient that is detectable.
As used herein, "reducing HBsAg levels and/or HBeAg levels" refers to reducing the amount of HBsAg and/or HBeAg in the blood of a patient that is detectable. The amount of HBsAg and/or HBeAg is generally closely related to the functional cure of hepatitis B.
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 reaction or interacting in an undesirable manner with any of the components contained in the composition.
As used herein, "carrier" refers to a relatively non-toxic substance that facilitates the introduction of the compounds of the present application into cells or tissues.
Detailed Description
The following examples are for illustrative purposes only and are not limiting of the application. Additional objects, advantages and novel features of the present application will become apparent to those of ordinary skill in the art upon examination of the following examples.
Examples
Example 1: evaluation of ZSTK474 by HepG2-NTCP cells the in vitro anti-HBV active compound formulation method was as follows:
taking the case of preparing a concentration of 20mM, the volume of solvent DMSO (. Mu.l) =sample mass (mg). Times.purity/(molecular weight/(20X 10)) 6
ZSTK474 was purchased from shanghai Tao Su biochemical technology limited. The control compound was entecavir (ETV, lot number: P1214012;99.0% purity), available from Shanghai Teartan technologies Co., ltd. The mother liquor concentrations of the above compounds were 20mM and stored at-20 ℃.
HepG2-NTCP cells were purchased from Shanghai Minkangde New drug development Co. The cell subculture medium was DMEM medium (Gibco product number 11960051) containing 10% fetal bovine serum (FBS, ex cell product number FSP 500), 500 μg/ml G418, 1% glutamine, 1% NEAA (non-essential amino acids), 1mM sodium pyruvate, 1% penicillin-streptomycin, mainly for subculturing cells. Cell plating medium was DMEM medium (Gibco accession number 11960051) containing 2% fetal bovine serum (FBS, ex cell accession number FSP 500), 500 μg/ml G418, 1% glutamine, 1% NEAA (non-essential amino acids), 1mM sodium pyruvate, 1% penicillin-streptomycin, mainly for cell plating and drug replacement.
The other major reagents and viruses used in this example are shown in Table 1.
TABLE 1 major reagents and viruses
Experimental protocol
Cell spreading and compound treatment
On day 0, hepG2-NTCP in cell plating medium was plated into 48-well plates (7.5X10) 4 Individual cells/well).
On day 1, the cell plating medium was replaced with 2% DMSO.
On day 2, cells were pretreated by adding a culture solution with a predetermined concentration of the compound for 2 hours, and then HepG2-NTCP cells were infected with the D-type HBV virus, and the culture solution with the predetermined concentration of the compound was added simultaneously with the infection. 3 single drug concentrations were set for the test compound ZSTK 474: 10. 1 and 0.1 μm, and 1 combination concentration: 10 μM ZSTK474+0.1nM ETV. Control compound ETV set 1 single drug concentration: 0.1nM. A blank containing DMSO alone without compound was also set. 2, multi-hole testing.
Fresh 2% dmso-containing cell plating medium containing compound was changed once on days 3, 5 and 7.
On day 9, the supernatant was collected, and the collected cell supernatant was assayed for HBeAg and HBsAg levels by ELISA and HBV DNA levels by qPCR. See table 2 for experimental procedures.
Table 2: experimental procedure
Days (days) Cell treatment
0 Cell plating
2 Pretreatment with drug for 2 hours, infection with virus (simultaneous drug addition)
3 Treatment with a compound
5 Treatment with a compound
7 Treatment with a compound
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 by reference to QIAamp 96 DNA Blood Kit instructions. The HBV DNA content was detected by qPCR method. And (3) PCR reaction: 95 ℃ for 10min;95 ℃,15sec,60 ℃,1min,40 cycles.
2) ELISA method for detecting HBeAg and HBsAg content in cell culture supernatant
Methods referring to the kit instructions, the methods are briefly described as follows: respectively adding 50 μl of standard substance, sample and reference substance into the detection plate, adding 50 μl of enzyme conjugate into each well, incubating at 37deg.C for 60 min, washing the plate with washing solution, blotting, adding 50 μl of premixed luminescent substrate, incubating at room temperature in dark for 10min, and measuring the luminescence value by enzyme-labeled instrument.
Data computation
HBV DNA inhibition (%) = (HBV copy number of 1-compound group sample/HBV copy number of DMSO control group) ×100%
HBsAg inhibition (%) = (HBsAg value of 1-sample/HBsAg value of DMSO control) ×100%
HBeAg inhibition (%) = (HBeAg value of 1-sample/HBeAg value of DMSO control group) ×100%
Analysis of results
The results of the measurements are shown in tables 3-5 and FIGS. 1-3.
TABLE 3 HBV DNA inhibition of test compounds
TABLE 4 test of HBsAg inhibition by compounds
TABLE 5 HBeAg inhibition of test compounds
As shown in Table 3 and FIG. 1, ZSTK474 has an inhibitory effect on HBV DNA. The ZSTK474 with the concentration of 10 mu M shows obvious inhibition on HBV DNA in HepG2-NTCP cells, and the inhibition rate is 34.17%; an ETV of 0.1nM inhibited HBV DNA of 50.93%; the inhibition rate of 10. Mu.M ZSTK474+0.1nM ETV on HBV DNA was 58.14%. When ZSTK474 is combined with ETV, the inhibition rate of HBV DNA can be improved.
As shown in Table 4 and FIG. 2, ZSTK474 has a remarkable inhibitory effect on HBsAg. Inhibition rates of 10. Mu.M, 1. Mu.M and 0.1. Mu.M ZSTK474 on HBsAg in HepG2-NTCP cells were 27.71%, 13.35% and 7.14%, respectively; ETV of 0.1nM had no inhibitory effect on HBsAg; whereas the inhibition of HBsAg by ETV of 10. Mu.M ZSTK474+0.1nM reached 31.76%. Compared with the single use of ZSTK474 and ETV, the inhibition rate of HBsAg is obviously improved, which indicates that the two can have synergistic effect.
As shown in table 5 and fig. 3, ZSTK474 has a significant inhibitory effect on HBeAg. The inhibition rates of 10. Mu.M, 1. Mu.M and 0.1. Mu.M ZSTK474 on HBeAg in HepG2-NTCP cells were 45.21%, 20.27% and 4.3%, respectively; ETV of 0.1nM had no inhibitory effect on HBeAg; and the inhibition rate of ETV of 10 mu M ZSTK474+0.1nM to HBeAg reaches 46.74%. The inhibition rate of HBsAg is improved when ZSTK474 is combined with ETV compared with that of the ZSTK474 when the ZSTK474 and the ETV are used singly, so that the ZSTK474 and the ETV possibly have synergistic effect.
The test results show that ZSTK474 can effectively reduce HBV viral load, and can simultaneously reduce HBsAg and HBeAg to 27.71% and 45.21% respectively under the condition of reducing HBV DNA to 34.17%. Whereas the control compound Entecavir (ETV), as reported in the literature, is only able to reduce HBV DNA, with little effect on HBeAg and HBsAg.
Notably, when ZSTK474 and entecavir were co-administered, a higher HBsAg and HBeAg reduction was exhibited, reaching 31.76% and 46.74%, respectively.
Compared with entecavir, ZSTK474 can effectively reduce the levels of HBeAg and HBsAg, thereby being hopeful to remove hepatitis B virus and achieving functional cure. Particularly, when ZSTK474 is combined with nucleoside analogues in the prior art, such as entecavir, which can reduce HBV titer but cannot reduce HBsAg and HBeAg levels, better effect of eliminating hepatitis B virus can be shown.
Although the present application has been described with reference to particular embodiments, those skilled in the art will recognize that changes and modifications may be made to the embodiments without departing from the spirit and scope of the application, which is defined by the appended claims.

Claims (5)

1. The use of a compound or a pharmaceutically acceptable salt thereof as the sole active ingredient in the manufacture of a medicament for the treatment or prophylaxis of hepatitis B,
wherein the compound is:
2. the use of claim 1, wherein the medicament is for reducing HBsAg levels and HBeAg levels simultaneously.
3. The use of claim 1 or 2, wherein the medicament is administered by a route selected from the group consisting of: oral and parenteral.
4. The use of claim 3, wherein the medicament is an oral formulation.
5. The use according to claim 4, wherein the medicament is in the form of a tablet or capsule.
CN202110247078.3A 2021-03-05 2021-03-05 Application of benzimidazole compound in treating or preventing hepatitis B Active CN114099517B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110247078.3A CN114099517B (en) 2021-03-05 2021-03-05 Application of benzimidazole compound in treating or preventing hepatitis B

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110247078.3A CN114099517B (en) 2021-03-05 2021-03-05 Application of benzimidazole compound in treating or preventing hepatitis B

Publications (2)

Publication Number Publication Date
CN114099517A CN114099517A (en) 2022-03-01
CN114099517B true CN114099517B (en) 2023-12-15

Family

ID=80359501

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110247078.3A Active CN114099517B (en) 2021-03-05 2021-03-05 Application of benzimidazole compound in treating or preventing hepatitis B

Country Status (1)

Country Link
CN (1) CN114099517B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012118978A1 (en) * 2011-03-03 2012-09-07 The Regents Of The University Of Colorado, A Body Corporate Methods for treating oncovirus positive cancers
CN105821039A (en) * 2016-03-09 2016-08-03 李旭 Specific sgRNA combined with immunogene to inhibit HBV replication, expression vector thereof, and application of specific sgRNA and expression vector
WO2020092528A1 (en) * 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012118978A1 (en) * 2011-03-03 2012-09-07 The Regents Of The University Of Colorado, A Body Corporate Methods for treating oncovirus positive cancers
CN105821039A (en) * 2016-03-09 2016-08-03 李旭 Specific sgRNA combined with immunogene to inhibit HBV replication, expression vector thereof, and application of specific sgRNA and expression vector
WO2020092528A1 (en) * 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Benzimidazole derivative, BM601, a novel inhibitor of hepatitis B virus and HBsAg secretion;Yi-Bin Xu等;《Antiviral Res》;第107卷;6-15 *
HBV X蛋白通过激活PI3K/Akt信号通路诱导大鼠肾系膜细胞增殖;陈军宝;张洪英;雷辉;陈小梅;卢宏柱;;中国现代医学杂志(08);全文 *
Molecular basis of benzimidazole inhibitors to hepatitis C virus envelope glycoprotein;Reaz Uddin等;《Chem Biol Drug Des.》;第92卷;1638 - 1646 *

Also Published As

Publication number Publication date
CN114099517A (en) 2022-03-01

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
JP6770098B2 (en) Methods for treating hepatitis B virus infections using NS5A, NS5B or NS3 inhibitors
CN114423423B (en) Pharmaceutical composition for treating or preventing viral hepatitis and application thereof
CN115944629A (en) Use of rapamycin for treating or preventing hepatitis B
CN114099517B (en) Application of benzimidazole compound in treating or preventing hepatitis B
CN114073702B (en) Use of quinolones for treating or preventing hepatitis B
CN114949216A (en) Use of PI3K and mTOR inhibitors for treating or preventing viral hepatitis
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
WO2022179575A1 (en) Application of rapamycin in treatment or prevention of 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
CN116173219A (en) Pharmaceutical composition for treating viral hepatitis
CN114681439A (en) Application of fenretinide in preparation of medicine for treating or preventing viral hepatitis
CN115245569A (en) A pharmaceutical composition for the treatment of viral hepatitis
CN116407638A (en) Pharmaceutical composition for treating viral hepatitis
CN114209706A (en) Use of vitamin D analogues for treating or preventing viral hepatitis
CN114533743A (en) A pharmaceutical composition 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
CN114681614A (en) Application of benzoxazinone compounds in treating or preventing viral hepatitis
CN115671099A (en) A pharmaceutical composition for treating viral hepatitis
CN115518072A (en) A pharmaceutical composition for treating 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