WO2016110173A1 - New inhibitor for influenza virus neuraminidase and uses thereof - Google Patents

New inhibitor for influenza virus neuraminidase and uses thereof Download PDF

Info

Publication number
WO2016110173A1
WO2016110173A1 PCT/CN2015/097322 CN2015097322W WO2016110173A1 WO 2016110173 A1 WO2016110173 A1 WO 2016110173A1 CN 2015097322 W CN2015097322 W CN 2015097322W WO 2016110173 A1 WO2016110173 A1 WO 2016110173A1
Authority
WO
WIPO (PCT)
Prior art keywords
influenza virus
icariin
use according
virus
influenza
Prior art date
Application number
PCT/CN2015/097322
Other languages
French (fr)
Chinese (zh)
Inventor
庞海
刘爽
Original Assignee
吉林省帝医生物科技有限公司
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 吉林省帝医生物科技有限公司 filed Critical 吉林省帝医生物科技有限公司
Publication of WO2016110173A1 publication Critical patent/WO2016110173A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin

Definitions

  • the present invention relates to a novel inhibitor of influenza virus neuraminidase, which is specifically icariin and a derivative thereof.
  • the present invention also relates to the use of the novel inhibitor for the preparation of a medicament, a food, a health supplement for preventing and/or treating a disease caused by an influenza virus. It belongs to the fields of pharmacy and pharmacology.
  • Influenza virus is an RNA virus that causes influenza in humans, dogs, horses, pigs, mammals, and poultry. Taxonomically, the influenza virus belongs to the Orthomyxoviridae family, which causes acute upper respiratory tract infections and can spread rapidly through the air, causing periodic pandemics around the world. The virus was first known in 1933 by the Englishman Wilson Smith, known as H1N1. H stands for hemagglutinin; N stands for neuraminidase. The numbers represent different types. Human influenza viruses are classified into three types: A (A), B (B), and C (C), which are the pathogens of influenza (flu). Among them, the influenza A virus antigen is susceptible to mutation and has caused a worldwide pandemic many times.
  • influenza virus is spherical, and the newly isolated strain is mostly filamentous, with a diameter between 80 and 120 nanometers, and a filamentous influenza virus with a length of up to 400 nanometers.
  • Influenza virus structure can be divided into envelope, matrix protein and core three parts from the outside.
  • the core of the virus contains the genetic material that stores the virus information and the enzymes necessary to replicate the information.
  • the genetic material of influenza virus is single-stranded negative-strand RNA, abbreviated as ss-RNA. Ss-RNA is combined with nuclear protein (NP) and entangled into ribonucleoprotein body (RNP), which exists in extremely high density. In addition to ribonucleoproteins, there are RNA polymerases responsible for RNA transcription.
  • the RNA of influenza A virus consists of 8 segments.
  • the influenza virus gene consists of eight single-stranded RNA fragments, the NA, HA, NP, M, NS, PB1, PB2, and PA genes, respectively. They encode 10 proteins: Hemagglutinin HA, Neuraminidase NA, Matrix protein 1 M1, Nucleoprotein NP, and RNA-dependent RNA polymerase PB1 , PB2 and PA) Ion channel protein M2 and non-structural proteins NS1 and NS2.
  • the matrix protein constitutes the outer shell skeleton of the virus, and in fact, in addition to the matrix protein (M1), the membrane protein (M2).
  • the M2 protein has an ion (mainly Na+) channel and regulates the pH in the membrane, but in a small amount.
  • the matrix protein binds tightly to the outermost envelope of the virus, protecting the core of the virus and maintaining the spatial structure of the virus.
  • the matrix protein is distributed on the inner wall of the host cell membrane, and the shaped viral nucleocapsid recognizes the site containing the matrix protein on the host cell membrane, binds to form a viral structure, and is budding.
  • the envelope is a layer of phospholipid bilayer membrane wrapped outside the matrix protein. This membrane is derived from the cell membrane of the host. The mature influenza virus sprouts from the host cell, and the host cell membrane is wrapped in the body and then detached from the cell to be infected. Next target.
  • hemagglutinin and neuraminic acid there are two very important glycoproteins in the envelope: hemagglutinin and neuraminic acid. Enzyme. These two types of proteins protrude from the virus in vitro and are about 10 to 40 nanometers in length and are called spikes.
  • an influenza virus surface is distributed with 500 hemagglutinin spikes and 100 neuraminidase spikes.
  • the antigens of hemagglutinin and neuraminidase mutate which is the basis for distinguishing virus strain subtypes.
  • Hemagglutinin is columnar and can bind to receptors on the surface of red blood cells of animals such as humans, birds, and pig guinea pigs to cause blood clotting, so it is called hemagglutinin. After hemagglutinin proteolysis, it is divided into two parts: light chain and heavy chain. The latter can be combined with the sialic acid receptor on the host cell membrane. The former can assist the fusion of the viral envelope and the host cell membrane. Hemagglutinin plays an important role in the introduction of the virus into host cells. Hemagglutinin is immunogenic and anti-hemagglutinin antibodies neutralize influenza viruses.
  • Neuraminidase is a mushroom-like tetrameric glycoprotein with activity in hydrolyzing sialic acid. After the mature influenza virus leaves the host cell in a budding manner, the hemagglutinin on the surface of the virus is kept in contact with the host cell membrane via the sialic acid receptor, and the sialic acid needs to be hydrolyzed by the neuraminidase to cut off the last connection between the virus and the host cell. So that the virus can be released from the host cell smoothly, and then infect the next host cell. Therefore, neuraminidase has also become a target of influenza treatment drugs, and oseltamivir (Duffy) designed for this enzyme is one of the most famous anti-influenza drugs.
  • Duffy oseltamivir
  • H can be divided into 17 subtypes (H1 to H17), and N has 10 subtypes (N1 to N10).
  • H1N1, H2N2, and H3N2 mainly infect humans, and many other subtypes of natural hosts are a variety of birds and animals.
  • H5, H7 and H9 subtype strains are the most harmful to birds.
  • the avian flu virus does not infect animals other than birds and pigs.
  • Hong Kong reported for the first time 18 cases of H5N1 human infection with avian influenza, of which 6 died, causing widespread concern worldwide. Since 1997, there have been several incidents of avian influenza virus infection in the world.
  • the highly pathogenic avian influenza viruses such as H5N1, H7N7, H7N9, and H9N2, once mutated and have human-to-human transmission ability, will lead to human avian influenza epidemic, indicating that the avian influenza virus has great potential for humans. Threat.
  • influenza virus antigens are highly variable, conventional vaccines are not yet effective in preventing influenza outbreaks and epidemics, so anti-influenza drug research is of great significance in influenza treatment.
  • Anti-influenza viruses that have been and are being studied include amantadines, influenza virus neuraminidase inhibitors, influenza virus receptor blockers, and anti-influenza antisense oligonucleotides. Amantadine and rimantadine are commonly used clinical treatment drugs, but they are ineffective against influenza B, easily resistant and neurotoxic; influenza virus neuraminidase inhibitors such as oseltamivir are currently the most effective.
  • influenza virus receptor blockers have obvious antiviral effects
  • anti-influenza virus antisense oligonucleotides have been shown to specifically inhibit viral replication in vitro, and may become a new generation of anti-influenza specific drugs, but They are still in the research stage.
  • Icariin Chinese name is Epimedium
  • English name is Icariin
  • Icariln Ieariline
  • English chemical name is 3-[(6-Deoxy-alpha-L-mannopyranosyl)oxy]-7-(beta-D- Glucpyranosy-loxy)-5-hydroxy-2-(4-methoxyphenyl)-8-(3-methyl-2-buten-1-yl)-4H-1-benzopyran-4-one, chemical formula: C 33 H 40 O 15 .
  • Natural icariin is found in the genus E. pimedium, mainly distributed in the north temperate zone. There are 23 species of genus Epimedium in the world, and 13 species in China, such as: kinky E.
  • Chinese Patent Application CN1535976A describes in detail a method for extracting and purifying Epimedium from Epimedium. Extracted from the dried stems and leaves of Epimedium, the purified icariin is a pale yellow needle-like crystalline powder. Epimedium and its extract, icariin, have been used in Chinese patent medicines and health care products for many years. Even for thousands of years, no toxic or side effects have been found. It is safe for clinical use in Europe for the treatment of acute and chronic hepatitis. .
  • drugs and health supplements containing icariin and its derivatives can effectively inhibit the activity of influenza virus neuraminidase, thereby achieving the effect of inhibiting the proliferation of influenza virus, and can be used for prevention and/or prevention.
  • the treatment of influenza diseases achieves the object of the present invention.
  • an inhibitor of influenza virus neuraminidase in particular icariin and a derivative thereof. It can be used for the purpose of preventing and/or treating drugs, foods, health products or cosmetics for pathological symptoms or diseases of human or animals caused by influenza viruses.
  • the present invention provides a pharmaceutical composition comprising icariin and a derivative thereof, wherein the pharmaceutical composition contains at least one active ingredient of icariin and a derivative thereof. And the composition may also contain at least one other compound or a herbal ingredient.
  • the present invention also provides a preparation comprising icariin and a derivative thereof or a pharmaceutical composition thereof, and a process for producing the same.
  • a specific object of the present invention is to provide an inhibitor of influenza virus neuraminidase, a composition containing the same, for use in the preparation of a cold medicine or a health supplement for preventing and/or treating influenza virus.
  • the influenza diseases include influenza A, B and C viruses and their respective subtypes and various serotypes thereof, various gene mutants, and serotype variants. That is, H1 to H17, N1 to N10 of influenza A virus, and various gene mutants and serotype variants of M1, M2, PA, PB1, PB2, NS1, NS2; type B and C viruses and their respective Various subtypes and their various serotypes, various gene mutants, serotype variants.
  • icariin an influenza virus neuraminidase inhibitor, icariin and its derivatives, which were screened from natural Chinese herbal medicines without any side effects, to effectively prevent and/or treat diseases caused by influenza virus for the first time. , proved the mechanism of action of the drug. To achieve scientific use of medicine, there is a targeted. It is also the first time that this function of icariin has been discovered, filling the domestic and international gaps in the modernization of Chinese herbal medicine, which is conducive to the rehabilitation of patients.
  • Icariin inhibits influenza virus staining in MDCK cells by fluorescent antibody staining.
  • A MDCK cells infected with influenza virus H5N1, no icariin, all cells are infected with virus
  • B MDCK cells infected with influenza virus H5N1, medium containing 5 ⁇ mol of icariin, only a small number of cells Infected with virus
  • C MDCK cells infected with influenza virus H9N2, no icariin, all cells infected with virus
  • D MDCK cells infected with influenza virus H9N2, medium containing 5 ⁇ mol of icariin, only A small number of cells are infected with the virus.
  • Icariin inhibits influenza virus proliferation in MDCK cells by flow cytometry analysis.
  • A MDCK cells infected with influenza virus H5N1, no icariin, almost all cells are infected with the virus;
  • B MDCK cells infected with influenza virus H5N1, the medium contains 5 ⁇ mol of icariin, only a small amount The cells are infected with the virus;
  • C MDCK cells infected with influenza virus H9N2, no icariin, almost all cells are infected with the virus;
  • D MDCK cells infected with influenza virus H9N2, the medium contains 5 ⁇ mol of icariin Only a small number of cells are infected with the virus.
  • Icariin inhibits the replication of influenza virus in MDCK cells in real time with unlabeled cell function.
  • A MDCK cells infected with influenza virus H5N1
  • the line with triangles is the result of the culture plate wells of virus-infected cells without icariin; the line with dots is the control wells without virus infection; no dots
  • the lines of the triangles and the triangles were plate wells with a gradient of concentration of 1 ⁇ mol, 5 ⁇ mol, 10 ⁇ mol, 20 ⁇ mol, 50 ⁇ mol, 100 ⁇ mol, and 200 ⁇ mol of icariin.
  • the line with triangles is the result of the culture plate wells of virus-infected cells without icariin; the line with dots is the control wells without virus infection; no dots
  • the lines of the triangles and the triangles were plate wells with a gradient of concentration of 1 ⁇ mol, 5 ⁇ mol, 10 ⁇ mol, 20 ⁇ mol, 50 ⁇ mol, 100 ⁇ mol, and 200 ⁇ mol of icariin.
  • FIG. 5 Animal experiments in which icariin inhibits influenza virus.
  • A the average weight gain and decrease of each experimental group after inoculation of H5N1 in mice, that is, the total weight of 10 mice - the total weight of the mice in the first 10 experiments was ⁇ 10, and the solid line represents the infected mice and used 30 mg / The icariin was used to prevent changes in body weight of the experimental group mice (experimental group 2); the dotted line represents the change in body weight of the experimental group mice that did not use icariin after infection with the virus (experimental group 1).
  • the survival rate of the mouse the solid line represents the condition of the mice in the experimental group infected with the infected mice and using 30 mg / icariin (experimental group 2); the dotted line represents the mice infected with the virus without using Epimedium Survival of mice in the experimental group of glycosides (experimental group 1).
  • FIG. 6 Analysis of pathological tissue sections.
  • A normal mouse lung tissue section (experimental group 4).
  • B Normal mice were administered 30 mg/time of icariin in lung tissue sections (experimental group 3), and there were increased immune cells such as macrophages.
  • C lung tissue sections of experimental mice without icariin after infection with mice (experimental group 1), thrombosis in small blood vessels of the lungs, and a small amount of neutrophil degeneration and necrosis in the widened alveolar septum There is a small amount of lymphocyte infiltration around the bronchi.
  • D mice were infected and 30 mg/time of icariin was used to prevent lung tissue sections of experimental mice (experimental group 2), and there was a widening of alveolar septum with a small amount of macrophage infiltration.
  • the icariin is defined by the following formula (I):
  • Xa, Xb, Xc, Xd, Xe, Xf, Xg, Xh, Xi, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently an oxygen atom (O) or a sulfur atom (S );
  • Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh and Ri are independently a hydrogen atom (H), a 1-5Ry-substituted alkyl group (C 1 -C 10 ) or (C 1 -C 10 )-O- (C 1 -C 10 ), each alkyl group (C 1 -C 10 ) is substituted with 1-5 Ry, each Ry is Rq, or (C 2 -C 10 )alkyl, (C 3 -C 10 An alkyl group, (C 3 -C 10 )cycloalkylalkyl group, (C 8 -C 14 )bicycloalkylalkyl group, (C 8 -C 14 )tricycloalkylalkyl group, —(C 5 - C 10 ) cycloalkylalkyl, (C 3 -C 10 )cycloalkyl, (C 8 -C 14 )bicycloalkylalkyl, (C 8 -C
  • Xa, Xb, Xb, Xd, Xe, Xf, Xg, Xh, Xi and X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently oxygen atoms (O And Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ri, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H) , Y 1 , Y 2 and Y 3 are independently CH 3 .
  • Xa, Xb, Xb, Xd, Xe, Xf, Xg, Xh, Xi and X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently oxygen atoms ( O)
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H)
  • Y 1 , Y 2 and Y 3 are independently CH 3 .
  • icariin and its derivatives include their individual isomers, racemic or non-racemic mixtures of isomers, oxides, or pharmaceutically acceptable salts thereof And hydrates.
  • the icariin compound can be modified by a group such as amination, carboxylation, aldehyde formation, benzylation or the like to prepare epidemis with different activities and physical and chemical properties.
  • a glycoside compound derivative can be modified by a group such as amination, carboxylation, aldehyde formation, benzylation or the like to prepare epidemis with different activities and physical and chemical properties.
  • the icariin and its icariin derivative used in the present invention may be a natural or chemically synthesized and modified compound having the above formula (I) or (II).
  • the compound can be produced by a known method. Solvents, temperatures, pressures, and other reaction conditions can be readily selected by those skilled in the art. Starting materials are commercially available or can be readily prepared by one skilled in the art by known methods.
  • the natural icariin used in the present invention is extracted and purified from Epimedium plants.
  • the natural icariin plant is extracted and purified from the Epimedium plant of the Berberidaceae family.
  • the Epimedium plants include E. davidii, E. baojingense, E. icoroanum, and E. icoroanum.
  • E. fargesii E. elongatum, E. acuminatum, E. simplicifolium, E. brachyrrhizum, multi-flower Epimedium (E. multiflorum), E. enshiense, E. reticulatum, E. sagittatum, E. latisepalum ), E.chlorandrum, E.platypetalum, E. truncatum, E.
  • one skilled in the art screens to inhibit influenza virus neuraminidase activity by inhibiting influenza virus replication by performing enzymatic kinetic experiments using potential candidate compounds.
  • the ratio of the addition of the influenza virus to the icariin may be 1:0.00001-10000, preferably 1:0.0001-1000; more preferably 1 by weight. 0.001-500; most preferably 1:0.01-100 to achieve complete inhibition of influenza virus neuraminidase, thereby inhibiting replication of influenza virus.
  • the prepared icariin and its derivatives treat or prevent pathological symptoms or diseases caused by influenza virus.
  • influenza virus includes influenza virus A, B and C and their various subtypes, serotypes, genotypes, gene mutant viruses; the pathological symptoms or diseases are caused by various viruses of the above viruses Type one or more diseases caused by influenza syndrome.
  • treating refers to a therapeutic treatment and a preventive or preventive measure, wherein the subject's pathological symptoms or disorders are prevented or slowed down (weakened). If a therapeutic amount of icariin and its derivatives according to the methods described herein is accepted, the subject's symptoms are successfully "treated” and the subject shows that symptoms can be observed and/or determined One or more signs and symptoms are reduced and disappeared. It should also be understood that the various modes of treating or preventing a medical condition described herein are intended to mean "significant", which includes all treatment or prevention and less than all treatment or prevention, wherein certain biologically relevant or medically relevant results are achieved.
  • suitable in vitro assays or in vivo assays are performed to determine the effect of a therapeutic agent based on a particular icariin and its derivatives and whether its administration is suitable for treatment.
  • in vitro assays can be performed on representative cells involved in a subject's condition to determine whether a given therapeutic drug based on icariin and its derivatives has an expectation of the virus type. Effect. If a therapeutic amount of icariin and its derivatives according to the methods described herein is accepted, the subject's symptoms are successfully treated, ie, the subject shows that the above pathological symptoms can be observed and/or determined Or one or more signs and symptoms of the disease are reduced and disappeared. It should also be understood that the various modes of treating or preventing a medical condition described herein are intended to mean "significant", which includes all treatment or prevention and less than all treatment or prevention, wherein certain biologically relevant or medically relevant results are achieved.
  • the subject is administered a prophylactically and/or therapeutically effective amount of a medicament comprising icariin and a derivative thereof.
  • the candidate compounds used in the treatment can be tested in a suitable animal model system including, but not limited to, non-human primates (eg, baboons, orangutans, prior to testing the human subject). Monkeys; pet animals such as cats, dogs, snakes, etc; farm animals of pigs, horses, cows, goats, etc.; any animal of laboratory animals such as rats, mice, monkeys, rabbits, and the like.
  • icariin and a derivative thereof are administered to a subject suffering from or at risk of the above-mentioned pathological symptoms or diseases (a state in which the symptoms are readily available) in an attempt to improve the pathological symptoms Or one or more factors of the disease.
  • the term "effective amount” as used herein refers to an amount sufficient to achieve the desired therapeutic and/or prophylactic effect, for example, to cause prevention or alleviation of symptoms associated with pathological symptoms or diseases.
  • the amount of the composition administered to the subject will depend on the type and severity of the disease as well as the nature of the individual, such as the usual health conditions, age, sex, weight, and tolerance to the drug. The amount will also depend on the severity, severity and type of disease. Those skilled in the art will be able to determine the appropriate dosage based on these and other factors.
  • the composition may also be administered in combination with one or more additional therapeutic compounds.
  • the compounds of the invention can be administered to a subject having one or more signs or pathological symptoms of pathological symptoms.
  • therapeutically effective amount refers to an average level that minimizes the physiological effects of symptoms.
  • the dosage will be capable of preventing or reducing the severity or spread of the condition or indication being treated.
  • the correct dosage will depend on the circumstances, such as the condition being treated, the schedule of administration, whether the compound is administered alone or in combination with another therapeutic agent, the plasma half-life of the compound, and the overall health of the subject.
  • the subject can be administered by oral, topical, inhalation, nasal, rectal, transdermal or injection administration.
  • a daily dosage is prepared containing the compound disclosed herein.
  • the regimen is from about 0.1 mg/kg to about 10000 mg/kg body weight, preferably from about 0.1 mg/kg to about 1000 mg/kg body weight, more preferably from 0.1 mg/kg to about 500 mg/kg, and most preferably from 0.5 mg/kg to About 200 mg/kg body weight. It can be administered from 1 to 6 times a day, preferably 2 or 3 times a day.
  • the spacing may also be irregular, and those skilled in the art recognize that the optimal amount of the compound or its pharmaceutically acceptable salt and the interval between single administrations will depend on the nature and extent of the condition to be treated, the form, route and location of administration.
  • the point and the particular condition of the subject being treated are determined and the most preferred protocol can be determined using conventional techniques. It will also be understood by those skilled in the art that the optimal course of treatment, i.e., the amount of the compound administered daily or a pharmaceutically acceptable salt thereof administered over a given number of days, can be utilized by those skilled in the art using conventional therapeutic test methods. determine. In therapeutic applications, relatively high doses in relatively short time intervals are sometimes required until the course of the disease slows or terminates, and preferably until the subject shows a partial or complete improvement in the disease or pathology symptom. Therefore, the patient can perform a prophylactic administration method to the patient.
  • treating a subject with a therapeutically effective amount of a therapeutic control composition described herein can include a single treatment or a series of treatments.
  • the compounds of the invention may be administered with at least one other active ingredient other compound and/or a Chinese medicinal ingredient.
  • the compounds can be administered simultaneously as a separate formulation or in a unit dosage form, or sequentially.
  • the compound is ribavirin (1), interferon, thymosin alpha , transfer factor, zidovudine (ZDV), didanosine, Lamivudine, stavudine, zalcitabine, abacavir, Ribavirin, (Virazole), nevirapine, diarrhife Delavirdine, efavirenz, saquinavir, ritonavir, nelfinavir, indinavir, amprenavir, Lopinavir, King Kong Amantadine, Rimantadine, Zanamivir, Acyclovir, Valacyclovir, Idoxuridine, Vidarabine , Lamivudine, Famciclovir, Penciclovir, os
  • the plurality of therapeutic agents can be administered in any order or even simultaneously. If at the same time, the plurality of therapeutic agents can be provided in a single, uniform form. Or in multiple forms (for example, as a single tablet or as separate tablets or capsules, several different formulations of pills). One of the therapeutic agents may be provided in a multi-dose form, or Several may be provided as a multi-dose type. If not simultaneously, the time interval between multiple doses may range from greater than 0 weeks to less than about 1 week, less than about 2 weeks, less than about 4 weeks, less than about 2 months, A change in the range of less than about 4 months or even less than about half a year.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit comprising a separate predetermined amount of compound or in combination with other agents, the amount being calculated Sufficient to combine with a pharmaceutically acceptable diluent, carrier or vehicle to produce the desired effect.
  • a pharmaceutical composition containing icariin or a derivative thereof for treating and/or preventing a pathological symptom or disease associated with an improvement of a cold disease caused by an influenza virus is prepared. It is administered to a subject by oral administration, injection, inhalation, or the like. Therefore, the pharmaceutical composition of the present invention can be prepared into various dosage forms.
  • the composition for oral administration can be a capsule, a dropping pill, a gel, a tablet, a powder, a granule, a lozenge, an effervescent tablet, or a syrup.
  • composition for external use may be a smear-type drug, a cream, an external application, an ointment, a topical syrup, a liquid spray, an air spray, or the like.
  • composition for administration by inhalation may be a solution, a dispersion, a dry powder or the like; or a high-purity icariin compound may be extracted from the Chinese herbal medicine Epimedium, and a liposome-encapsulated nano icariin is prepared and prepared. Injectable preparations and sustained release pharmaceutical dosage forms.
  • the proportion of icariin or a derivative thereof is from 0.0% to 99.0%, preferably from 0.0% to 90%, more preferably 10% by weight based on the total weight of the entire preparation. -80%, most preferably 25-70%.
  • the above preparation may be a controlled preparation, including a sustained release tablet or a sustained release capsule.
  • the controlled release tablet comprises a core and a controlled release layer that encapsulates the core.
  • the core contains at least two active ingredients, at least one of which is icariin and a derivative thereof.
  • the core may further comprise at least one other active ingredient of Interferon, Thymosin alpha , Ribavirin, Amantadine, Rimantadine oseltamivir and Zanamivir.
  • the core may also contain at least one other traditional Chinese medicine active ingredient which is one or more combinations of traditional Chinese medicine ingredients such as Radix, Eucalyptus, Forsythia, Honeysuckle, Bupleurum, Andrographis paniculata.
  • the controlled release layer of the controlled release tablet comprises a copolymer of methacrylic acid and ethyl acetate, a copolymer of methacrylic acid and methyl methacrylate, ethyl cellulose, and triethyl citrate.
  • the ratio of the core and the controlled release layer of the package core is 1:0.01-100.
  • a method of preparing a sustained release tablet is provided.
  • the controlled release formulations produced provide pH independent release characteristics in the range of pH 1 to 10.0.
  • the oral preparation is exemplified, and the pharmaceutically acceptable excipients used include, but are not limited to, lubricants and co-solvents such as magnesium stearate, calcium stearate, zinc stearate, stearic acid.
  • the carrier particles may be crystals or spheres of lactose or sucrose; or composite spheres or granules, such as spheres of sugar formed by granulating sucrose with starch as a binder, calcium carbonate spheres or malt paste formed by starch as a binder fine.
  • the carrier particles can also be particles of any other pharmaceutically acceptable excipient such as hydroxypropylcellulose granules, guar gum granules, xanthan gum granules.
  • the carrier can take a wide variety of forms, and all such selections and amounts of adjustments are clearly within the abilities of those skilled in the art.
  • the compound of icariin or a derivative thereof according to the present invention or a composition thereof may comprise a liquid or a water-alcohol which is dissolved in a food or a health care product liquid, such as optionally dissolved in a flavoring. Class solution. It can be incorporated into a swallowable solid excipient, for example in the form of granules, pills, tablets, enteric coated tablets. It can also be placed in a liquid in a food or health supplement, optionally in the swallow capsule. For swallowing, a variety of oral composition embodiments can be employed, and in particular, can be oral composition embodiments of food addenda.
  • enteric coated tablets, colloidal capsules, gels, emulsions, tablets, capsules or solutions are prepared by conventional methods.
  • the active agent according to the invention may be incorporated into a food supplement or any other form of fortified food or health supplement, such as a food or health care product stick, or compressed or uncompressed Among the powders.
  • These powders can be diluted with water, diluted with water in soda, cheese products or soy derivatives, or can be incorporated into food or health care bars.
  • Example 1 Preparation of influenza virus neuraminidase and analysis of its effect on inhibition of influenza virus neuraminidase activity by icariin
  • H5N1 A/chicken/Guangdong/174/04 (H5N1)
  • H9N2 A/chicken/Guangdong/SS/94 (H9N2)
  • the inhibitory effect of the enzyme was also selected from the H9N2 virus strain to prepare a neuraminidase solution, and Triflurane X-100 (Sigma) was added to a concentration of 1% to prepare an influenza virus neuraminidase solution for assay of inhibitor activity.
  • the 98% pure icariin standard (ChromaDex, USA) was dissolved in 30% dimethyl nitrous oxide (Sigma) at a concentration of 10 mmol/L; Methylumbelliferyl-aDN-acetylneuramimic acid (MUNANA, sigma) was formulated into 20 Micromol/L concentration; oseltamivir (Oseltamivir, ChromaDex, USA) was formulated with water to a concentration of 10 mmol/L; 10 times buffer (325 mmol MES (Sigma), 40 mmol CaCl (Sigma), pH 6.5).
  • the enzyme activity assay reaction system was 100 microliters and was carried out in a black 96-well plate.
  • Influenza virus neuraminidase solution was pre-charged with different concentrations of icariin, and then added 3 ⁇ L of reaction buffer, 27 ⁇ L of water, which contained 1 ⁇ mol, 5 ⁇ mol, 50 ⁇ mol, 100 ⁇ mol, 200 ⁇ mol of icariin concentration gradient (final reaction system) concentration).
  • oseltamivir was set as a control experiment according to the above method, and the final reaction system concentration gradient was 1 ⁇ mol, 5 ⁇ mol, 50 ⁇ mol, 100 ⁇ mol, and 200 ⁇ mol.
  • influenza virus neuraminidase solution other components are added at the above concentration, the influenza virus neuraminidase solution is replaced with water
  • no icariin and oseltamivir are added (other ingredients are added according to the above concentration, Replenish the reaction volume with water)
  • add no icariin and oseltami but add 1% DMSO additional of other components at the above concentration, 1% DMSO is the final reaction system concentration).
  • Add the reaction before MUNANA The system is 40 ⁇ L, and each reaction system is shown in Table 1.
  • reaction After reacting at 37 ° C for 1 hour, all the reaction systems were added with 60 ⁇ l of 20 ⁇ mol/L MUNANA, reacted at 37 ° C for 1 hour, and terminated with 0.002 mol/L sodium hydroxide. reaction. Fluorescence intensity was measured using a super-microplate reader (Synergy HT, blo-tek, USA) at 360 excitation light and 460 emission light.
  • icariin Inhibition of influenza virus neuraminidase activity by icariin (Fig. 1) revealed that icariin inhibits influenza virus neuraminidase activity, which is nearly 40 times higher than oseltamivir, suggesting epimedium Glycoside can be used as a lead compound for the prevention and/or treatment of influenza virus diseases, and it is expected to develop a better drug for oseltamivir.
  • this experiment also confirmed that icariin can also inhibit the neuraminidase activity of different subtypes, which can inhibit the activity of N1 (H5N1) and N2 (H9N2) enzymes, suggesting that icariin may inhibit Neuraminidase activity of various subtypes.
  • Example 2 Inhibition of influenza virus by icariin at the in vitro cell level
  • MDCK Mesarby Canine Kidney cells (provided by Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences) were selected as influenza virus proliferating cells, and DMEM (Gibco) was used as the base medium for MDCK cells.
  • MDCK according to the method of Xie et al. (Yi Xie, James A. Schafer. Inhibition of ENaC by intracellular Cl - in an MDCK clone with high ENaC expression. Am J Physiol Renal Physiol.
  • Cell culture 190 mL of DMEM medium was added to 20 mL of fetal bovine serum, 2 mL of green chain double antibody, 7.5% NaHCO 3 to adjust the pH to 7.4. The above components were mixed into MDCK complete medium, 15 passages of cell bottles were added, and the amount of digestive juice was added. It is advisable to cover the cells just before, and gently put them in a 37 ° C incubator for about 10 minutes. When the cells are rounded and do not associate with each other, gently digest the digestive juice, add the culture solution, and repeatedly suck and blow until the cells are The cells were dissociated, and the cells were counted.
  • ⁇ 10 5 /mL cells were seeded in a new 25 mL culture flask, and cultured at 37 ° C in a 5% CO 2 atmosphere.
  • the influenza virus was inoculated into the 9-day-old chicken embryoal allantoic cavity, incubated at 35 ° C for 48-72 h, then the chicken embryo was placed in a refrigerator at 4 ° C overnight, the allantoic fluid was harvested the next day, centrifuged at 3000 rpm for 10 min, and the precipitate was discarded.
  • the serum was subjected to a blood coagulation test, and it was confirmed that the titer was greater than 1:64, and the supernatant was harvested and stored at -80 °C.
  • Influenza virus-infected cells were determined for virus titer: cultured cells were passaged and counted, transferred to a 6-well culture plate, and the cell density per well was adjusted to 1.0 ⁇ 10 5 /mL. Take H5N1 subtype, H9N2 (in order to try to inhibit the effect of icariin on other subtypes of neuraminidase, we also selected H9N2 strain) subtype influenza virus 1mL was added to the A and B cells, The same amount of physiological saline was added to the C well as a negative control. The final volume of each well culture system was adjusted to 5 mL, and A and B cells were incubated for 72 h for a total of 3 groups. Each group was repeated 5 times.
  • Cytopathic effect was observed under a microscope to determine the virus titer.
  • Hatakeyama et al. Shuji Hatakeyama, Yuko Sakai-Tagawa, Maki Kiso, Hideo Goto, Chiharu Kawakami, Keiko Mitamura, Norio Sugaya, Yasuo Suzuki and Yoshihiro Kawaoka.
  • Icariin for Influenza Virus Inhibition Icariin with 30% The dimethyl sigma was dissolved at a concentration of 1 mmol/mL and added to the DMEM cell culture medium. The experimental results of Example 1 were used to determine the concentration of 5 ⁇ mol for the detection of icariin inhibition virus replication assay. Icariin was replaced with PBS as a negative control to inhibit viral replication.
  • Flow cytometry (Beckman Coulter, USA), refer to the method of Liu et al. (Liu H, Zhang GL, Shen L, Zeng Z, Zhou BL, Liu CH, Nie G.
  • a mixture of virus and inhibitor was then added to a 96-well cell electronic assay plate in which MDCK cells had been grown, and placed in a 37 ° C incubator for 190-200 hours.
  • icariin concentration gradients ie, the medium containing 1 ⁇ mol, 5 ⁇ mol, 10 ⁇ mol, 20 ⁇ mol, 50 ⁇ mol, 100 ⁇ mol, 200 ⁇ mol, and a virus-negative control and icariin-free inoculated virus control .
  • Culture and detection were performed using a real-time label-free cell function meter, and data acquisition was performed every 15 minutes.
  • Example 2 Inhibition of influenza virus by icariin at the cellular level in vitro (Example 2), protection of MDCK cells from influenza virus destruction by viral pathogenicity and icariin of MDCK cells, including detection of infection using fluorescent antibodies Influenza-infected cells (Fig. 2), flow cytometry analysis (Fig. 3), and real-time unlabeled cell function analyzer analysis (Fig. 4) are all well demonstrated that icariin can inhibit influenza virus proliferation and protect it. Cells are protected from viruses.
  • Fig. 2 Influenza-infected cells
  • Fig. 3 flow cytometry analysis
  • Fig. 4 real-time unlabeled cell function analyzer analysis
  • neuraminidase virus strain may be a broad-spectrum inhibitor of influenza virus subtypes, serotypes, genotypes, and icariin can be used as a preventive and/or therapeutic agent.
  • the flu virus causes pathological symptoms or diseases of drugs, foods, and health products.
  • mice the Experimental Animal Center of Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, were reproduced.
  • the experimental animal quality license number was Heilongjiang-SYXK 2007-167.
  • mice The animal laboratory air is regularly ventilated and well lit, keeping the room temperature at the implementation room. Five animals were kept per cage, and the puffed feed specially made for mice was fed freely. Before the start of the experiment, observe the animals' feeding, activity and feces for a week. Only healthy mice were selected for the next experiment.
  • the acute toxicity test was used to evaluate the safety of icariin in order to provide a basis for the application of icariin.
  • the maximum tolerance method for acute toxicity test was used.
  • 20 healthy BALB/C mice weighing about 20 g were selected, male and female.
  • the clinically recommended route of administration is oral, and the equity experiment is administered by intragastric administration.
  • Icariin was prepared into a mushy paste with distilled water and administered to mice once a day (about 200 mg of icariin per mouse).
  • the animals were fasted overnight and given free access to water before gavage.
  • the stomach was given a normal diet, the symptoms of poisoning and death were observed and weighed once a week for 2 weeks. Under the same conditions, the same batch of mice were fed with normal food and observed as the control group.
  • the results of the experiment showed that all mice had any symptoms of poisoning, which proved that icariin is a non-toxic substance.
  • Icariin for the prevention of influenza virus infection in animals 6-week-old male BALB/c mice were selected, each group was inoculated with 50 ⁇ L of 102 LD50 H5N1 virus (since H9N2 is pathogenic to mice, so this H9N2) was not selected in the experiment (experimental group 1 and experimental group 2), PBS was used as a negative control (experimental group 4), and a control group (experimental group 3) administered with icariin alone was infected with the virus-infected mouse.
  • four experimental animal groups were set up, each group of 10 mice, each of which was orally administered with 1 mg, which was equivalent to oral administration of icariin 25 mg/kg body weight per day, twice a day for 14 days.
  • mice were weighed once a day, and the changes in body weight and survival rate were observed. Histopathological examination: Selecting the diseased, treatment group, oral administration of icariin and normal mouse lung tissue with 10% neutral-buffered formalin, then correcting and using different concentrations Dehydrated ethanol, soaked in xylene (Sima), embedded in paraffin (Paraffin, Sigma), cut into 3 micron thick slices, placed on glass slides, dewaxed continuously with alcohol, with hematoxylin (Hematoxylin) and Eosin (Sigma) staining were observed under light microscopy.
  • icariin inhibits influenza virus
  • Example 3 An animal experiment in which icariin inhibits influenza virus (Example 3) showed that icariin was able to protect against infection with highly pathogenic avian influenza virus (H5N1) (Fig. 5).
  • H5N1 highly pathogenic avian influenza virus
  • icariin can also promote immune cell infiltration in lung tissue and improve the immune protection of lung tissue (Fig. 6).
  • Icariin can be used in the preparation of icariin and its derivatives for the treatment or prevention of drugs, foods, and health products for pathological symptoms or diseases caused by influenza virus infection.
  • the icariin 0-side composition used in the examples of the present invention may be icariin for purchasing a drug, 200 g of icariin, and pulverized into a fine powder.
  • the component was passed through a 120 mesh sieve, weighed, and the amount of starch and magnesium stearate added in the above table were mixed and poured into a lower hopper.
  • the prepared pellets were filled with a capsule containing amyl saponin by using a hard capsule drug filling machine to fill 200 mg of icariin per two capsules, respectively.
  • Active agent Add amount (g) Icariin 300 Excipient Cassava starch 30 Galactose 20 Microcrystalline cellulose 30
  • Preparation process weigh 200g of icariin, pass 100 mesh sieve, tapioca starch, galactose, microcrystalline cellulose over 80 mesh sieve, mix evenly, add 100g of 6% PVP absolute ethanol solution to prepare granules, 60 °C Dry, 18 mesh sieve dry pellets, 2 g of magnesium stearate was added to the dry pellets.
  • Designing a controlled release formulation for oral administration in accordance with the present invention requires selection and evaluation of the in vitro release profile and timing of the formulation, making it optimal for obtaining the desired in vivo plasma profile, preferably once daily.
  • formulation principles for single unit matrix tablets have been investigated, namely providing formulations with different release profiles.
  • Active agent Add amount (g) Icariin 200 Excipient starch 200 Magnesium stearate 3 Adhesive glucose 2 Maltodextrin 3 Vinyl acetate copolymer 1.5
  • the icariin component or icariin used in the examples of the present invention may be a purchased drug, and 200 g of icariin is weighed and pulverized into a fine powder. The component was passed through a 120 mesh sieve, weighed, and the amount of starch, magnesium stearate, glucose, maltodextrin and vinyl acetate copolymer added in the above table was mixed, passed through a 250 mesh sieve, and then poured into a lower hopper. in.
  • pellets prepared by a and b were filled with a hard capsule drug filling machine according to the coated fine particles to prepare a capsule, that is, a mixed controlled release capsule preparation of the present invention containing 0.5 g of icariin was obtained.
  • Active agent Add amount (g) Icariin 200 Excipient Cassava starch 30 Galactose 20 Microcrystalline cellulose 30
  • Preparation process weigh icariin 200g over 100 mesh sieve, tapioca starch, galactose, microcrystalline cellulose over 80 mesh sieve, mix well, add 100g 6% PVP absolute ethanol solution to granulate, dry at 60 ° C 18 mesh sieved dry granules, and 2 g of magnesium stearate was added to the dry granules.
  • the extended release tablets of the present invention utilize a swellable and partially aggressive polymeric matrix. Based on the hypothetical matrix, the release profile can describe the in vitro release profile of the sample approximately at the square root of time. Obtaining the in vitro release profile that is not affected by the pH value is not affected by the pH value of the gastrointestinal tract when absorbed in the body, and is beneficial to avoid the difference in individual absorption due to the difference in pH of the gastrointestinal tract.
  • the polymer of the present invention constitutes an extended release matrix, and slowly releases icariin as a active ingredient and derivatives thereof and combinations thereof. After administration, after contact with the aqueous liquid, the polymer swells to form a viscous gel layer to regulate drug release.
  • the viscosity of the polymer preferably ranges from 180 to 110,000 mPa.s (apparent viscosity of a 2% aqueous solution at 20 ° C).
  • Example 6 or 7 of the present invention were simultaneously subjected to a release test at different pH values, and a release test curve was drawn based on the release test data. As shown in FIG. 7, the preparation of Example 7 can be seen. The sample has a consistent release profile in each pH medium and its release is virtually unaffected by the pH of the medium.
  • icariin can inhibit the activity of influenza virus neuraminidase, thereby inhibiting the replication of influenza virus, and can be used for the prevention and treatment of various diseases caused by influenza virus infection, food, health care products. Development.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A new inhibitor for influenza virus neuraminidase and uses thereof in the preparation for medicines, food, and health care products for the prevention and/or treatment of human or animal diseases caused by influenza viruses. The neuraminidase inhibitor for is icariin and derivatives thereof.

Description

一种流感病毒神经氨酸酶的新型抑制剂及其用途Novel inhibitor of influenza virus neuraminidase and use thereof 技术领域Technical field
本发明涉及一种流感病毒神经氨酸酶的新型抑制剂,其具体为淫羊藿苷及其衍生物。本发明还涉及该新型抑制剂在制备用于预防和/或治疗流感病毒引起的疾病的药物、食品、保健品中的用途。属于药学和药理学领域。The present invention relates to a novel inhibitor of influenza virus neuraminidase, which is specifically icariin and a derivative thereof. The present invention also relates to the use of the novel inhibitor for the preparation of a medicament, a food, a health supplement for preventing and/or treating a disease caused by an influenza virus. It belongs to the fields of pharmacy and pharmacology.
背景技术Background technique
以下描述利于读者理解。所提供的信息或引用的参考文献都不能被认为是本发明的现有技术。The following description is intended to help the reader understand. Neither the information provided nor the references cited are considered to be prior art to the present invention.
流行性感冒病毒(influenza virus),简称流感病毒,是一种能够引起人、狗、马、猪哺乳动物及禽类等流行性感冒的RNA病毒。在分类学上,流感病毒属于正粘病毒科(Orthomyxoviridae),它会造成急性上呼吸道感染,并可通过空气迅速传播,在世界各地会发生周期性大流行。该病毒最早是在1933年由英国人威尔逊·史密斯(Wilson Smith)认识的,称为H1N1。H代表血凝素;N代表神经氨酸酶。数字代表不同类型。人流感病毒分为甲(A)、乙(B)、丙(C)三型,是流行性感冒(流感)的病原体。其中甲型流感病毒抗原易发生变异,曾多次引起世界性大流行。Influenza virus, or influenza virus, is an RNA virus that causes influenza in humans, dogs, horses, pigs, mammals, and poultry. Taxonomically, the influenza virus belongs to the Orthomyxoviridae family, which causes acute upper respiratory tract infections and can spread rapidly through the air, causing periodic pandemics around the world. The virus was first known in 1933 by the Englishman Wilson Smith, known as H1N1. H stands for hemagglutinin; N stands for neuraminidase. The numbers represent different types. Human influenza viruses are classified into three types: A (A), B (B), and C (C), which are the pathogens of influenza (flu). Among them, the influenza A virus antigen is susceptible to mutation and has caused a worldwide pandemic many times.
流感病毒呈球形,新分离的毒株则多呈丝状,其直径在80至120纳米之间,丝状流感病毒的长度可达400纳米。流感病毒结构自外而内可分为包膜、基质蛋白以及核心三部分。病毒的核心包含了存贮病毒信息的遗传物质以及复制这些信息必须的酶。流感病毒的遗传物质是单股负链RNA,简写为ss-RNA,ss-RNA与核蛋白(NP)相结合,缠绕成核糖核蛋白体(RNP),以密度极高的形式存在。除了核糖核蛋白体,还有负责RNA转录的RNA多聚酶。甲型流感病毒的RNA由8个节段组成。流感病毒的基因由8个单链RNA片段组成,分别为NA、HA、NP、M、NS、PB1、PB2和PA基因。它们编码10种蛋白:膜蛋白血凝素(Hemagglutinin HA),神经氨酸(Neuraminidase NA),基质蛋白(Matrix protein1 M1,核蛋白(Nucleoprotein NP),3种RNA依赖聚(RNA-dependent RNA polymerase PB1、PB2和PA)离子通道蛋白(Ion channel protein M2)和非结构蛋白(Non-structural protein NS1和NS2)。The influenza virus is spherical, and the newly isolated strain is mostly filamentous, with a diameter between 80 and 120 nanometers, and a filamentous influenza virus with a length of up to 400 nanometers. Influenza virus structure can be divided into envelope, matrix protein and core three parts from the outside. The core of the virus contains the genetic material that stores the virus information and the enzymes necessary to replicate the information. The genetic material of influenza virus is single-stranded negative-strand RNA, abbreviated as ss-RNA. Ss-RNA is combined with nuclear protein (NP) and entangled into ribonucleoprotein body (RNP), which exists in extremely high density. In addition to ribonucleoproteins, there are RNA polymerases responsible for RNA transcription. The RNA of influenza A virus consists of 8 segments. The influenza virus gene consists of eight single-stranded RNA fragments, the NA, HA, NP, M, NS, PB1, PB2, and PA genes, respectively. They encode 10 proteins: Hemagglutinin HA, Neuraminidase NA, Matrix protein 1 M1, Nucleoprotein NP, and RNA-dependent RNA polymerase PB1 , PB2 and PA) Ion channel protein M2 and non-structural proteins NS1 and NS2.
基质蛋白构成了病毒的外壳骨架,实际上骨架中除了基质蛋白(M1)之外还有膜蛋白(M2)。M2蛋白具有离子(主要是Na+)通道和调节膜内PH值的作用,但数量很少。基质蛋白与病毒最外层的包膜紧密结合,起到保护病毒核心和维系病毒空间结构的作用。当流感病毒在宿主细胞内完成其繁殖之后,基质蛋白是分布在宿主细胞膜内壁上的,成型的病毒核衣壳能够识别宿主细胞膜上含有基质蛋白的部位,与之结合形成病毒结构,并以出芽的形式突出释放成熟病毒颗粒(Virion)。包膜是包裹在基质蛋白之外的一层磷脂双分子层膜,这层膜来源于宿主的细胞膜,成熟的流感病毒从宿主细胞出芽,将宿主的细胞膜包裹在自己身上之后脱离细胞,去感染下一个目标。包膜中除了磷脂分子之外,还有两种非常重要的糖蛋白:血凝素和神经氨酸 酶。这两类蛋白突出病毒体外,长度约为10至40纳米,被称作刺突。一般一个流感病毒表面会分布有500个血凝素刺突和100个神经氨酸酶刺突。在甲型流感病毒中血凝素和神经氨酸酶的抗原会发生变异,这是区分病毒毒株亚型的依据。The matrix protein constitutes the outer shell skeleton of the virus, and in fact, in addition to the matrix protein (M1), the membrane protein (M2). The M2 protein has an ion (mainly Na+) channel and regulates the pH in the membrane, but in a small amount. The matrix protein binds tightly to the outermost envelope of the virus, protecting the core of the virus and maintaining the spatial structure of the virus. After the influenza virus completes its reproduction in the host cell, the matrix protein is distributed on the inner wall of the host cell membrane, and the shaped viral nucleocapsid recognizes the site containing the matrix protein on the host cell membrane, binds to form a viral structure, and is budding. The form of the prominent release of mature virus particles (Virion). The envelope is a layer of phospholipid bilayer membrane wrapped outside the matrix protein. This membrane is derived from the cell membrane of the host. The mature influenza virus sprouts from the host cell, and the host cell membrane is wrapped in the body and then detached from the cell to be infected. Next target. In addition to phospholipid molecules, there are two very important glycoproteins in the envelope: hemagglutinin and neuraminic acid. Enzyme. These two types of proteins protrude from the virus in vitro and are about 10 to 40 nanometers in length and are called spikes. Generally, an influenza virus surface is distributed with 500 hemagglutinin spikes and 100 neuraminidase spikes. In the influenza A virus, the antigens of hemagglutinin and neuraminidase mutate, which is the basis for distinguishing virus strain subtypes.
血凝素(HA)呈柱状,能与人、鸟、猪豚鼠等动物红细胞表面的受体相结合引起凝血,故而被称作血凝素。血凝素蛋白水解后分为轻链和重链两部分,后者可以与宿主细胞膜上的唾液酸受体相结合,前者则可以协助病毒包膜与宿主细胞膜相互融合。血凝素在病毒导入宿主细胞的过程中扮演了重要角色。血凝素具有免疫原性,抗血凝素抗体可以中和流感病毒。神经氨酸酶(NA)是一个呈蘑菇状的四聚体糖蛋白,具有水解唾液酸的活性。成熟的流感病毒以出芽的方式脱离宿主细胞之后,病毒表面的血凝素会经由唾液酸受体与宿主细胞膜保持联系,需要由神经氨酸酶将唾液酸水解,切断病毒与宿主细胞的最后联系,使病毒能顺利从宿主细胞中释放,继而感染下一个宿主细胞。因此神经氨酸酶也成为流感治疗药物的一个作用靶点,针对此酶设计的奥司他韦(达菲类)是最著名的抗流感药物之一。H可分为17个亚型(H1~H17),N有10个亚型(N1~N10)。其中仅H1N1、H2N2、H3N2主要感染人类,其它许多亚型的自然宿主是多种禽类和动物。其中对禽类危害最大的为H5、H7和H9亚型毒株。一般情况下,禽流感病毒不会感染鸟类和猪以外的动物。但1997年香港首次报道发生18例H5N1人感染禽流感病例,其中6例死亡,引起全球广泛关注。1997年以后,世界上又先后几次发生了禽流感病毒感染人的事件。具有高致病性的H5N1、H7N7、H7N9、H9N2等禽流感病毒,一旦发生变异而具有人与人的传播能力,会导致人禽流感流行,预示着禽流感病毒对人类已具有很大的潜在威胁。Hemagglutinin (HA) is columnar and can bind to receptors on the surface of red blood cells of animals such as humans, birds, and pig guinea pigs to cause blood clotting, so it is called hemagglutinin. After hemagglutinin proteolysis, it is divided into two parts: light chain and heavy chain. The latter can be combined with the sialic acid receptor on the host cell membrane. The former can assist the fusion of the viral envelope and the host cell membrane. Hemagglutinin plays an important role in the introduction of the virus into host cells. Hemagglutinin is immunogenic and anti-hemagglutinin antibodies neutralize influenza viruses. Neuraminidase (NA) is a mushroom-like tetrameric glycoprotein with activity in hydrolyzing sialic acid. After the mature influenza virus leaves the host cell in a budding manner, the hemagglutinin on the surface of the virus is kept in contact with the host cell membrane via the sialic acid receptor, and the sialic acid needs to be hydrolyzed by the neuraminidase to cut off the last connection between the virus and the host cell. So that the virus can be released from the host cell smoothly, and then infect the next host cell. Therefore, neuraminidase has also become a target of influenza treatment drugs, and oseltamivir (Duffy) designed for this enzyme is one of the most famous anti-influenza drugs. H can be divided into 17 subtypes (H1 to H17), and N has 10 subtypes (N1 to N10). Among them, only H1N1, H2N2, and H3N2 mainly infect humans, and many other subtypes of natural hosts are a variety of birds and animals. Among them, H5, H7 and H9 subtype strains are the most harmful to birds. Under normal circumstances, the avian flu virus does not infect animals other than birds and pigs. However, in 1997, Hong Kong reported for the first time 18 cases of H5N1 human infection with avian influenza, of which 6 died, causing widespread concern worldwide. Since 1997, there have been several incidents of avian influenza virus infection in the world. The highly pathogenic avian influenza viruses such as H5N1, H7N7, H7N9, and H9N2, once mutated and have human-to-human transmission ability, will lead to human avian influenza epidemic, indicating that the avian influenza virus has great potential for humans. Threat.
由于流感病毒抗原极易变异,常规疫苗目前尚不能及时有效地预防流感暴发与流行,因此抗流感病毒药物研究在流感治疗中具有重要意义。已应用和正在研究的抗流感病毒药物有金刚烷胺类药物、流感病毒神经氨酸苷酶抑制剂、流感病毒受体阻断剂和抗流感病毒反义寡核苷酸等。金刚烷胺和金刚乙胺是常用临床治疗药物,但对B型流感无效,易产生耐药性并具有神经毒性;流感病毒神经氨酸苷酶抑制剂,如奥司他韦是目前最为有效的抗流感病毒一线药物;流感病毒受体阻滞剂具有明显的抗病毒作用、抗流感病毒反义寡核苷酸体外实验证明能特异性抑制病毒复制,有可能成为新一代抗流感特效药,但它们尚处于研究阶段。Because influenza virus antigens are highly variable, conventional vaccines are not yet effective in preventing influenza outbreaks and epidemics, so anti-influenza drug research is of great significance in influenza treatment. Anti-influenza viruses that have been and are being studied include amantadines, influenza virus neuraminidase inhibitors, influenza virus receptor blockers, and anti-influenza antisense oligonucleotides. Amantadine and rimantadine are commonly used clinical treatment drugs, but they are ineffective against influenza B, easily resistant and neurotoxic; influenza virus neuraminidase inhibitors such as oseltamivir are currently the most effective. First-line drugs against influenza virus; influenza virus receptor blockers have obvious antiviral effects, anti-influenza virus antisense oligonucleotides have been shown to specifically inhibit viral replication in vitro, and may become a new generation of anti-influenza specific drugs, but They are still in the research stage.
淫羊藿苷,中文别名为淫羊藿甙,英文名为Icariin、Icariln、Ieariline,英文化学名是3-[(6-Deoxy-alpha-L-mannopyranosyl)oxy]-7-(beta-D-glucopyranosy-loxy)-5-hydroxy-2-(4-methoxyphenyl)-8-(3-methyl-2-buten-1-yl)-4H-1-benzopyran-4-one,化学分子式是:C33H40O15。天然的淫羊藿苷存在于小檗科淫羊藿属(E.pimedium)植物中,主要分布于北温带,世界上常见的淫羊藿属植物有23种,中国有13种,例如:淫羊藿(E.brevicornum),箭叶淫羊藿(E.sagitatum),朝鲜淫羊藿(E.icoroanum)等。中国专利申请CN1535976A详细地描述了从淫羊藿属植物中提取纯化淫羊藿甙的方法。从淫羊藿属植物的干燥茎叶中提取,纯化后的淫羊藿苷为淡黄色针状结晶粉末。淫羊藿及它的提取物淫羊藿苷,在国内已经用于中成药和保健品,多年甚至上千年的使用没有发现任何毒副作用,在欧洲用于治疗急慢性肝炎,临床应用是安全的。Icariin, Chinese name is Epimedium, English name is Icariin, Icariln, Ieariline, English chemical name is 3-[(6-Deoxy-alpha-L-mannopyranosyl)oxy]-7-(beta-D- Glucpyranosy-loxy)-5-hydroxy-2-(4-methoxyphenyl)-8-(3-methyl-2-buten-1-yl)-4H-1-benzopyran-4-one, chemical formula: C 33 H 40 O 15 . Natural icariin is found in the genus E. pimedium, mainly distributed in the north temperate zone. There are 23 species of genus Epimedium in the world, and 13 species in China, such as: kinky E. brevicornum, E. sagitatum, E. icoroanum, etc. Chinese Patent Application CN1535976A describes in detail a method for extracting and purifying Epimedium from Epimedium. Extracted from the dried stems and leaves of Epimedium, the purified icariin is a pale yellow needle-like crystalline powder. Epimedium and its extract, icariin, have been used in Chinese patent medicines and health care products for many years. Even for thousands of years, no toxic or side effects have been found. It is safe for clinical use in Europe for the treatment of acute and chronic hepatitis. .
基于发明者的知识,没有报道显示淫羊藿甙流感病毒神经氨酸酶活性相关性,特别是淫羊藿苷及其衍生物在制备用于预防和/或治疗流感病毒引起的流行性感冒药物、保健品的用途。 Based on the knowledge of the inventors, there is no report showing the correlation of the activity of neuraminidase of the influenza virus, especially icariin and its derivatives in the preparation of influenza drugs for the prevention and/or treatment of influenza viruses. The use of health products.
出乎意料地,申请人发现含有淫羊藿苷及其衍生物的药物、保健品能够有效地抑制流感病毒神经氨酸酶活性,进而达到抑制流感病毒增值的药物效果,可用于预防和/或治疗流感疾病,实现本发明的目的。Unexpectedly, Applicants have found that drugs and health supplements containing icariin and its derivatives can effectively inhibit the activity of influenza virus neuraminidase, thereby achieving the effect of inhibiting the proliferation of influenza virus, and can be used for prevention and/or prevention. The treatment of influenza diseases achieves the object of the present invention.
发明内容Summary of the invention
在本发明的一个方面是提供一种流感病毒神经氨酸酶的抑制剂,具体为淫羊藿苷及其衍生物。其可以用于预防和/或治疗流感病毒引起的人或动物病理症状或疾病的药物、食品、保健品或化妆品中的用途。In one aspect of the invention, there is provided an inhibitor of influenza virus neuraminidase, in particular icariin and a derivative thereof. It can be used for the purpose of preventing and/or treating drugs, foods, health products or cosmetics for pathological symptoms or diseases of human or animals caused by influenza viruses.
在本发明的又一个方面,本发明还提供含有淫羊藿苷及其衍生物的药物组合物,其中,所述药物组合物含有至少一种活性成分为淫羊藿苷及其衍生物。且所述组合物还可以含有至少一种其它的化合物或中草药成分。In still another aspect of the present invention, the present invention provides a pharmaceutical composition comprising icariin and a derivative thereof, wherein the pharmaceutical composition contains at least one active ingredient of icariin and a derivative thereof. And the composition may also contain at least one other compound or a herbal ingredient.
在本发明的另一个方面,本发明还提供含有淫羊藿苷及其衍生物或其药物组合物的制剂及其制造方法。In another aspect of the invention, the present invention also provides a preparation comprising icariin and a derivative thereof or a pharmaceutical composition thereof, and a process for producing the same.
本发明的具体目的是提供流感病毒神经氨酸酶的抑制剂、含有该抑制剂的组合物在制备用于预防和/或治疗流感病毒引起的感冒药物、保健品中的用途。所述的流感疾病包括流感病毒的甲型、乙型和丙型病毒及其他们各自的各种亚型及其各种血清型、各种基因突变体、血清型变异体。即甲型流感病毒的H1~H17、N1~N10以及M1、M2、PA、PB1、PB2、NS1、NS2的各种基因突变体、血清型变异体;乙型和丙型病毒及其他们各自的各种亚型及其各种血清型、各种基因突变体、血清型变异体。A specific object of the present invention is to provide an inhibitor of influenza virus neuraminidase, a composition containing the same, for use in the preparation of a cold medicine or a health supplement for preventing and/or treating influenza virus. The influenza diseases include influenza A, B and C viruses and their respective subtypes and various serotypes thereof, various gene mutants, and serotype variants. That is, H1 to H17, N1 to N10 of influenza A virus, and various gene mutants and serotype variants of M1, M2, PA, PB1, PB2, NS1, NS2; type B and C viruses and their respective Various subtypes and their various serotypes, various gene mutants, serotype variants.
在国际上,发明人使用从天然中草药中筛选获得了一个无任何毒副作用的流感病毒神经氨酸酶抑制剂淫羊藿苷及其衍生物,首次有效地防止和/或治疗流感病毒导致的疾病,证明了该药的作用机理。做到科学用药,有的放矢。也是首次发现了淫羊藿苷的这种功能,填补了中草药现代化的国内、国际空白,利于患者康复。Internationally, the inventors used an influenza virus neuraminidase inhibitor, icariin and its derivatives, which were screened from natural Chinese herbal medicines without any side effects, to effectively prevent and/or treat diseases caused by influenza virus for the first time. , proved the mechanism of action of the drug. To achieve scientific use of medicine, there is a targeted. It is also the first time that this function of icariin has been discovered, filling the domestic and international gaps in the modernization of Chinese herbal medicine, which is conducive to the rehabilitation of patients.
附图说明DRAWINGS
图1、淫羊藿苷抑制流感病毒神经氨酸酶活性分析。Figure 1. Icariin inhibits influenza virus neuraminidase activity assay.
图2、淫羊藿苷抑制流感病毒在MDCK细胞复制荧光抗体染色法分析。A,为流感病毒H5N1传染的MDCK细胞,未加淫羊藿苷,所有细胞都感染了病毒;B,为流感病毒H5N1传染的MDCK细胞,培养基含有5μmol淫羊藿苷,仅有少量的细胞感染了病毒;C,为流感病毒H9N2传染的MDCK细胞,未加淫羊藿苷,所有细胞都感染了病毒;D,为流感病毒H9N2传染的MDCK细胞,培养基含有5μmol淫羊藿苷,仅有少量的细胞感染了病毒。Figure 2. Icariin inhibits influenza virus staining in MDCK cells by fluorescent antibody staining. A, MDCK cells infected with influenza virus H5N1, no icariin, all cells are infected with virus; B, MDCK cells infected with influenza virus H5N1, medium containing 5 μmol of icariin, only a small number of cells Infected with virus; C, MDCK cells infected with influenza virus H9N2, no icariin, all cells infected with virus; D, MDCK cells infected with influenza virus H9N2, medium containing 5 μmol of icariin, only A small number of cells are infected with the virus.
图3、淫羊藿苷抑制流感病毒在MDCK细胞增殖流式细胞仪分析。A,为流感病毒H5N1传染的MDCK细胞,未加淫羊藿苷,几乎所有细胞都感染了病毒;B,为流感病毒H5N1传染的MDCK细胞,培养基含有5μmol淫羊藿苷,仅有少量的细胞感染了病毒;C,为流感病毒H9N2传染的MDCK细胞,未加淫羊藿苷,几乎所有细胞都感染了病毒;D,为流感病毒H9N2传染的MDCK细胞,培养基含有5μmol淫羊藿苷,仅有少量的细胞感染了病毒。 Figure 3. Icariin inhibits influenza virus proliferation in MDCK cells by flow cytometry analysis. A, MDCK cells infected with influenza virus H5N1, no icariin, almost all cells are infected with the virus; B, MDCK cells infected with influenza virus H5N1, the medium contains 5μmol of icariin, only a small amount The cells are infected with the virus; C, MDCK cells infected with influenza virus H9N2, no icariin, almost all cells are infected with the virus; D, MDCK cells infected with influenza virus H9N2, the medium contains 5 μmol of icariin Only a small number of cells are infected with the virus.
图4、淫羊藿苷抑制流感病毒在MDCK细胞复制实时无标记细胞功能分仪。A,为流感病毒H5N1传染的MDCK细胞,带有三角的线为病毒感染细胞未加淫羊藿苷的培养板孔的结果;带有圆点的线为未感染病毒的对照孔;无圆点和三角的线为加有淫羊藿苷的1μmol、5μmol、10μmol、20μmol、50μmol、100μmol、200μmol浓度梯度的板孔。B,为流感病毒H9N2传染的MDCK细胞,带有三角的线为病毒感染细胞未加淫羊藿苷的培养板孔的结果;带有圆点的线为未感染病毒的对照孔;无圆点和三角的线为加有淫羊藿苷的1μmol、5μmol、10μmol、20μmol、50μmol、100μmol、200μmol浓度梯度的板孔。Figure 4. Icariin inhibits the replication of influenza virus in MDCK cells in real time with unlabeled cell function. A, MDCK cells infected with influenza virus H5N1, the line with triangles is the result of the culture plate wells of virus-infected cells without icariin; the line with dots is the control wells without virus infection; no dots The lines of the triangles and the triangles were plate wells with a gradient of concentration of 1 μmol, 5 μmol, 10 μmol, 20 μmol, 50 μmol, 100 μmol, and 200 μmol of icariin. B, MDCK cells infected with influenza virus H9N2, the line with triangles is the result of the culture plate wells of virus-infected cells without icariin; the line with dots is the control wells without virus infection; no dots The lines of the triangles and the triangles were plate wells with a gradient of concentration of 1 μmol, 5 μmol, 10 μmol, 20 μmol, 50 μmol, 100 μmol, and 200 μmol of icariin.
图5、淫羊藿苷抑制流感病毒的动物实验。A,为小鼠接种H5N1后各个实验组的平均体重增减情况,即10只小鼠总重量-实验起始10只小鼠总重量÷10的数值,实线代表感染小鼠并使用30mg/次淫羊藿苷预防患病的实验组小鼠体重变化(实验组2);虚线代表小鼠感染病毒后未使用淫羊藿苷的实验组小鼠体重变化(实验组1)。B,为小鼠生存率,实线代表感染小鼠并使用30mg/次淫羊藿苷预防患病的实验组小鼠情况(实验组2);虚线代表小鼠感染病毒后未使用淫羊藿苷的实验组小鼠生存情况(实验组1)。Figure 5. Animal experiments in which icariin inhibits influenza virus. A, the average weight gain and decrease of each experimental group after inoculation of H5N1 in mice, that is, the total weight of 10 mice - the total weight of the mice in the first 10 experiments was ÷ 10, and the solid line represents the infected mice and used 30 mg / The icariin was used to prevent changes in body weight of the experimental group mice (experimental group 2); the dotted line represents the change in body weight of the experimental group mice that did not use icariin after infection with the virus (experimental group 1). B, the survival rate of the mouse, the solid line represents the condition of the mice in the experimental group infected with the infected mice and using 30 mg / icariin (experimental group 2); the dotted line represents the mice infected with the virus without using Epimedium Survival of mice in the experimental group of glycosides (experimental group 1).
图6、病理组织切片分析。A,正常小鼠肺组织切片(实验组4)。B,正常小鼠投服30mg/次淫羊藿苷的肺组织切片(实验组3),见有巨噬细胞等免疫细胞增加。C,小鼠感染病毒后未使用淫羊藿苷的实验组小鼠肺组织切片(实验组1),肺脏小血管内有血栓形成,增宽的肺泡隔内有少量嗜中性粒细胞变性坏死,支气管周围有少量淋巴细胞浸润。D,感染小鼠并使用30mg/次淫羊藿苷预防患病的实验组小鼠肺组织切片(实验组2),见有肺泡隔增宽,伴有少量巨噬细胞浸润。Figure 6. Analysis of pathological tissue sections. A, normal mouse lung tissue section (experimental group 4). B. Normal mice were administered 30 mg/time of icariin in lung tissue sections (experimental group 3), and there were increased immune cells such as macrophages. C, lung tissue sections of experimental mice without icariin after infection with mice (experimental group 1), thrombosis in small blood vessels of the lungs, and a small amount of neutrophil degeneration and necrosis in the widened alveolar septum There is a small amount of lymphocyte infiltration around the bronchi. D, mice were infected and 30 mg/time of icariin was used to prevent lung tissue sections of experimental mice (experimental group 2), and there was a widening of alveolar septum with a small amount of macrophage infiltration.
图7、本发明实施例7制备的缓释片剂在不同pH值下的溶出曲线对比图。Figure 7. Comparison of dissolution profiles of sustained release tablets prepared in Example 7 of the present invention at different pH values.
具体实施方式detailed description
为了提供对本发明的实质性理解,在下文中以不同的详细程度描述了本发明的某些方面、模式、实施方式、变型和特征。在实施本发明的过程中,使用了分子生物学、蛋白质生物化学、细胞生物学、免疫学、微生物学和重组DNA方面的很多传统技术。这些技术是熟知的。In order to provide a substantial understanding of the present invention, certain aspects, modes, embodiments, variations and features of the present invention are described in various detail. In the practice of the present invention, many conventional techniques in molecular biology, protein biochemistry, cell biology, immunology, microbiology, and recombinant DNA have been used. These techniques are well known.
在一个实施方式中,制备由以下式(I)所定义的淫羊藿苷:In one embodiment, the icariin is defined by the following formula (I):
Figure PCTCN2015097322-appb-000001
Figure PCTCN2015097322-appb-000001
Figure PCTCN2015097322-appb-000002
Figure PCTCN2015097322-appb-000002
在另一个实施方式中,制备由以下式(II)所定义的淫羊藿苷衍生物:In another embodiment, the icariin derivative defined by the following formula (II) is prepared:
Figure PCTCN2015097322-appb-000003
Figure PCTCN2015097322-appb-000003
其中,among them,
Xa、Xb、Xc、Xd、Xe、Xf、Xg、Xh、Xi、X1、X2、X3、X4、X5、X6和X7独立为氧原子(O)或硫原子(S);Xa, Xb, Xc, Xd, Xe, Xf, Xg, Xh, Xi, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently an oxygen atom (O) or a sulfur atom (S );
Ra、Rb、Rc、Rd、Re、Rf、Rg、Rh和Ri独立为氢原子(H)、1-5Ry替代的烷基(C1-C10)或(C1-C10)-O-(C1-C10)、每一个烷基(C1-C10)用1-5个Ry取代,每一个Ry是Rq、或(C2-C10)烷基、(C3-C10)烷基、(C3-C10)环烷基烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、—(C5-C10)环烷基烷基、(C3-C10)环烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、苯基、萘基、(C14)芳香基团,它们的每一个可以用一个或更多的Rz取代,每一个Rz是(C1-C6)烷基、(C2–C6)烯基、(C1–C6)炔基、(C3–C8)环烷基、(C6-C8)环烯基、苯基、3-到5-元)杂环、C(卤素)3或CH(卤素)2;R1、R2、R2、R4、R5、R6、R7和R8独立为氢原子(H),或(C1-C10)烷基、(C2-C10)烯基、(C2-C10)炔基、(C3-C10)环烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、苯基、萘基、(C14)芳香基团,它们的每一个可以是非取代的、一个或更多的Rz或Rq取代的、一个或更多的带有1个以上Rz取代的Rq替换,每一个Rq可以是CN、OH、卤素、N3、NO2、N(Rz)2、═NR z、CH═NRz、NRz OH、ORz、CORz、C(O)ORz、OC(O)Rz、OC(O)ORz、SRz、S(O)Rz或S(O)2Rz;Y1、Y2和Y3独立为CH3、(C1-C12)烷基、也可以是用Ry组合取代的(C1-C12)烷基,每一个(C1-C12)烷基可以用1-5个R y取代或一个平衡离子取代。Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh and Ri are independently a hydrogen atom (H), a 1-5Ry-substituted alkyl group (C 1 -C 10 ) or (C 1 -C 10 )-O- (C 1 -C 10 ), each alkyl group (C 1 -C 10 ) is substituted with 1-5 Ry, each Ry is Rq, or (C 2 -C 10 )alkyl, (C 3 -C 10 An alkyl group, (C 3 -C 10 )cycloalkylalkyl group, (C 8 -C 14 )bicycloalkylalkyl group, (C 8 -C 14 )tricycloalkylalkyl group, —(C 5 - C 10 ) cycloalkylalkyl, (C 3 -C 10 )cycloalkyl, (C 8 -C 14 )bicycloalkylalkyl, (C 8 -C 14 )tricycloalkylalkyl, phenyl , naphthyl, (C 14 ) aromatic groups, each of which may be substituted with one or more Rz, each Rz being (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 1 -C 6 )alkynyl, (C 3 -C 8 )cycloalkyl, (C 6 -C 8 )cycloalkenyl, phenyl, 3- to 5-membered heterocyclic ring, C(halogen) 3 Or CH(halogen) 2 ; R 1 , R 2 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H), or a (C 1 -C 10 )alkyl group, ( C 2 -C 10 )alkenyl, (C 2 -C 10 )alkynyl, (C 3 -C 10 )cycloalkyl, (C 8 -C 14 )bicycloalkylalkyl, (C 8 -C 14 ) tricyclo alkyl, phenyl, naphthyl, (C 14) Fragrance groups, each of which may be unsubstituted, substituted by one or more Rz or Rq, one or more Rq substitutions with more than one Rz substitution, each Rq may be CN, OH, halogen , N 3 , NO 2 , N(Rz) 2 , ═NR z, CH═NRz, NRz OH, ORz, CORz, C(O)ORz, OC(O)Rz, OC(O)ORz, SRz, S( O) Rz or S(O) 2 Rz; Y 1 , Y 2 and Y 3 are independently CH 3 , (C 1 -C 12 )alkyl, or may be a (C 1 -C 12 ) alkane substituted with a combination of Ry Each of the (C 1 -C 12 )alkyl groups may be substituted with from 1 to 5 R y or with one counter ion.
在一个优选的方面,Xa、Xb、Xb、Xd、Xe、Xf、Xg、Xh、Xi和X1、X2、X3、X4、X5、X6和X7独立为氧原子(O),Ra、Rb、Rc、Rd、Re、Rf、Rg、Rh、Ri、R1、R2、R3、R4、R5、R6、R7和R8独立为氢原子(H),Y1、Y2和Y3独立为CH3In a preferred aspect, Xa, Xb, Xb, Xd, Xe, Xf, Xg, Xh, Xi and X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently oxygen atoms (O And Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ri, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H) , Y 1 , Y 2 and Y 3 are independently CH 3 .
在一个特定实施方式中,Xa、Xb、Xb、Xd、Xe、Xf、Xg、Xh、Xi和X1、X2、X3、X4、X5、 X6和X7独立为氧原子(O),Ra、Rb、Rc、Rd、Re、Rf、Rg、Rh、Ri、R1、R2、R3、R4、R5、R6、R7和R8独立为氢原子(H),Y1、Y2和Y3独立为CH3In a specific embodiment, Xa, Xb, Xb, Xd, Xe, Xf, Xg, Xh, Xi and X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently oxygen atoms ( O), Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ri, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H) ), Y 1 , Y 2 and Y 3 are independently CH 3 .
本领域技术人员可以理解,所述淫羊藿苷及其衍生物包括其单独的异构体、异构体的外消旋或非外消旋混合物、氧化物、或其药学上可接受的盐和水合物。It will be understood by those skilled in the art that the icariin and its derivatives include their individual isomers, racemic or non-racemic mixtures of isomers, oxides, or pharmaceutically acceptable salts thereof And hydrates.
在本发明的一个具体实施方式中,通过对淫羊藿苷化合物进行基团修饰,如氨基化、羧基化、醛基化、苄基化等化学修饰,可制备活性以及理化特性不同的淫羊藿苷化合物衍生物。In a specific embodiment of the present invention, the icariin compound can be modified by a group such as amination, carboxylation, aldehyde formation, benzylation or the like to prepare epidemis with different activities and physical and chemical properties. A glycoside compound derivative.
在一个具体实施方式中,本发明中使用的淫羊藿苷及其淫羊藿苷衍生物可以为天然的或化学合成的和改性的具有上述式(I)或(II)的化合物。化合物可以通过已知方法制备。本领域技术人员可以容易地选择溶剂、温度、压力和其他反应条件。起始原料是市售的或本领域技术人员可以通过已知方法很容易制备。In a specific embodiment, the icariin and its icariin derivative used in the present invention may be a natural or chemically synthesized and modified compound having the above formula (I) or (II). The compound can be produced by a known method. Solvents, temperatures, pressures, and other reaction conditions can be readily selected by those skilled in the art. Starting materials are commercially available or can be readily prepared by one skilled in the art by known methods.
在一个优选实施方式中,本发明中使用的天然的淫羊藿苷为淫羊藿属植物中提取和纯化的。In a preferred embodiment, the natural icariin used in the present invention is extracted and purified from Epimedium plants.
可选地,天然的淫羊藿苷植物是从小檗科(Berberidaceae)淫羊藿属(Epimedium)植物提取纯化而成的。所述淫羊藿属植物包括由宝兴淫羊藿(E.davidii)、保靖淫羊藿(E.baojingense)、朝鲜淫羊藿朝鲜淫羊藿(E.icoroanum)、川鄂淫羊藿(E.fargesii)、川西淫羊藿(E.elongatum)、粗毛淫羊藿(E.acuminatum)、单叶淫羊藿(E.simplicifolium)、短茎淫羊藿(E.brachyrrhizum)、多花淫羊藿(E.multiflorum)、恩施淫羊藿(E.enshiense)、革叶淫羊藿(E.reticulatum)、光叶淫羊藿(E.sagittatum)、宽萼淫羊藿(E.latisepalum)、绿药淫羊藿(E.chlorandrum)、茂汶淫羊藿(E.platypetalum)、偏斜淫羊藿(E.truncatum)、强茎淫羊藿(E.rhizomatosum)、三枝九叶草(E.sagittatum)、少花淫羊藿(E.pauciflorum)、天平山淫羊藿(E.myrianthum)、无距淫羊藿(E.ecalcaratum)、腺毛淫羊藿(E.glandulosopilosum)、小叶淫羊藿(E.parvifolium)、星花淫羊藿(E.stellulatum)、镇坪淫羊藿(E.ilicifolium)组成的组的一种或多种混合物。本说明书中使用的一些术语的定义,除非另有说明,本文中使用的所有技术和科学用语通常具有和本发明所属领域的普通技术人员通常理解的意思相同的意思。Alternatively, the natural icariin plant is extracted and purified from the Epimedium plant of the Berberidaceae family. The Epimedium plants include E. davidii, E. baojingense, E. icoroanum, and E. icoroanum. (E. fargesii), E. elongatum, E. acuminatum, E. simplicifolium, E. brachyrrhizum, multi-flower Epimedium (E. multiflorum), E. enshiense, E. reticulatum, E. sagittatum, E. latisepalum ), E.chlorandrum, E.platypetalum, E. truncatum, E. rhizomatosum, three-leaf clover (E.sagittatum), E. pauciflorum, E. myrianthum, E. ecalcaratum, E. glandulosopilosum, One or more mixtures of the group consisting of E. parvifolium, E. stellulatum, and E. ilicifolium. The definitions of some terms used in the specification, unless otherwise indicated, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
淫羊藿苷及其衍生物的预防用途和治疗用途Prophylactic and therapeutic uses of icariin and its derivatives
在本发明的一个实施方式中,通过使用潜在的候选者化合物进行酶促动力学实验,本领域技术人员筛选具有抑制流感病毒神经氨酸酶活性,从而抑制流感病毒复制。特别地,通过候选者化合物的酶促动力学筛选实验确定淫羊藿苷及其衍生物能够完全抑制流感病毒神经氨酸酶活性,用于预防和/或治疗流感病毒疾病。In one embodiment of the invention, one skilled in the art screens to inhibit influenza virus neuraminidase activity by inhibiting influenza virus replication by performing enzymatic kinetic experiments using potential candidate compounds. In particular, it has been determined by enzymatic kinetic screening experiments of candidate compounds that icariin and its derivatives are capable of completely inhibiting influenza virus neuraminidase activity for the prevention and/or treatment of influenza virus diseases.
在各种流感病毒引起的疾病的具体实施方式中,以重量计,流感病毒与淫羊藿苷的添加的比例关系可以为1:0.00001-10000,优选为1:0.0001-1000;更优选为1:0.001-500;最优选为1:0.01-100以达到完全抑制流感病毒神经氨酸酶,进而抑制流感病毒的复制。In a specific embodiment of the diseases caused by various influenza viruses, the ratio of the addition of the influenza virus to the icariin may be 1:0.00001-10000, preferably 1:0.0001-1000; more preferably 1 by weight. 0.001-500; most preferably 1:0.01-100 to achieve complete inhibition of influenza virus neuraminidase, thereby inhibiting replication of influenza virus.
在本发明的另一个具体实施方式中,制备的淫羊藿苷及其衍生物治疗或预防流感病毒导致的病理症状或疾病。所述的流感病毒包括由流感病毒甲型、乙型和丙型及其他们的各种亚型、血清型、基因型、基因突变体病毒;所述的病理症状或疾病由上述病毒各种亚型引起的流感综合症组成的一种或多种的疾病。优选为感冒综合症、咳嗽、发烧、喷嚏、鼻塞、流清水样鼻涕、畏寒、头痛、乏力、全身酸痛、肺炎、扁桃体炎、咽炎、喉炎、流泪、声音嘶哑、呼吸综合症、 心肺症、四肢冰冷,恶心呕吐、干咳,口干舌燥,头昏欲睡,腹泻、感冒流产、心肌炎、高血压、心肌梗塞、脑中风、鼻腔黏膜充血、水肿、有分泌物、咽部轻度充血、颌下淋巴结肿大、咽部充血水肿。In another embodiment of the invention, the prepared icariin and its derivatives treat or prevent pathological symptoms or diseases caused by influenza virus. The influenza virus includes influenza virus A, B and C and their various subtypes, serotypes, genotypes, gene mutant viruses; the pathological symptoms or diseases are caused by various viruses of the above viruses Type one or more diseases caused by influenza syndrome. Preferred are cold syndrome, cough, fever, sneezing, nasal congestion, runny nose, chills, headache, fatigue, body aches, pneumonia, tonsillitis, pharyngitis, laryngitis, tearing, hoarseness, respiratory syndrome, Cardiopulmonary disease, cold limbs, nausea and vomiting, dry cough, dry mouth, drowsiness, diarrhea, cold abortion, myocarditis, hypertension, myocardial infarction, stroke, nasal mucosal congestion, edema, secretions, pharyngeal light Degree of congestion, submandibular lymphadenopathy, pharyngeal congestion and edema.
本文中使用的术语“治疗”或指的是治疗处理措施和预防的或防止的措施,其中,防止或减缓(减弱)受试者的患有的病理症状或失调。如果接受了根据本文所述的方法的治疗量的淫羊藿苷及其衍生物,则受试者的症状得到成功地“治疗”,受试者显示,可以观察到和/或测定到症状的一种或多种迹象和症状减少和消失。还应当理解,本文描述的治疗或防止医疗状况的各种模式意在表示“显著”,其包括全部治疗或者预防以及小于全部治疗或者预防,其中达到了某种生物学相关或医学相关的结果。The term "treating" as used herein, or refers to a therapeutic treatment and a preventive or preventive measure, wherein the subject's pathological symptoms or disorders are prevented or slowed down (weakened). If a therapeutic amount of icariin and its derivatives according to the methods described herein is accepted, the subject's symptoms are successfully "treated" and the subject shows that symptoms can be observed and/or determined One or more signs and symptoms are reduced and disappeared. It should also be understood that the various modes of treating or preventing a medical condition described herein are intended to mean "significant", which includes all treatment or prevention and less than all treatment or prevention, wherein certain biologically relevant or medically relevant results are achieved.
在各种实施方式中,进行合适的体外测定或体内测定来测定基于特定的淫羊藿苷及其衍生物的治疗药物的效果以及其给药是否适于治疗。在各种实施方式中,可以对受试者的病症所涉及的代表性细胞进行体外测定,来测定给定的基于淫羊藿苷及其衍生物的治疗药物是否对所述病毒类型产生了期望的效果。如果接受了根据本文所述的方法的治疗量的淫羊藿苷及其衍生物,则受试者的症状得到成功地治疗,即受试者显示,可以观察到和/或测定到上述病理症状或疾病的一种或多种迹象和症状减少和消失。还应当理解,本文描述的治疗或防止医疗状况的各种模式意在表示“显著”,其包括全部治疗或者预防以及小于全部治疗或者预防,其中达到了某种生物学相关或医学相关的结果。In various embodiments, suitable in vitro assays or in vivo assays are performed to determine the effect of a therapeutic agent based on a particular icariin and its derivatives and whether its administration is suitable for treatment. In various embodiments, in vitro assays can be performed on representative cells involved in a subject's condition to determine whether a given therapeutic drug based on icariin and its derivatives has an expectation of the virus type. Effect. If a therapeutic amount of icariin and its derivatives according to the methods described herein is accepted, the subject's symptoms are successfully treated, ie, the subject shows that the above pathological symptoms can be observed and/or determined Or one or more signs and symptoms of the disease are reduced and disappeared. It should also be understood that the various modes of treating or preventing a medical condition described herein are intended to mean "significant", which includes all treatment or prevention and less than all treatment or prevention, wherein certain biologically relevant or medically relevant results are achieved.
本发明的一个具体实施方式中,对受试者施用预防和/或治疗有效量的含有淫羊藿苷及其衍生物的药物。In a specific embodiment of the invention, the subject is administered a prophylactically and/or therapeutically effective amount of a medicament comprising icariin and a derivative thereof.
在对人类受试者进行测试之前,可以在合适的动物模型系统中测试治疗中所用的候补化合物,所述动物受试者模型系统包括但不局限于非人灵长类(例如狒狒、猩猩、猴);诸如猫、狗、蛇等的宠物动物;猪、马、牛、山羊等的农场动物;诸如大鼠、小鼠、猴、兔等的实验室动物的任何动物。在一个实施方式中,将淫羊藿苷及其衍生物给药至患有或处于上述病理症状或疾病的危险中(易得该症状的状态)的受试者,来试图改善造成该病理症状或疾病的一个或多个因素。The candidate compounds used in the treatment can be tested in a suitable animal model system including, but not limited to, non-human primates (eg, baboons, orangutans, prior to testing the human subject). Monkeys; pet animals such as cats, dogs, snakes, etc; farm animals of pigs, horses, cows, goats, etc.; any animal of laboratory animals such as rats, mice, monkeys, rabbits, and the like. In one embodiment, icariin and a derivative thereof are administered to a subject suffering from or at risk of the above-mentioned pathological symptoms or diseases (a state in which the symptoms are readily available) in an attempt to improve the pathological symptoms Or one or more factors of the disease.
本文中使用的术语“有效量”指的是足以获得所需的治疗和/或预防效果,例如引起预防或减轻与病理症状或疾病的症状的量。给药至受试者的组合物的量将取决于疾病的类型和严重性以及个体的性质,比如平时的健康情况、年龄、性别、体重和对药物的耐受力。所述量还取决于患病程度、严重性和类型。本领域技术人员将能够根据这些因素和其他因素来确定合适的剂量。所述组合物还可结合一种或多种其他的治疗化合物来给药。在本文描述的方法中,本发明的化合物可以给药至具有病理症状的一种或多种迹象或病理症状的受试者。例如,“治疗有效量”是指最小程度地减轻症状的生理作用的平均水平。The term "effective amount" as used herein refers to an amount sufficient to achieve the desired therapeutic and/or prophylactic effect, for example, to cause prevention or alleviation of symptoms associated with pathological symptoms or diseases. The amount of the composition administered to the subject will depend on the type and severity of the disease as well as the nature of the individual, such as the usual health conditions, age, sex, weight, and tolerance to the drug. The amount will also depend on the severity, severity and type of disease. Those skilled in the art will be able to determine the appropriate dosage based on these and other factors. The composition may also be administered in combination with one or more additional therapeutic compounds. In the methods described herein, the compounds of the invention can be administered to a subject having one or more signs or pathological symptoms of pathological symptoms. For example, "therapeutically effective amount" refers to an average level that minimizes the physiological effects of symptoms.
通常,所述剂量将能够预防或减轻所治疗状况或指征的严重性或扩展。正确的剂量将依赖于环境,例如所治疗的状况、给药时间表、所述化合物是否单独施用或者与另一种治疗剂结合施用、所述化合物的血浆半衰期和受试者的整体健康状况。Generally, the dosage will be capable of preventing or reducing the severity or spread of the condition or indication being treated. The correct dosage will depend on the circumstances, such as the condition being treated, the schedule of administration, whether the compound is administered alone or in combination with another therapeutic agent, the plasma half-life of the compound, and the overall health of the subject.
可以采用包括口服,外用,吸入,经鼻,经直肠,经皮或注射施用方式对受试者用药。The subject can be administered by oral, topical, inhalation, nasal, rectal, transdermal or injection administration.
在本发明的一个具体实施方式中,制备了含有对于本文公开的所述化合物,每日给药剂量 方案为约0.1mg/kg至约10000mg/kg体重、优选为约0.1mg/kg至约1000mg/kg体重、更有选为0.1mg/kg至约500mg/kg,最优选为0.5mg/kg至约200mg/kg体重。每日可施用1至6次,优选每日施用2或3次。间隔也可以是不规则的,本领域技术人员公认化合物或其药学上可接受的盐的最佳量和单次给药的间隔将由待治疗状况的性质和程度,给药的形式、途径和位点以及所治疗的受试者的具体情况而确定,并且可以常规技术确定最优选方案。本领域技术人员还应该理解,治疗的最佳过程,即在给定的天数内每日给予的化合物或其药学上可接受的盐的给药数量可以由本领域技术人员利用常规的治疗测试方法而确定。在治疗应用中,在相对短的时间间隔中有相对高的剂量有时是需要的,直到疾病的进程减缓或终止为止,且优选地直到受试者显示部分或完全地改善了所述疾病或病理症状。因此,患者可以对患者实行预防性的给药方式。本领域技术人员将认识到,某些因素可以影响有效地治疗一受试者的剂量和时间,包括但不局限于疾病或失调的严重程度、先前的治疗、健康情况和/或受试者的年龄以及存在的其它疾病。而且,利用本文描述的治疗有效量的治疗对照组合物来治疗一受试者可以包括单次治疗或一系列治疗。In a specific embodiment of the invention, a daily dosage is prepared containing the compound disclosed herein. The regimen is from about 0.1 mg/kg to about 10000 mg/kg body weight, preferably from about 0.1 mg/kg to about 1000 mg/kg body weight, more preferably from 0.1 mg/kg to about 500 mg/kg, and most preferably from 0.5 mg/kg to About 200 mg/kg body weight. It can be administered from 1 to 6 times a day, preferably 2 or 3 times a day. The spacing may also be irregular, and those skilled in the art recognize that the optimal amount of the compound or its pharmaceutically acceptable salt and the interval between single administrations will depend on the nature and extent of the condition to be treated, the form, route and location of administration. The point and the particular condition of the subject being treated are determined and the most preferred protocol can be determined using conventional techniques. It will also be understood by those skilled in the art that the optimal course of treatment, i.e., the amount of the compound administered daily or a pharmaceutically acceptable salt thereof administered over a given number of days, can be utilized by those skilled in the art using conventional therapeutic test methods. determine. In therapeutic applications, relatively high doses in relatively short time intervals are sometimes required until the course of the disease slows or terminates, and preferably until the subject shows a partial or complete improvement in the disease or pathology symptom. Therefore, the patient can perform a prophylactic administration method to the patient. One of skill in the art will recognize that certain factors can affect the dosage and timing of effective treatment of a subject, including but not limited to the severity of the disease or disorder, prior treatment, health conditions, and/or subject's Age and other diseases that exist. Moreover, treating a subject with a therapeutically effective amount of a therapeutic control composition described herein can include a single treatment or a series of treatments.
本发明的化合物可以与至少另外一种活性成分其他化合物和/或中药成分一起施用。所述化合物可以作为单独的制剂或者组合在单位剂型中同时施用,或者顺次施用。所述的化合物为利巴韦林(Ribavirin1)、干扰素(Interferon)、胸腺肽α(Thymosinα)转移因子(Transfer factor)、齐多夫定(zidovudine,ZDV)、地丹诺辛(didanosine)、拉米夫定(lamivudine)、斯塔夫定(stavudine)、扎西他宾(zalcitabine)、abacavir、病毒唑(Ribavirin)、(三氮畦唑核苷(Virazole)、奈韦拉平(nevirapine)、地拉夫定(delavirdine)、efavirenz、沙奎那韦(saquinavir)、利托那韦(ritonavir)、奈非那韦(nelfinavir)、英地那韦(indinavir)、安泼那韦(amprenavir)、Lopinavir、金刚烷胺(Amantadine)、金刚乙胺(Rimantadine)、扎那米韦(Zanamivir)、阿昔洛韦(Acyclovir)、伐昔洛韦(Valacyclovir)、碘苷(Idoxuridine)、阿糖腺苷(Vidarabine)、拉米夫定(Lamivudine)、泛昔洛韦(Famciclovir)、喷昔洛韦(Penciclovir)、奥司他韦和扎那米韦。其中所述至少另外一种中药活性成分为板蓝根、大青叶、银翘、连翘、荆芥、金银花、鱼腥草、野菊花、夏桑菊、柴胡、穿心莲等中药成分的一种或多种组合。在任何情况下,多种治疗剂可以以任何顺序或甚至同时给药。如果同时地,所述多种治疗剂可以提供为单一、统一的形式或者以多个形式(例如,作为单一的片剂或者各自独立的片剂或胶囊、滴丸的几种不同制剂)。所述治疗剂中的一种治疗剂可能以多剂量型提供,或者其中的几种可以作为多剂量型提供。如果不同时地,则多次剂量之间的时间间隔可以从大于0周到小于约1周、小于约2周、小于约4周、小于约2个月、小于约4个月或甚至小于约半年的范围中变化。The compounds of the invention may be administered with at least one other active ingredient other compound and/or a Chinese medicinal ingredient. The compounds can be administered simultaneously as a separate formulation or in a unit dosage form, or sequentially. The compound is ribavirin (1), interferon, thymosin alpha , transfer factor, zidovudine (ZDV), didanosine, Lamivudine, stavudine, zalcitabine, abacavir, Ribavirin, (Virazole), nevirapine, diarrhife Delavirdine, efavirenz, saquinavir, ritonavir, nelfinavir, indinavir, amprenavir, Lopinavir, King Kong Amantadine, Rimantadine, Zanamivir, Acyclovir, Valacyclovir, Idoxuridine, Vidarabine , Lamivudine, Famciclovir, Penciclovir, oseltamivir and zanamivir, wherein at least one other active ingredient of the traditional Chinese medicine is Radix, Folium, Silver Aster, Forsythia, Nepeta, Honeysuckle, Houttuynia, Wild Chrysanthemum, Summer Sangju, Chai One or more combinations of traditional Chinese medicine ingredients such as andrographis paniculata. In any case, the plurality of therapeutic agents can be administered in any order or even simultaneously. If at the same time, the plurality of therapeutic agents can be provided in a single, uniform form. Or in multiple forms (for example, as a single tablet or as separate tablets or capsules, several different formulations of pills). One of the therapeutic agents may be provided in a multi-dose form, or Several may be provided as a multi-dose type. If not simultaneously, the time interval between multiple doses may range from greater than 0 weeks to less than about 1 week, less than about 2 weeks, less than about 4 weeks, less than about 2 months, A change in the range of less than about 4 months or even less than about half a year.
本文使用的术语“单位剂型”是指适合作为用于人和动物受试者的单位剂量的在物理上分离的单元,各单元包含单独的预定量的化合物或者与其它试剂组合,其量经计算足以与药学上可接受的稀释剂、载体或媒介组合产生期望效果。The term "unit dosage form" as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit comprising a separate predetermined amount of compound or in combination with other agents, the amount being calculated Sufficient to combine with a pharmaceutically acceptable diluent, carrier or vehicle to produce the desired effect.
在本发明的具体实施方式中,制备含有淫羊藿苷或其衍生物的药物组合物,其用于治疗和/或预防由流感病毒引起的感冒性疾病得到改善相关的病理症状或疾病。其通过口服、注射、吸入法等方式,对受试者给药而发挥作用。因此本发明的药物组合物可以制备成各种剂型,例如,用于口服用药的组合物可以为胶囊、滴丸、凝胶剂、片剂、粉剂、颗粒剂、含片、泡腾片、糖浆、乳化物、控释的制备物(包括缓释片剂或缓释胶囊等)、快速溶解的制备物、口服液体剂 型等;用于外用用药的组合物可以为涂抹剂型药物、霜剂、外敷、软膏、外用药水、液体喷雾剂和气喷雾剂等。用于吸入法给药的组合物可以为溶液、分散液、干粉等;也可以从中草药淫羊藿中提取高纯度的淫羊藿苷化合物,制备脂质体包裹的纳米淫羊藿苷,制备成注射制剂以及缓释药物剂型。在所述的药物组合物中,淫羊藿苷或者其衍生物所占比例,以重量占整个制剂的总重量为0.0%-99.0%,优选地0.0%-90%,更优选地为10%-80%,最优选为25-70%。In a specific embodiment of the present invention, a pharmaceutical composition containing icariin or a derivative thereof for treating and/or preventing a pathological symptom or disease associated with an improvement of a cold disease caused by an influenza virus is prepared. It is administered to a subject by oral administration, injection, inhalation, or the like. Therefore, the pharmaceutical composition of the present invention can be prepared into various dosage forms. For example, the composition for oral administration can be a capsule, a dropping pill, a gel, a tablet, a powder, a granule, a lozenge, an effervescent tablet, or a syrup. , emulsion, controlled release preparation (including sustained release tablets or sustained release capsules, etc.), fast-dissolving preparations, oral liquid preparations The composition for external use may be a smear-type drug, a cream, an external application, an ointment, a topical syrup, a liquid spray, an air spray, or the like. The composition for administration by inhalation may be a solution, a dispersion, a dry powder or the like; or a high-purity icariin compound may be extracted from the Chinese herbal medicine Epimedium, and a liposome-encapsulated nano icariin is prepared and prepared. Injectable preparations and sustained release pharmaceutical dosage forms. In the pharmaceutical composition, the proportion of icariin or a derivative thereof is from 0.0% to 99.0%, preferably from 0.0% to 90%, more preferably 10% by weight based on the total weight of the entire preparation. -80%, most preferably 25-70%.
上述制剂可以为控制制剂,包括缓释片剂或缓释胶囊。所述控释片剂包括核芯和包裹核芯的控释层。所述核芯含有至少两种活性成分,其中至少一种活性成分为淫羊藿苷及其衍生物。所述核芯还可以包含至少另外一种活性成分为干扰素(Interferon)、胸腺肽α(Thymosinα)病毒唑(Ribavirin)、金刚烷胺(Amantadine)、金刚乙胺(Rimantadine)奥司他韦和扎那米韦。所述核芯也可以含有至少另外一种中药活性成分为板蓝根、大青叶、连翘、金银花、柴胡、穿心莲等中药成分的一种或多种组合。所述的控释片剂的控释层含有甲基丙烯酸和醋酸乙酯共聚物、甲基丙烯酸和甲基丙烯酸甲酯共聚物、乙基纤维素、柠檬酸三乙酯。其中,按照重量比,所述核芯和包裹核芯的控释层的比例为1:0.01-100。The above preparation may be a controlled preparation, including a sustained release tablet or a sustained release capsule. The controlled release tablet comprises a core and a controlled release layer that encapsulates the core. The core contains at least two active ingredients, at least one of which is icariin and a derivative thereof. The core may further comprise at least one other active ingredient of Interferon, Thymosin alpha , Ribavirin, Amantadine, Rimantadine oseltamivir and Zanamivir. The core may also contain at least one other traditional Chinese medicine active ingredient which is one or more combinations of traditional Chinese medicine ingredients such as Radix, Eucalyptus, Forsythia, Honeysuckle, Bupleurum, Andrographis paniculata. The controlled release layer of the controlled release tablet comprises a copolymer of methacrylic acid and ethyl acetate, a copolymer of methacrylic acid and methyl methacrylate, ethyl cellulose, and triethyl citrate. Wherein, according to the weight ratio, the ratio of the core and the controlled release layer of the package core is 1:0.01-100.
在本发明的一个具体实施方式中,提供了一种制备缓释片剂的方法。所制造的控释制剂,在pH 1至10.0范围内提供与pH无关的体外释放特性。In a specific embodiment of the invention, a method of preparing a sustained release tablet is provided. The controlled release formulations produced provide pH independent release characteristics in the range of pH 1 to 10.0.
在本发明药物制剂中,以口服制剂为例,所用药学上可接受的辅料包括但不局限于润滑剂和助溶剂,如硬脂酸镁、硬脂酸钙、硬脂酸锌、硬脂酸、山嵛酸甘油酯、十八烷基富马酸钠、滑石、无水胶体二氧化硅、硅胶;崩解剂,如淀粉、环糊精、麦芽糊精、羧甲基纤维素、甲基丙烯酸和醋酸乙酯共聚物、乙烯基醋酸酯共聚物、甲基丙烯酸和甲基丙烯酸甲酯共聚物、乙基纤维素、柠檬酸三乙酯、交联羧甲基纤维素、交联聚乙烯吡咯烷酮、明胶、微晶纤维素;稀释剂或压缩剂,如麦芽糊精葡萄糖、乳糖、半乳糖、蔗糖、甘露醇木糖醇、赤藻糖醇、山梨糖醇、微晶纤维素;涂覆剂,干漆、N-乙烯基丁内酰胺、二氧化钛;调味料或其它成分,如草莓、桔子、香蕉、薄荷、蜂蜜;助溶剂,如酒类(啤酒、果酒、米酒、白酒)、植物油、动物油、乙醇、异丙醇、甘油、甘露醇、丙二醇、苯甲醇、二甲基亚风、聚乙二醇、阿拉伯胶、卵磷脂、吡咯酮、油酸、氮酮;着色剂。In the pharmaceutical preparation of the present invention, the oral preparation is exemplified, and the pharmaceutically acceptable excipients used include, but are not limited to, lubricants and co-solvents such as magnesium stearate, calcium stearate, zinc stearate, stearic acid. , behenic acid glyceride, octadecyl fumarate, talc, anhydrous colloidal silica, silica gel; disintegrating agents such as starch, cyclodextrin, maltodextrin, carboxymethyl cellulose, methyl Acrylic acid and ethyl acetate copolymer, vinyl acetate copolymer, methacrylic acid and methyl methacrylate copolymer, ethyl cellulose, triethyl citrate, croscarmellose, crosslinked polyethylene Pyrrolidone, gelatin, microcrystalline cellulose; diluent or compressive agent, such as maltodextrin glucose, lactose, galactose, sucrose, mannitol xylitol, erythritol, sorbitol, microcrystalline cellulose; coating , dry paint, N-vinyl butyrolactam, titanium dioxide; seasonings or other ingredients, such as strawberries, oranges, bananas, mint, honey; cosolvents, such as alcohol (beer, fruit wine, rice wine, white wine), vegetable oil, Animal oil, ethanol, isopropanol, glycerin , mannitol, propylene glycol, benzyl alcohol, dimethyl sulfite, polyethylene glycol, gum arabic, lecithin, pyrrolidone, oleic acid, azone; colorant.
载体颗粒可以为乳糖或蔗糖的晶体或球体;或复合球体或颗粒,如通过用淀粉作为粘合剂使蔗糖成粒状形成的糖的球体,通过淀粉作为粘合剂形成的碳酸钙球体或麦芽糊精。载体颗粒也可以是任何其他的医药可以接受的赋形剂的颗粒,如羟丙基纤维素颗粒、瓜尔豆胶颗粒、黄原胶颗粒。载体可以有多种形式,所有这些选择和量的调整显然在本领域技术人员的能力范围内。The carrier particles may be crystals or spheres of lactose or sucrose; or composite spheres or granules, such as spheres of sugar formed by granulating sucrose with starch as a binder, calcium carbonate spheres or malt paste formed by starch as a binder fine. The carrier particles can also be particles of any other pharmaceutically acceptable excipient such as hydroxypropylcellulose granules, guar gum granules, xanthan gum granules. The carrier can take a wide variety of forms, and all such selections and amounts of adjustments are clearly within the abilities of those skilled in the art.
通过经口途径,根据本发明的淫羊藿苷或其衍生物的化合物或其组合物可以包含溶解在食品或保健品液体中的,如可选地溶解在调味的水性的液体或水-醇类溶液。其可以被纳入可吞服的固体赋形剂,例如在颗粒形式,丸剂,片剂,肠溶片的形式中。其也可以被放置在食物或保健品中的液体中,其本身可选的条件是在吞服胶囊中。对于吞服而言,可以为多种口服组合物实施方式,特别是,可以是食品附加物的口服组合物实施方式。通过常规的方法制造肠溶片,胶体胶囊,凝胶,乳化物,片剂,胶囊或溶液而制剂。具体地,根据本发明的活性剂可以纳入食物补充剂或强化食品或保健品的任何其它形式,例如食品或保健品棒,或者压缩或非压缩的 粉剂之中。这些粉剂可以用水稀释,可以在苏打,乳酪产品或大豆衍生物中用水稀释,或可以纳入到食品或保健品棒中。By the oral route, the compound of icariin or a derivative thereof according to the present invention or a composition thereof may comprise a liquid or a water-alcohol which is dissolved in a food or a health care product liquid, such as optionally dissolved in a flavoring. Class solution. It can be incorporated into a swallowable solid excipient, for example in the form of granules, pills, tablets, enteric coated tablets. It can also be placed in a liquid in a food or health supplement, optionally in the swallow capsule. For swallowing, a variety of oral composition embodiments can be employed, and in particular, can be oral composition embodiments of food addenda. The enteric coated tablets, colloidal capsules, gels, emulsions, tablets, capsules or solutions are prepared by conventional methods. In particular, the active agent according to the invention may be incorporated into a food supplement or any other form of fortified food or health supplement, such as a food or health care product stick, or compressed or uncompressed Among the powders. These powders can be diluted with water, diluted with water in soda, cheese products or soy derivatives, or can be incorporated into food or health care bars.
通过如下实施例记载,更好地理解本发明,但不局限于该实施例。The present invention will be better understood by the following examples, but is not limited to the examples.
实施例Example
实施例1.流感病毒神经氨酸酶的制备及其淫羊藿苷抑制流感病毒神经氨酸酶活性分析Example 1. Preparation of influenza virus neuraminidase and analysis of its effect on inhibition of influenza virus neuraminidase activity by icariin
取104TCID50的流感病毒H5N1(A/chicken/Guangdong/174/04(H5N1))、H9N2(A/chicken/Guangdong/SS/94(H9N2),为了尝试淫羊藿苷是否对其它亚型神经氨酸酶的抑制效果,也选择了H9N2病毒株制备神经氨酸酶液),加入曲拉通X-100(Sigma)到1%浓度,制备流感病毒神经氨酸酶液,用于抑制剂活性测定。纯度98%的淫羊藿苷标准品(美国ChromaDex公司),用30%的二甲基亚风(Sigma)溶解,浓度为10mmol/L;Methylumbelliferyl-a-D-N-acetylneuramimic acid(MUNANA,sigma)配成20微摩尔/L浓度;奥司他韦(Oseltamivir,美国ChromaDex公司)用水配置成10mmol/L的浓度;10倍的缓冲液(325mmol MES(Sigma)、40mmol CaCl(Sigma),pH6.5)。酶活性测定反应体系为100微升,在黑色96孔板中进行。流感病毒神经氨酸酶液预先加入不同浓度的淫羊藿苷,再加入3μL反应缓冲液,水27μL,其中设有1μmol、5μmol、50μmol、100μmol、200μmol的淫羊藿苷浓度梯度(最终反应体系浓度)。同时按上述方法设奥司他韦作对照实验,最终反应体系浓度梯度为1μmol、5μmol、50μmol、100μmol、200μmol。另外,设不加流感病毒神经氨酸酶液(其它成分按上述浓度加,流感病毒神经氨酸酶液用水替代)、不加淫羊藿苷和奥司他韦(其它成分按上述浓度加,用水补充反应体积)、不加淫羊藿苷和奥司他韦但加入1%的DMSO(其它成分按上述浓度加,1%的DMSO是最终反应体系浓度)的对照实验,加MUNANA前各个反应体系为40μL,各个反应体系详见表1。37℃反应1小时后,所有反应体系加60微升20微摩尔/L MUNANA,37℃反应1小时,用0.002mol/L的氢氧化钠终止所有反应。用超级酶标仪(Synergy HT,blo-tek,USA)在360的激发光和460的发射光下测定荧光强度。Take 104TCID50 of influenza virus H5N1 (A/chicken/Guangdong/174/04 (H5N1)), H9N2 (A/chicken/Guangdong/SS/94 (H9N2), in order to try to determine whether icariin is related to other subtypes of neuraminic acid The inhibitory effect of the enzyme was also selected from the H9N2 virus strain to prepare a neuraminidase solution, and Triflurane X-100 (Sigma) was added to a concentration of 1% to prepare an influenza virus neuraminidase solution for assay of inhibitor activity. The 98% pure icariin standard (ChromaDex, USA) was dissolved in 30% dimethyl nitrous oxide (Sigma) at a concentration of 10 mmol/L; Methylumbelliferyl-aDN-acetylneuramimic acid (MUNANA, sigma) was formulated into 20 Micromol/L concentration; oseltamivir (Oseltamivir, ChromaDex, USA) was formulated with water to a concentration of 10 mmol/L; 10 times buffer (325 mmol MES (Sigma), 40 mmol CaCl (Sigma), pH 6.5). The enzyme activity assay reaction system was 100 microliters and was carried out in a black 96-well plate. Influenza virus neuraminidase solution was pre-charged with different concentrations of icariin, and then added 3 μL of reaction buffer, 27 μL of water, which contained 1 μmol, 5 μmol, 50 μmol, 100 μmol, 200 μmol of icariin concentration gradient (final reaction system) concentration). At the same time, oseltamivir was set as a control experiment according to the above method, and the final reaction system concentration gradient was 1 μmol, 5 μmol, 50 μmol, 100 μmol, and 200 μmol. In addition, it is not necessary to add influenza virus neuraminidase solution (other components are added at the above concentration, the influenza virus neuraminidase solution is replaced with water), no icariin and oseltamivir are added (other ingredients are added according to the above concentration, Replenish the reaction volume with water), add no icariin and oseltami but add 1% DMSO (addition of other components at the above concentration, 1% DMSO is the final reaction system concentration). Add the reaction before MUNANA. The system is 40 μL, and each reaction system is shown in Table 1. After reacting at 37 ° C for 1 hour, all the reaction systems were added with 60 μl of 20 μmol/L MUNANA, reacted at 37 ° C for 1 hour, and terminated with 0.002 mol/L sodium hydroxide. reaction. Fluorescence intensity was measured using a super-microplate reader (Synergy HT, blo-tek, USA) at 360 excitation light and 460 emission light.
表1、加MUNANA前各个反应体系的成分组成Table 1. Composition of each reaction system before adding MUNANA
Figure PCTCN2015097322-appb-000004
Figure PCTCN2015097322-appb-000004
Figure PCTCN2015097322-appb-000005
Figure PCTCN2015097322-appb-000005
由淫羊藿苷抑制流感病毒神经氨酸酶活性分析(图1),揭示了淫羊藿苷抑制流感病毒神经氨酸酶活性很强,比奥司他韦高近40倍,提示淫羊藿苷可以作为预防和/或治疗流感病毒疾病的候补药物先导化合物,有望开发出一种奥司他韦更好的药物。另外,本实验也证实了淫羊藿苷也同样能够很好地抑制不同亚型的神经氨酸酶活性,即能够抑制N1(H5N1)和N2(H9N2)酶活性,提示淫羊藿苷可能抑制各种亚型的神经氨酸酶活性。Inhibition of influenza virus neuraminidase activity by icariin (Fig. 1) revealed that icariin inhibits influenza virus neuraminidase activity, which is nearly 40 times higher than oseltamivir, suggesting epimedium Glycoside can be used as a lead compound for the prevention and/or treatment of influenza virus diseases, and it is expected to develop a better drug for oseltamivir. In addition, this experiment also confirmed that icariin can also inhibit the neuraminidase activity of different subtypes, which can inhibit the activity of N1 (H5N1) and N2 (H9N2) enzymes, suggesting that icariin may inhibit Neuraminidase activity of various subtypes.
实施例2.体外细胞水平上的淫羊藿苷对流感病毒抑制作用Example 2. Inhibition of influenza virus by icariin at the in vitro cell level
选择MDCK(Madin-Darby Canine Kidney)细胞(中国农业科学院哈尔滨兽医研究所提供)作为流感病毒增殖细胞,DMEM(Gibco公司)作为MDCK细胞基础培养基。按照Xie等方法(Yi Xie,James A.Schafer.Inhibition of ENaC by intracellular Cl-in an MDCK clone with high ENaC expression.Am J Physiol Renal Physiol.2004Oct;287(4):F722-31.),进行MDCK细胞培养:DMEM培养液190mL加入胎牛血清20mL,青链双抗2mL,7.5%NaHCO3调节pH为7.4,上述各组分混匀为MDCK完全培养液,取15代细胞瓶,加入消化液适量,以恰好盖过细胞为宜,轻放于37℃培养箱中约10min,待细胞圆缩,彼此互不相联时,轻轻吸弃消化液,加入培养液,反复轻吸吹打,至细胞脱壁,变匀,计数细胞,1.0×105/mL细胞接种于新的25mL培养瓶中,置37℃、5%CO2环境静置培养。将流感病毒接种于9~11日龄鸡胚尿囊腔,35℃孵育48~72h,然后将鸡胚置4℃冰箱过夜,次日收获尿囊液,3000rpm离心10min,弃去沉淀,取上清液进行血凝实验,确定其滴度大于1:64者收获上清液,-80℃保存。流感病毒感染细胞确定病毒滴度:培养细胞传代并计数,转移至6孔培养板中,调整每孔细胞密度为1.0×105/mL。取H5N1亚型、H9N2(为了尝试淫羊藿苷是否对其它亚型神经氨酸酶的抑制效果,我们同时选择了H9N2病毒株)亚型流感病毒1mL分别加入到其中A和B孔细胞中,C孔加入等量生理盐水作阴性对照。每孔培养体系最终体积调整为5mL,A和B孔细胞孵育72h共3组,每组再重复5次,显微镜下观察细胞病变(Cytopathic effect,CPE)情况,确定病毒滴度。参考Hatakeyama等的方法(Shuji Hatakeyama,Yuko Sakai-Tagawa,Maki  Kiso,Hideo Goto,Chiharu Kawakami,Keiko Mitamura,Norio Sugaya,Yasuo Suzuki and Yoshihiro Kawaoka.Enhanced Expression of anα2,6-Linked Sialic Acid on MDCK Cells Improves Isolation of Human Influenza Viruses and Evaluation of Their Sensitivity to a Neuraminidase Inhibitor.J Clin Microbiol.Aug 2005;43(8):4139–4146.),进行淫羊藿苷对于流感病毒抑制实验:淫羊藿苷用30%的二甲基亚风(sigma)溶解,浓度为1mmol/mL,分别加入到DMEM细胞培养基中,参考实施例1的实验结果,设浓度为5μmol进行检测淫羊藿苷抑制病毒复制实验检测,用PBS替代淫羊藿苷作抑制病毒复制的阴性对照。用96孔培养板,设5个平行实验,1.0×105/mL MDCK细胞100μL,加入100μL的含200TCID50滴度的病毒和各种浓度的淫羊藿苷,置37℃、5%CO2环境静置培养3天,观察病毒增值情况。分别使用流式细胞仪(Flow cytometry,美国贝克曼库尔特公司),参考Liu等的方法(Liu H,Zhang GL,Shen L,Zeng Z,Zhou BL,Liu CH,Nie G.Application and evaluation of a pseudotyped virus assay for screening herbs for anti-H5N1 avian influenza virus.Zhong Xi Yi Jie He Xue Bao.2010Nov;8(11):1036-40.)、荧光抗体(Anti-Influenza A H5N1 Hemagglutinin Antibody和Anti-Influenza A H9N2 Hemagglutinin Antibody,购自Sino Biotlogical Inc.),参考Ebrahimi等的方法(Ebrahimi SM,Tebianian M,Aghaiypour K,Nili H,Mirjalili A.Prokaryotic expression and characterization of avian influenza A virus M2 gene as a candidate for universal recombinant vaccine against influenza A subtypes;specially H5N1 and H9N2.Mol Biol Rep.2010 Jul;37(6):2909-14.),利用染色法分析病毒增殖情况,荧光显微镜(德国奥林巴斯)检测。实时无标记细胞功能分析仪(xCELLigence RTCA MP,ACEA Biosciences Inc)分析:将200TCID50/mL流感病毒H5N1和H9N2与已稀释的抑制剂按照1:1比例进行混合,置于37℃培养箱中进行孵育1小时,然后将病毒与抑制剂的混合物加入至已生长好MDCK细胞的96孔细胞电子检测板中,置于37℃培养箱中培养190-200小时。实验时,设有7个淫羊藿苷浓度梯度,即培养基含有1μmol、5μmol、10μmol、20μmol、50μmol、100μmol、200μmol,并设有病毒阴性对照和不含淫羊藿苷的接种病毒的对照。使用实时无标记细胞功能分仪进行培养和检测,每15min进行一次数据采集。MDCK (Madin-Darby Canine Kidney) cells (provided by Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences) were selected as influenza virus proliferating cells, and DMEM (Gibco) was used as the base medium for MDCK cells. MDCK according to the method of Xie et al. (Yi Xie, James A. Schafer. Inhibition of ENaC by intracellular Cl - in an MDCK clone with high ENaC expression. Am J Physiol Renal Physiol. 2004 Oct; 287(4): F722-31.) Cell culture: 190 mL of DMEM medium was added to 20 mL of fetal bovine serum, 2 mL of green chain double antibody, 7.5% NaHCO 3 to adjust the pH to 7.4. The above components were mixed into MDCK complete medium, 15 passages of cell bottles were added, and the amount of digestive juice was added. It is advisable to cover the cells just before, and gently put them in a 37 ° C incubator for about 10 minutes. When the cells are rounded and do not associate with each other, gently digest the digestive juice, add the culture solution, and repeatedly suck and blow until the cells are The cells were dissociated, and the cells were counted. 1.0 × 10 5 /mL cells were seeded in a new 25 mL culture flask, and cultured at 37 ° C in a 5% CO 2 atmosphere. The influenza virus was inoculated into the 9-day-old chicken embryoal allantoic cavity, incubated at 35 ° C for 48-72 h, then the chicken embryo was placed in a refrigerator at 4 ° C overnight, the allantoic fluid was harvested the next day, centrifuged at 3000 rpm for 10 min, and the precipitate was discarded. The serum was subjected to a blood coagulation test, and it was confirmed that the titer was greater than 1:64, and the supernatant was harvested and stored at -80 °C. Influenza virus-infected cells were determined for virus titer: cultured cells were passaged and counted, transferred to a 6-well culture plate, and the cell density per well was adjusted to 1.0 × 10 5 /mL. Take H5N1 subtype, H9N2 (in order to try to inhibit the effect of icariin on other subtypes of neuraminidase, we also selected H9N2 strain) subtype influenza virus 1mL was added to the A and B cells, The same amount of physiological saline was added to the C well as a negative control. The final volume of each well culture system was adjusted to 5 mL, and A and B cells were incubated for 72 h for a total of 3 groups. Each group was repeated 5 times. Cytopathic effect (CPE) was observed under a microscope to determine the virus titer. Refer to the method of Hatakeyama et al. (Shuji Hatakeyama, Yuko Sakai-Tagawa, Maki Kiso, Hideo Goto, Chiharu Kawakami, Keiko Mitamura, Norio Sugaya, Yasuo Suzuki and Yoshihiro Kawaoka. Enhanced Expression of an α2, 6-Linked Sialic Acid on MDCK Cells Improves Isolation Of Human Influenza Viruses and Evaluation of Their Sensitivity to a Neuraminidase Inhibitor.J Clin Microbiol.Aug 2005;43(8):4139–4146.), Icariin for Influenza Virus Inhibition Experiment: Icariin with 30% The dimethyl sigma was dissolved at a concentration of 1 mmol/mL and added to the DMEM cell culture medium. The experimental results of Example 1 were used to determine the concentration of 5 μmol for the detection of icariin inhibition virus replication assay. Icariin was replaced with PBS as a negative control to inhibit viral replication. Using a 96-well culture plate, set up 5 parallel experiments, 1.0 × 10 5 /mL MDCK cells 100 μL, add 100 μL of virus containing 200 TCID 50 titer and various concentrations of icariin, set at 37 ° C, 5% CO 2 environment The culture was allowed to stand for 3 days, and the virus value added was observed. Flow cytometry (Beckman Coulter, USA), refer to the method of Liu et al. (Liu H, Zhang GL, Shen L, Zeng Z, Zhou BL, Liu CH, Nie G. Application and evaluation of a pseudotyped virus assay for screening herbs for anti-H5N1 avian influenza virus.Zhong Xi Yi Jie He Xue Bao.2010Nov;8(11):1036-40.), fluorescent antibody (Anti-Influenza A H5N1 Hemagglutinin Antibody and Anti-Influenza A H9N2 Hemagglutinin Antibody, purchased from Sino Biotlogical Inc., with reference to the method of Ebrahimi et al. (Ebrahimi SM, Tebianian M, Aghaiypour K, Nili H, Mirjalili A. Prokaryotic expression and characterization of avian influenza A virus M2 gene as a candidate for universal Recombinant vaccine against influenza A subtypes;specially H5N1 and H9N2.Mol Biol Rep.2010 Jul;37(6):2909-14.), the virus proliferation was analyzed by staining and detected by fluorescence microscopy (Olympus, Germany). Real-time label-free cell function analyzer (xCELLigence RTCA MP, ACEA Biosciences Inc) analysis: 200 TCID50/mL influenza virus H5N1 and H9N2 were mixed with diluted inhibitors in a 1:1 ratio and placed in a 37 ° C incubator for incubation. One hour, a mixture of virus and inhibitor was then added to a 96-well cell electronic assay plate in which MDCK cells had been grown, and placed in a 37 ° C incubator for 190-200 hours. During the experiment, there were 7 icariin concentration gradients, ie, the medium containing 1 μmol, 5 μmol, 10 μmol, 20 μmol, 50 μmol, 100 μmol, 200 μmol, and a virus-negative control and icariin-free inoculated virus control . Culture and detection were performed using a real-time label-free cell function meter, and data acquisition was performed every 15 minutes.
体外细胞水平上的淫羊藿苷对流感病毒抑制作用研究(实施例2),通过MDCK细胞的病毒致病性和淫羊藿苷保护MDCK细胞免受流感病毒毁灭实验,包括使用荧光抗体检测感染流感病感染细胞(图2)、流式细胞仪分析(图3)和实时无标记细胞功能分析仪分析(图4),都很好地证明了淫羊藿苷能够抑制流感病毒增殖,保护了细胞免受病毒破坏。在这个实验中,我们使用了两种流感病毒神经氨酸酶的病毒株,即H5N1(N1)和H9N2(N2),淫羊藿苷对这两种不同的神经氨酸酶的病毒株显示了几乎同等的抑制效果,表明神经氨酸酶的病毒株可能是一个广谱的抗流感病毒亚型、血清型、基因型的抑制剂化合物,淫羊藿苷可以作为预防和/或治疗各种类型的流感病毒导致的病理症状或疾病的药物、食品、保健品。Inhibition of influenza virus by icariin at the cellular level in vitro (Example 2), protection of MDCK cells from influenza virus destruction by viral pathogenicity and icariin of MDCK cells, including detection of infection using fluorescent antibodies Influenza-infected cells (Fig. 2), flow cytometry analysis (Fig. 3), and real-time unlabeled cell function analyzer analysis (Fig. 4) are all well demonstrated that icariin can inhibit influenza virus proliferation and protect it. Cells are protected from viruses. In this experiment, we used two strains of influenza virus neuraminidase, namely H5N1 (N1) and H9N2 (N2), and icariin showed these two different neuraminidase strains. Almost the same inhibitory effect, suggesting that the neuraminidase virus strain may be a broad-spectrum inhibitor of influenza virus subtypes, serotypes, genotypes, and icariin can be used as a preventive and/or therapeutic agent. The flu virus causes pathological symptoms or diseases of drugs, foods, and health products.
实施例3.淫羊藿苷抑制流感病毒的动物实验Example 3. Animal experiment of icariin inhibiting influenza virus
3.1实验动物品种和特征3.1 Experimental animal species and characteristics
BALB/C小鼠,中国农业科学院哈尔滨兽医研究所实验动物中心繁殖,实验动物质量许可证号为黑龙江-SYXK 2007-167。BALB/C mice, the Experimental Animal Center of Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, were reproduced. The experimental animal quality license number was Heilongjiang-SYXK 2007-167.
年龄9周~10周,体重20g左右,雌雄各半。 Age 9 weeks to 10 weeks, weight 20g, male and female.
3.2饲养条件3.2 feeding conditions
动物实验室空气定时通风、光照良好,保持实施室室温。每笼饲养5只动物,喂以专门为小鼠制作的膨化饲料,自由饮水。实验开始前,观察动物进食、活动情况及其粪便等一周,只有健康的老鼠选择为下一步实验。The animal laboratory air is regularly ventilated and well lit, keeping the room temperature at the implementation room. Five animals were kept per cage, and the puffed feed specially made for mice was fed freely. Before the start of the experiment, observe the animals' feeding, activity and feces for a week. Only healthy mice were selected for the next experiment.
3.3淫羊藿苷的动物安全性评价3.3 Animal safety evaluation of icariin
首选,运用急性毒性试验对淫羊藿苷进行了安全性评价,以便为淫羊藿苷的应用提供依据。本实验采用急性毒性试验最大耐受量法,首先选取体重20g左右的健康BALB/C小鼠20只,雌雄各半。临床拟用的给药途径为口服,股本实验采用了灌胃法给药。淫羊藿苷用蒸馏水调制成黏糊状,1次灌胃给予小鼠(每只鼠200毫克淫羊藿苷左右),灌胃前动物禁食过夜,自由饮水。灌胃后给予正常饮食,观察中毒症状、死亡情况,每周称重一次,观察期为2周。在同样条件下,取未淫羊藿苷添加正常食物喂食同批老鼠作为对照组进行观察。实验结果结果显示所有小鼠均为出现任何中毒症状,证明淫羊藿苷属无毒物质。First, the acute toxicity test was used to evaluate the safety of icariin in order to provide a basis for the application of icariin. In this experiment, the maximum tolerance method for acute toxicity test was used. First, 20 healthy BALB/C mice weighing about 20 g were selected, male and female. The clinically recommended route of administration is oral, and the equity experiment is administered by intragastric administration. Icariin was prepared into a mushy paste with distilled water and administered to mice once a day (about 200 mg of icariin per mouse). The animals were fasted overnight and given free access to water before gavage. After the stomach was given a normal diet, the symptoms of poisoning and death were observed and weighed once a week for 2 weeks. Under the same conditions, the same batch of mice were fed with normal food and observed as the control group. The results of the experiment showed that all mice had any symptoms of poisoning, which proved that icariin is a non-toxic substance.
淫羊藿苷对感染流感病毒动物预防治疗实验:选择6周龄雄性BALB/c小鼠,每组小鼠分别接种50μL 102 LD50的H5N1病毒(由于H9N2对于小鼠致病性不稳定,所以本实验没有选择H9N2)(实验组1和实验组2),用PBS作阴性对照(实验组4),并设单独给以淫羊藿苷的对照组(实验组3),接种病毒的小鼠感染4小时后,设4个实验动物组,每组10只小鼠,每只小鼠口服给药1mg,相当于每天口服淫羊藿苷25毫克/千克体重,每天早晚2次,共14天,每天称量小鼠体重1次,观察小鼠体重变化和生存率情况。病理组织学检查:选择病死、治疗组、经口单纯给以淫羊藿苷和正常小鼠的肺组织用10%中性福尔马林(Neutral-buffered formalin)固定,然后修正和用不同浓度的乙醇脱水,二甲苯(Xylene,Sima)浸泡后,用石蜡(Paraffin,Sigma)镶嵌封固,切成3微米厚的薄片,放在载玻片上,用酒精连续进行脱蜡,用苏木精(Hematoxylin)和伊红(Eosin,Sigma)染色,在光学显微镜(Light microscopy)下观察。Icariin for the prevention of influenza virus infection in animals: 6-week-old male BALB/c mice were selected, each group was inoculated with 50 μL of 102 LD50 H5N1 virus (since H9N2 is pathogenic to mice, so this H9N2) was not selected in the experiment (experimental group 1 and experimental group 2), PBS was used as a negative control (experimental group 4), and a control group (experimental group 3) administered with icariin alone was infected with the virus-infected mouse. Four hours later, four experimental animal groups were set up, each group of 10 mice, each of which was orally administered with 1 mg, which was equivalent to oral administration of icariin 25 mg/kg body weight per day, twice a day for 14 days. The mice were weighed once a day, and the changes in body weight and survival rate were observed. Histopathological examination: Selecting the diseased, treatment group, oral administration of icariin and normal mouse lung tissue with 10% neutral-buffered formalin, then correcting and using different concentrations Dehydrated ethanol, soaked in xylene (Sima), embedded in paraffin (Paraffin, Sigma), cut into 3 micron thick slices, placed on glass slides, dewaxed continuously with alcohol, with hematoxylin (Hematoxylin) and Eosin (Sigma) staining were observed under light microscopy.
表2、动物实验分组实验情况Table 2, animal experiment group experiment
组别Group 接种病毒株Vaccination strain 经口给予淫羊藿苷Icariin PBS PBS
实验组1Experimental group 1 感染infection no no
实验组2 Experimental group 2 感染infection Yes no
实验组3 Experimental group 3 未感染Uninfected Yes no
实验组4 Experimental group 4 未感染Uninfected no Yes
淫羊藿苷抑制流感病毒的动物实验(实施例3),结果显示淫羊藿苷能够防护高致病性禽流感病毒(H5N1)的感染(图5)。另外,组织病理切片显示,淫羊藿苷还能促进肺组织免疫细胞浸润,提高肺组织的免疫防护作用(图6)。淫羊藿苷可以用于制备的淫羊藿苷及其衍生物治疗或预防由流感病毒感染导致的病理症状或疾病的药物、食品、保健品。An animal experiment in which icariin inhibits influenza virus (Example 3) showed that icariin was able to protect against infection with highly pathogenic avian influenza virus (H5N1) (Fig. 5). In addition, histopathological sections showed that icariin can also promote immune cell infiltration in lung tissue and improve the immune protection of lung tissue (Fig. 6). Icariin can be used in the preparation of icariin and its derivatives for the treatment or prevention of drugs, foods, and health products for pathological symptoms or diseases caused by influenza virus infection.
实施例4:胶囊型制剂Example 4: Capsule preparation
Figure PCTCN2015097322-appb-000006
Figure PCTCN2015097322-appb-000006
Figure PCTCN2015097322-appb-000007
Figure PCTCN2015097322-appb-000007
用于本发明实施例的淫羊藿0苷组合物可以是购买药品的淫羊藿苷,淫羊藿苷200g,粉碎成细粉。将该组分过120目筛,称取,加入上述表中量的淀粉和硬脂酸镁进行混合,然后倒入下料斗中。开造粒包衣机,入风压力O.6bar,入风温度30℃,喷枪压力(CYL)3b缸,雾化压力(CAP1)0.8bar,倒入250g空白丸芯,造粒,造粒速度4rpm,蠕泵12%,转盘转速145rpm,喷200g的7%PVP溶液(溶剂为90%乙醇)。造粒结束后,50℃烘干,出料。The icariin 0-side composition used in the examples of the present invention may be icariin for purchasing a drug, 200 g of icariin, and pulverized into a fine powder. The component was passed through a 120 mesh sieve, weighed, and the amount of starch and magnesium stearate added in the above table were mixed and poured into a lower hopper. Open granulation coating machine, inlet air pressure O.6bar, inlet air temperature 30 °C, spray gun pressure (CYL) 3b cylinder, atomization pressure (CAP1) 0.8 bar, pour 250g blank pellet core, granulation, granulation speed 4 rpm, 12% creep pump, 145 rpm rotary speed, spray 200 g of 7% PVP solution (solvent is 90% ethanol). After the granulation is finished, it is dried at 50 ° C and discharged.
将制得的小丸采用硬胶囊药物填充机按照每两粒胶囊中含淫羊藿苷的量分别为200mg进行填充制备含有淫羊藿苷的胶囊。The prepared pellets were filled with a capsule containing amyl saponin by using a hard capsule drug filling machine to fill 200 mg of icariin per two capsules, respectively.
实施例5片剂制备aExample 5 Tablet Preparation a
活性剂Active agent 添加量(g)Add amount (g)
淫羊藿苷Icariin 300300
   
辅料 Excipient  
木薯淀粉Cassava starch 3030
半乳糖 Galactose 2020
微晶纤维素 Microcrystalline cellulose 3030
制备工艺:称取淫羊藿苷200g,过100目筛,木薯淀粉、半乳糖、微晶纤维素过80目筛,混合均匀,加入100g的6%PVP无水乙醇溶液适量制粒,60℃干燥,18目筛整型干颗粒,干颗粒中加入2g的硬脂酸镁。Preparation process: weigh 200g of icariin, pass 100 mesh sieve, tapioca starch, galactose, microcrystalline cellulose over 80 mesh sieve, mix evenly, add 100g of 6% PVP absolute ethanol solution to prepare granules, 60 °C Dry, 18 mesh sieve dry pellets, 2 g of magnesium stearate was added to the dry pellets.
c、将上述a采用双层压片机冲压片即得每片含淫羊藿苷的量为200mg。c. The above a is punched with a double laminator to obtain 200 mg of icariin per tablet.
实施例6.混合型胶囊型控释制剂Example 6. Mixed capsule controlled release preparation
根据本发明设计口服给药的控释制剂需要挑选和评价制剂的体外释放特性和定时性,使其最适于获得想要的体内血浆特征,优选每日给药一次。因此,已经研究了单个单位骨架片的几种不同的制剂原理,即提供具有不同的释放度类型制剂。Designing a controlled release formulation for oral administration in accordance with the present invention requires selection and evaluation of the in vitro release profile and timing of the formulation, making it optimal for obtaining the desired in vivo plasma profile, preferably once daily. Thus, several different formulation principles for single unit matrix tablets have been investigated, namely providing formulations with different release profiles.
a、活性药芯的制备a, preparation of active drug core
活性剂Active agent 添加量(g)Add amount (g)
淫羊藿苷 Icariin 200200
   
辅料 Excipient  
淀粉starch 200200
硬脂酸镁 Magnesium stearate 33
黏合剂 Adhesive  
葡萄糖glucose 22
麦芽糊精 Maltodextrin 33
乙烯基醋酸酯共聚物Vinyl acetate copolymer 1.51.5
用于本发明实施例的淫羊藿苷组分或淫羊藿苷可以是购买药品,称取淫羊藿苷200g,粉碎成细粉。将该组分过120目筛,称取,加入上述表中量的淀粉、硬脂酸镁、葡萄糖、麦芽糊精和乙烯基醋酸酯共聚物进行混合,过250目筛,然后倒入下料斗中。开造粒包衣机,入风压力O.6bar,入风温度30℃,喷枪压力(CYL)3b缸,雾化压力(CAP1)0.8bar,倒入250g空白丸芯,造粒,造粒速度4rpm,蠕泵12%,转盘转速145rpm,喷200g的7%PVP溶液(溶剂为90%乙醇)。造粒结束后,50℃烘干,出料。The icariin component or icariin used in the examples of the present invention may be a purchased drug, and 200 g of icariin is weighed and pulverized into a fine powder. The component was passed through a 120 mesh sieve, weighed, and the amount of starch, magnesium stearate, glucose, maltodextrin and vinyl acetate copolymer added in the above table was mixed, passed through a 250 mesh sieve, and then poured into a lower hopper. in. Open granulation coating machine, inlet air pressure O.6bar, inlet air temperature 30 °C, spray gun pressure (CYL) 3b cylinder, atomization pressure (CAP1) 0.8 bar, pour 250g blank pellet core, granulation, granulation speed 4 rpm, 12% creep pump, 145 rpm rotary speed, spray 200 g of 7% PVP solution (solvent is 90% ethanol). After the granulation is finished, it is dried at 50 ° C and discharged.
b、包衣的制备b, preparation of coating
包衣组成Coating composition 添加量(g)Add amount (g)
甲基丙烯酸和醋酸乙酯共聚物Methacrylic acid and ethyl acetate copolymer 9090
甲基丙烯酸和甲基丙烯酸甲酯共聚物Methyl methacrylate and methyl methacrylate copolymer 135135
乙基纤维素 Ethyl cellulose 180180
柠檬酸三乙酯Triethyl citrate 450450
在室温,在流化床中,用丙酮/异丙醇/水混合物(54/36/10,重量百分比)溶解90g甲基丙烯酸和醋酸乙酯共聚物、135g甲基丙烯酸和甲基丙烯酸甲酯共聚物、180g乙基纤维素和45g柠檬酸三乙酯溶解,同时用该混合物来包覆450g的a中得到的细粒。喷射后得到包覆的微粒。平均直径为270μm。90 g of methacrylic acid and ethyl acetate copolymer, 135 g of methacrylic acid and methyl methacrylate were dissolved in a fluidized bed at room temperature with an acetone/isopropanol/water mixture (54/36/10, weight percent) The copolymer, 180 g of ethyl cellulose and 45 g of triethyl citrate were dissolved while using the mixture to coat 450 g of the fine particles obtained in a. The coated particles are obtained after spraying. The average diameter is 270 μm.
c、将a和b制得的小丸采用硬胶囊药物填充机按照包覆好的细粒进行填充制备胶囊,即得到含有0.5g淫羊藿苷的本发明的混合型控释胶囊制剂。c. The pellets prepared by a and b were filled with a hard capsule drug filling machine according to the coated fine particles to prepare a capsule, that is, a mixed controlled release capsule preparation of the present invention containing 0.5 g of icariin was obtained.
实施例7本发明控释片剂的制备Example 7 Preparation of Controlled Release Tablets of the Present Invention
a、a,
活性剂Active agent 添加量(g)Add amount (g)
淫羊藿苷 Icariin 200200
   
辅料 Excipient  
木薯淀粉Cassava starch 3030
半乳糖 Galactose 2020
微晶纤维素 Microcrystalline cellulose 3030
制备工艺:称取淫羊藿苷200g过100目筛,木薯淀粉、半乳糖、微晶纤维素过80目筛,混合均匀,加入100g的6%PVP无水乙醇溶液适量制粒,60℃干燥,18目筛整型干颗粒,干颗粒中加入2g的硬脂酸镁。Preparation process: weigh icariin 200g over 100 mesh sieve, tapioca starch, galactose, microcrystalline cellulose over 80 mesh sieve, mix well, add 100g 6% PVP absolute ethanol solution to granulate, dry at 60 ° C 18 mesh sieved dry granules, and 2 g of magnesium stearate was added to the dry granules.
b、包衣的制备b, preparation of coating
Figure PCTCN2015097322-appb-000008
Figure PCTCN2015097322-appb-000008
Figure PCTCN2015097322-appb-000009
Figure PCTCN2015097322-appb-000009
在室温,在流化床中,用丙酮/异丙醇/水混合物(54/36/10,重量百分比)溶解至90g甲基丙烯酸和醋酸乙酯共聚物、135g甲基丙烯酸和甲基丙烯酸甲酯共聚物、180g乙基纤维素和45g柠檬酸三乙酯溶解,同时用该混合物来包覆450g的a中得到的细粒。喷射后得到包覆的微粒。平均直径为270μm。为了获得多层的包衣,在原有的包衣基础上,再次进行喷射,反复进行上述步骤就可以得到所需的多层控释层。Dissolve in a fluidized bed at room temperature with an acetone/isopropanol/water mixture (54/36/10, weight percent) to 90 g of methacrylic acid and ethyl acetate copolymer, 135 g of methacrylic acid and methacrylic acid The ester copolymer, 180 g of ethyl cellulose and 45 g of triethyl citrate were dissolved while using this mixture to coat 450 g of the fine particles obtained in a. The coated particles are obtained after spraying. The average diameter is 270 μm. In order to obtain a multi-layer coating, the spray is again carried out on the basis of the original coating, and the above steps are repeated to obtain the desired multilayer controlled release layer.
c、将上述a,b两种组分采用双层压片机冲压片即得双层片,每片淫羊藿苷0.5g。c. The above two components a and b are punched by a double laminating machine to obtain a two-layer tablet, each of which is 0.5 g of icariin.
本发明的延长释放片剂使用可溶胀的和部分侵蚀性聚合物基质。基于假设的基质,释放曲线可大约按时间的平方根来描述样品的体外释放特性。取得不受pH值影响的体外释放曲线,则体内吸收时则比较不受胃肠道pH值的影响,有利于避免因胃肠道pH值不同而带来的个体吸收差异。本发明的聚合物其构成延长释放基质,缓慢地释放作为活性成分的淫羊藿苷及其衍生物与其组合物。给药后,制剂与含水液体接触后,聚合物溶胀,形成粘性的凝胶层来调节药物释放。聚合物的黏度优选范围是从180至110,000mPa.s(在20℃下2%水溶液的表观粘度)。The extended release tablets of the present invention utilize a swellable and partially aggressive polymeric matrix. Based on the hypothetical matrix, the release profile can describe the in vitro release profile of the sample approximately at the square root of time. Obtaining the in vitro release profile that is not affected by the pH value is not affected by the pH value of the gastrointestinal tract when absorbed in the body, and is beneficial to avoid the difference in individual absorption due to the difference in pH of the gastrointestinal tract. The polymer of the present invention constitutes an extended release matrix, and slowly releases icariin as a active ingredient and derivatives thereof and combinations thereof. After administration, after contact with the aqueous liquid, the polymer swells to form a viscous gel layer to regulate drug release. The viscosity of the polymer preferably ranges from 180 to 110,000 mPa.s (apparent viscosity of a 2% aqueous solution at 20 ° C).
我们将本发明实施例6或7制备的控释样品在不同pH值下同时进行释放度试验,并根据释放实验数据绘制了释放试验曲线.如图7所示,可以看出实施例7制备的样品在各pH值介质中有一致的释放曲线,其释放几乎不受介质pH值的影响。The controlled release samples prepared in Example 6 or 7 of the present invention were simultaneously subjected to a release test at different pH values, and a release test curve was drawn based on the release test data. As shown in FIG. 7, the preparation of Example 7 can be seen. The sample has a consistent release profile in each pH medium and its release is virtually unaffected by the pH of the medium.
实施例8本发明组合物的注射剂型的制备Example 8 Preparation of Injectable Formulations of the Compositions of the Invention
配方:10g淫羊藿苷Formula: 10g icariin
20g甘露醇20g mannitol
称取淫羊藿苷10g,用注射用水配制成1000ml溶液,无菌过滤,按每支1ml灌装,冷冻干燥,压塞,轧盖得到成品注射液。Weigh 10g of icariin, prepare 1000ml solution with water for injection, sterilely filter, fill with 1ml each, freeze-dry, tampon, and cover to obtain the finished injection.
实施例9:涂覆片剂制备Example 9: Preparation of coated tablets
Figure PCTCN2015097322-appb-000010
Figure PCTCN2015097322-appb-000010
Figure PCTCN2015097322-appb-000011
Figure PCTCN2015097322-appb-000011
本发明的有益效果是:淫羊藿苷能够抑制流感病毒神经氨酸酶的活性,从而抑制流感病毒的复制,可用于流感病毒感染而导致的各种疾病的预防和治疗药物、食品、保健品的开发。The beneficial effects of the present invention are: icariin can inhibit the activity of influenza virus neuraminidase, thereby inhibiting the replication of influenza virus, and can be used for the prevention and treatment of various diseases caused by influenza virus infection, food, health care products. Development.
本发明不局限在本申请中描述的特定实施方式,用作本发明的单独的方面的单一说明。本领域技术人员将理解,可以不脱离本申请的精神和范围的情况下进行各种修改和改动。根据以上描述,除了本文中列举的以外,本公开的范围内的功能上等同的用途对于本领域的本领域技术人员而言是明显的。这样的改动和修改意欲落在所附权利要求的范围内。本公开仅受所附权利要求以及与这样的权利要求的的范围所等同的全部范围限制。应当理解,本公开不局限于特定的方法、试剂、组合物和生物系统,当然,所述方法、试剂、组合物和生物系统可以变化。还可理解,本文中使用的术语仅用于描述特定的实施方式,不用来是限制性的。The invention is not limited to the specific embodiments described in the application, and is used as a single description of the individual aspects of the invention. A person skilled in the art will appreciate that various modifications and changes can be made without departing from the spirit and scope of the application. Functionally equivalent uses within the scope of the present disclosure will be apparent to those skilled in the art from this disclosure. Such modifications and variations are intended to fall within the scope of the appended claims. The present disclosure is only limited by the scope of the appended claims and the scope of the claims. It should be understood that the present disclosure is not limited to specific methods, reagents, compositions, and biological systems, of course, the methods, reagents, compositions, and biological systems may vary. It is also understood that the terminology used herein is for the purpose of describing the particular embodiments
此外,在以马库什组的方式描述本公开的特征或方面的情况下,本领域的本领域技术人员将意识到,本公开也是以马库什组中的任一单个成员或亚组成员的方式所描述的。Moreover, where features or aspects of the disclosure are described in the form of a Markush group, those skilled in the art will recognize that the present disclosure is also a single member or subgroup member of the Markush group. The way it is described.
本文所参考的或引用的所有专利、专利申请、在先申请和出版物通过引用而全文并入本文,包括所有的附图和表格,使得它们不与本说明书的明确教导相矛盾。在权利要求范围内提出其他的实施方式。 All of the patents, patent applications, prior applications and publications are hereby incorporated by reference in their entirety in their entirety in their entireties in the entireties in Other embodiments are proposed within the scope of the claims.

Claims (54)

  1. 一种流感病毒神经氨酸酶的新型抑制剂,其特征在于所述抑制剂为淫羊藿苷及其衍生物,且所述淫羊藿苷及其衍生物分别为下式(I)和(II)所定义、包括其单独的异构体、异构体的外消旋或非外消旋混合物、氧化物、或其药学上可接受的盐和水合物,A novel inhibitor of influenza virus neuraminidase characterized in that the inhibitor is icariin and a derivative thereof, and the icariin and its derivatives are respectively of the following formula (I) and II) as defined, including its individual isomers, racemic or non-racemic mixtures of isomers, oxides, or pharmaceutically acceptable salts and hydrates thereof,
    Figure PCTCN2015097322-appb-100001
    Figure PCTCN2015097322-appb-100001
    其中,Xa、Xb、Xc、Xd、Xe、Xf、Xg、Xh、Xi、X1、X2、X3、X4、X5、X6和X7独立为氧原子(O)或硫原子(S);Wherein Xa, Xb, Xc, Xd, Xe, Xf, Xg, Xh, Xi, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 and X 7 are independently an oxygen atom (O) or a sulfur atom. (S);
    Ra、Rb、Rc、Rd、Re、Rf、Rg、Rh和Ri独立为氢原子(H)、1-5Ry替代的烷基(C1-C10)或(C1-C10)-O-(C1-C10)、每一个烷基(C1-C10)用1-5个Ry取代,每一个Ry是Rq、或(C2-C10)烷基、(C3-C10)烷基、(C3-C10)环烷基烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、—(C5-C10)环烷基烷基、(C3-C10)环烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、苯基、萘基、(C14) 芳香基团,它们的每一个可以用一个或更多的Rz取代,每一个Rz是(C1-C6)烷基、(C2–C6)烯基、(C1–C6)炔基、(C3–C8)环烷基、(C6-C8)环烯基、苯基、3-到5-元)杂环、C(卤素)3或CH(卤素)2;R1、R2、R2、R4、R5、R6、R7和R8独立为氢原子(H),或(C1-C10)烷基、(C2-C10)烯基、(C2-C10)炔基、(C3-C10)环烷基、(C8-C14)二环烷基烷基、(C8-C14)三环烷基烷基、苯基、萘基、(C14)芳香基团,它们的每一个可以是非取代的、一个或更多的Rz或Rq取代的、一个或更多的带有1个以上Rz取代的Rq替换,每一个Rq可以是CN、OH、卤素、N3、NO2、N(Rz)2、═NRz、CH═NRz、NRz OH、ORz、CORz、C(O)ORz、OC(O)Rz、OC(O)ORz、SRz、S(O)Rz或S(O)2Rz;Y1、Y2和Y3独立为CH3、(C1-C12)烷基、也可以是用Ry组合取代的(C1-C12)烷基,每一个(C1-C12)烷基可以用1-5个Ry取代或一个平衡离子取代。Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh and Ri are independently a hydrogen atom (H), a 1-5Ry-substituted alkyl group (C 1 -C 10 ) or (C 1 -C 10 )-O- (C 1 -C 10 ), each alkyl group (C 1 -C 10 ) is substituted with 1-5 Ry, each Ry is Rq, or (C 2 -C 10 )alkyl, (C 3 -C 10 An alkyl group, (C 3 -C 10 )cycloalkylalkyl group, (C 8 -C 14 )bicycloalkylalkyl group, (C 8 -C 14 )tricycloalkylalkyl group, —(C 5 - C 10 ) cycloalkylalkyl, (C 3 -C 10 )cycloalkyl, (C 8 -C 14 )bicycloalkylalkyl, (C 8 -C 14 )tricycloalkylalkyl, phenyl , naphthyl, (C 14 ) aromatic groups, each of which may be substituted with one or more Rz, each Rz being (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 1 -C 6 )alkynyl, (C 3 -C 8 )cycloalkyl, (C 6 -C 8 )cycloalkenyl, phenyl, 3- to 5-membered heterocyclic ring, C(halogen) 3 Or CH(halogen) 2 ; R 1 , R 2 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 are independently a hydrogen atom (H), or a (C 1 -C 10 )alkyl group, ( C 2 -C 10 )alkenyl, (C 2 -C 10 )alkynyl, (C 3 -C 10 )cycloalkyl, (C 8 -C 14 )bicycloalkylalkyl, (C 8 -C 14 ) tricyclo alkyl, phenyl, naphthyl, (C 14) Fragrance groups, each of which may be unsubstituted, substituted by one or more Rz or Rq, one or more Rq substitutions with more than one Rz substitution, each Rq may be CN, OH, halogen , N 3 , NO 2 , N(Rz) 2 , ═NRz, CH═NRz, NRz OH, ORz, CORz, C(O)ORz, OC(O)Rz, OC(O)ORz, SRz, S(O Rz or S(O) 2 Rz; Y 1 , Y 2 and Y 3 are independently CH 3 , (C 1 -C 12 )alkyl, or (C 1 -C 12 )alkyl substituted with a combination of Ry Each (C 1 -C 12 )alkyl group may be substituted with 1-5 Ry or a counter ion.
  2. 如权利要求1所述的新型抑制剂,其特征在于所述流感病毒包括流感病毒甲型、乙型和丙型及其各种亚型、血清型、基因型、基因突变体病毒。The novel inhibitor according to claim 1, characterized in that the influenza virus comprises influenza virus A, B and C and various subtypes thereof, serotypes, genotypes, gene mutant viruses.
  3. 如权利要求2所述的抑制剂,其特征在于所述流感病毒为甲型流感病毒。The inhibitor according to claim 2, wherein the influenza virus is an influenza A virus.
  4. 如权利要求3所述的抑制剂,其特征在于所述流感病毒为H5N1或H9N2病毒。The inhibitor according to claim 3, characterized in that the influenza virus is an H5N1 or H9N2 virus.
  5. 如权利要求1所述的抑制剂,其特征在于与奥司他韦相比,淫羊藿苷及其衍生物对流感病毒神经氨酸酶活性具有高近40倍的抑制作用。The inhibitor according to claim 1, characterized in that icariin and its derivative have a nearly 40-fold inhibitory effect on influenza virus neuraminidase activity compared to oseltamivir.
  6. 一种流感病毒神经氨酸酶的新型抑制剂的筛选方法,其特征在于所述方法包括将淫羊藿苷及其衍生物作为流感病毒神经氨酸酶的抑制剂,添加到细胞或动物体内。A method for screening novel inhibitors of influenza virus neuraminidase, characterized in that the method comprises adding icariin and a derivative thereof as an inhibitor of influenza virus neuraminidase to a cell or an animal.
  7. 如权利要求6所述的方法,其特征在于所述细胞为MDCK细胞。The method of claim 6 wherein said cells are MDCK cells.
  8. 如权利要求6所述的方法,其特征在于所述动物为鸡或小鼠。The method of claim 6 wherein said animal is a chicken or a mouse.
  9. 如权利要求1-5任一项所述的抑制剂在制备用于预防和/或治疗流感病毒引起的人或动物病理症状或疾病的药物、食品、保健品或化妆品中的用途。Use of the inhibitor according to any one of claims 1 to 5 for the preparation of a medicament, food, health supplement or cosmetic for preventing and/or treating a human or animal pathological condition or disease caused by influenza virus.
  10. 如权利要求9所述的用途,其特征在于所述淫羊藿苷来自于天然的淫羊藿苷植物,是从小檗科(Berberidaceae)淫羊藿属(Epimedium)植物提取纯化而成的,所述淫羊藿属植物包括由宝兴淫羊藿(E.davidii)、保靖淫羊藿(E.baojingense)、朝鲜淫羊藿朝鲜淫羊藿(E.icoroanum)、川鄂淫羊藿(E.fargesii)、川西淫羊藿(E.elongatum)、粗毛淫羊藿(E.acuminatum)、单叶淫羊藿(E.simplicifolium)、短茎淫羊藿(E.brachyrrhizum)、多花淫羊藿(E.multiflorum)、恩施淫羊藿(E.enshiense)、革叶淫羊藿(E.reticulatum)、光叶淫羊藿(E.sagittatum)、宽萼淫羊藿(E.latisepalum)、绿药淫羊藿(E.chlorandrum)、茂汶淫羊藿(E.platypetalum)、偏斜淫羊藿(E.truncatum)、强茎淫羊藿(E.rhizomatosum)、三枝九叶草(E.sagittatum)、少花淫羊藿(E.pauciflorum)、天平山淫羊藿(E.myrianthum)、无距淫羊藿(E.ecalcaratum)、腺毛淫羊藿(E.glandulosopilosum)、小叶淫羊藿(E.parvifolium)、星花淫羊藿(E.stellulatum)、镇坪淫羊藿(E.ilicifolium)组成的组的一种或多种混合物。The use according to claim 9, wherein the icariin is derived from a natural icariin plant and is obtained by extracting and purifying a plant of the genus Epimedium of the genus Berberidaceae. The genus Epimedium includes E. davidii, E. baojingense, E. icoroanum, and E. icoroum. E. fargesii), E. elongatum, E. acuminatum, E. simplicifolium, E. brachyrrhizum, and more E. multiflorum, E. enshiense, E. reticulatum, E. sagittatum, E. latisepalum E.chlorandrum, E.platypetalum, E. truncatum, E. rhizomatosum, and three-leaf clover (E.rhizomatosum) E.sagittatum), E. pauciflorum, E. myrianthum, E. ecalcaratum, E. glandulosopilosum, leaflets Epimedium (E.parvifolium), star flower Epimedium (E.stellulatum), one of the group of Epimedium Zhenping (E.ilicifolium) consisting of a mixture of one or more.
  11. 如权利要求9或10所述的用途,其特征在于所述流感病毒包括流感病毒甲型、乙型和丙型及其各种亚型、血清型、基因型、基因突变体病毒。The use according to claim 9 or 10, characterized in that the influenza virus comprises influenza virus A, B and C and various subtypes thereof, serotypes, genotypes, gene mutant viruses.
  12. 如权利要求11所述的用途,其特征在于所述流感病毒为甲型流感病毒。 The use according to claim 11, characterized in that the influenza virus is an influenza A virus.
  13. 如权利要求12所述的用途,其特征在于所述流感病毒为H5N1或H9N2病毒。The use according to claim 12, characterized in that the influenza virus is an H5N1 or H9N2 virus.
  14. 如权利要求9或10所述的用途,其特征在于所述的病理症状或疾病包括咳嗽、发烧、喷嚏、鼻塞、流清水样鼻涕、畏寒、头痛、乏力、全身酸痛、肺炎、扁桃体炎、咽炎、喉炎、流泪、声音嘶哑、呼吸综合症、心肺症、四肢冰冷,恶心呕吐、干咳、口干舌燥、头昏欲睡、腹泻、感冒流产、心肌炎、高血压、心肌梗塞、脑中风、鼻腔黏膜充血、水肿、有分泌物、咽部轻度充血、颌下淋巴结肿大、咽部充血水肿、感冒综合征组成的组的一种或多种的疾病。The use according to claim 9 or 10, characterized in that the pathological symptoms or diseases include cough, fever, sneezing, nasal congestion, runny nose, chills, headache, fatigue, body aches, pneumonia, tonsillitis, Pharyngitis, laryngitis, tearing, hoarseness, respiratory syndrome, cardiopulmonary disease, cold limbs, nausea and vomiting, dry cough, dry mouth, drowsiness, diarrhea, cold abortion, myocarditis, hypertension, myocardial infarction, stroke One or more diseases of the group consisting of nasal mucosa congestion, edema, secretion, mild hyperemia of the pharynx, enlarged submandibular lymph nodes, pharyngeal congestion and edema, and cold syndrome.
  15. 如权利要求14所述的用途,其特征在于所述的病理症状或疾病包括咳嗽、发烧、鼻塞、畏寒、头痛、乏力、全身酸痛、流泪、四肢冰冷,恶心呕吐、头昏欲睡、水肿、感冒综合征组成的组的一种或多种的疾病。The use according to claim 14, characterized in that the pathological symptoms or diseases include cough, fever, stuffy nose, chills, headache, fatigue, body aches, tears, cold limbs, nausea and vomiting, drowsiness, edema One or more diseases of the group consisting of cold syndrome.
  16. 如权利要求9或10所述的用途,其特征在于将所述淫羊藿苷及其衍生物通过口服、外用、吸入、经鼻、经直肠、经皮或注射途径对受试者给药。The use according to claim 9 or 10, characterized in that the icariin and its derivative are administered to a subject by oral, topical, inhalation, nasal, rectal, transdermal or injection routes.
  17. 如权利要求16所述的用途,其特征在于将所述淫羊藿苷及其衍生物通过口服或注射途径对受试者给药。The use according to claim 16, wherein the icariin and a derivative thereof are administered to a subject by an oral or injection route.
  18. 如权利要求16或17所述的用途,其特征在于所述受试者包括人、农场动物、宠物动物、实验室动物在内的动物。The use according to claim 16 or 17, wherein the subject comprises an animal such as a human, a farm animal, a pet animal, or a laboratory animal.
  19. 如权利要求18所述的用途,其特征在于所述受试者为人。The use according to claim 18, characterized in that the subject is a human.
  20. 如权利要求16-19任一项所述的用途,其特征在于所述受试者已经患有或正在患有或处于流感病毒感染的病理症状或疾病的风险。The use according to any of claims 16-19, characterized in that the subject already has or is at risk of having or being at a pathological condition or disease of an influenza virus infection.
  21. 如权利要求20所述的用途,其特征在于人已经患有流感病毒感染的病理症状或疾病。The use according to claim 20, characterized in that the human has already suffered from a pathological symptom or disease of influenza virus infection.
  22. 如权利要求9-21任一项所述的用途,其特征在于对所述的受试者施用含有预防和/或治疗的有效量的权利要求1所述的淫羊藿苷及其衍生物。The use according to any one of claims 9 to 21, wherein the subject is administered an effective amount of the icariin of claim 1 and a derivative thereof according to the prophylactic and/or therapeutic effect.
  23. 如权利要求22所述的用途,其特征在于所述有效量包括对人每天施用约0.1mg/kg至约10000mg/kg体重的淫羊藿苷及其衍生物。The use according to claim 22, wherein the effective amount comprises administering from about 0.1 mg/kg to about 10000 mg/kg of body weight of icariin and a derivative thereof per day to a human.
  24. 如权利要求23所述的用途,其特征在于所述有效量包括对人每天施用约0.5mg/kg至约200mg/kg体重的淫羊藿苷及其衍生物。The use according to claim 23, wherein the effective amount comprises administering icariin and a derivative thereof to the human daily from about 0.5 mg/kg to about 200 mg/kg of body weight.
  25. 如权利要求24所述的用途,其特征在于所述有效量包括对人每天施用25mg/kg体重的淫羊藿苷及其衍生物。The use according to claim 24, wherein the effective amount comprises administering 25 mg/kg body weight of icariin and a derivative thereof to a human daily.
  26. 如权利要求9-25任一项所述的用途,其特征在于对所述的受试者使用施用1至6次淫羊藿苷及其衍生物。Use according to any of claims 9-25, characterized in that icariin and its derivatives are administered 1 to 6 times to the subject.
  27. 如权利要求26所述的用途,其特征在于对所述的受试者使用施用2次或3次淫羊藿苷及其衍生物。The use according to claim 26, characterized in that icariin and a derivative thereof are administered twice or three times to said subject.
  28. 含有权利要求1所述新型抑制剂的药物组合物,其特征在于所述药物组合物含有至少两种活性成分,其中至少一种活性成分为淫羊藿苷及其衍生物。A pharmaceutical composition comprising the novel inhibitor of claim 1, characterized in that the pharmaceutical composition contains at least two active ingredients, at least one of which is icariin and a derivative thereof.
  29. 如权利要求28所述的药物组合物,其中所述至少另外一种活性成分为利巴韦林(Ribavirin1)、干扰素(Interferon)、胸腺肽α(Thymosinα)、转移因子(Transfer factor)、齐多夫定(zidovudine, ZDV)、地丹诺辛(didanosine)、拉米夫定(lamivudine)、斯塔夫定(stavudine)、扎西他宾(zalcitabine)、abacavir、病毒唑(Ribavirin)、(三氮畦唑核苷(Virazole)、奈韦拉平(nevirapine)、地拉夫定(delavirdine)、efavirenz、沙奎那韦(saquinavir)、利托那韦(ritonavir)、奈非那韦(nelfinavir)、英地那韦(indinavir)、安泼那韦(amprenavir)、Lopinavir、金刚烷胺(Amantadine)、金刚乙胺(Rimantadine)、扎那米韦(Zanamivir)、阿昔洛韦(Acyclovir)、伐昔洛韦(Valacyclovir)、碘苷(Idoxuridine)、阿糖腺苷(Vidarabine)、拉米夫定(Lamivudine)、泛昔洛韦(Famciclovir)、喷昔洛韦(Penciclovir)、奥司他韦和扎那米韦。The pharmaceutical composition according to claim 28, wherein said at least one other active ingredient is Ribavirin 1 , Interferon, Thymosin α, Transfer factor, Qiduo Zidovudine ZDV), didanosine, lamivudine, stavudine, zalcitabine, abacavir, ribavirin, ribavirin (Virazole), nevirapine, delavirdine, efavirenz, saquinavir, ritonavir, nelfinavir, indinavir, Amprenavir, Lopinavir, Amantadine, Rimantadine, Zanamivir, Acyclovir, Valacyclovir, Iodine (Idoxuridine), Vidarabine, Lamivudine, Famciclovir, Penciclovir, oseltamivir and zanamivir.
  30. 如权利要求28所述的所述的药物组合物,其中所述至少另外一种中药活性成分为板蓝根、大青叶、银翘、连翘、荆芥、金银花、鱼腥草、野菊花、夏桑菊、柴胡、穿心莲等中药成分的一种或多种组合。The pharmaceutical composition according to claim 28, wherein the at least one other traditional Chinese medicine active ingredient is Radix Isatidis, Folium Isatidis, Yinqiao, Forsythia, Nepeta, Honeysuckle, Houttuynia, Wild Chrysanthemum, Summer Sangju One or more combinations of traditional Chinese medicine ingredients such as Bupleurum and Andrographis paniculata.
  31. 如权利要求28-30任一项所述的组合物在制备用于预防和/或治疗流感病毒引起的人或动物病理症状或疾病的药物、食品、保健品或化妆品中的用途。Use of a composition according to any one of claims 28 to 30 for the manufacture of a medicament, food, nutraceutical or cosmetic for the prevention and/or treatment of pathological conditions or diseases of human or animal caused by influenza virus.
  32. 如权利要求31所述的用途,其特征在于所述流感病毒包括流感病毒甲型、乙型和丙型及其各种亚型、血清型、基因型、基因突变体病毒。The use according to claim 31, characterized in that the influenza virus comprises influenza virus A, B and C and various subtypes thereof, serotypes, genotypes, gene mutant viruses.
  33. 如权利要求32所述的用途,其特征在于所述流感病毒为甲型流感病毒。The use according to claim 32, characterized in that the influenza virus is an influenza A virus.
  34. 含有如权利要求1所述的新型抑制剂或如权利要求28-30所述药物组合物的胶囊、滴丸、凝胶剂、片剂、粉剂、颗粒剂、含片、泡腾片、糖浆、乳化物、控释制剂、快速溶解的制备物、口服液体的制剂。A capsule, a dropping pill, a gel, a tablet, a powder, a granule, a lozenge, an effervescent tablet, a syrup, or the like, comprising the novel inhibitor of claim 1 or the pharmaceutical composition according to claims 28-30, Emulsions, controlled release formulations, fast-dissolving preparations, oral liquid formulations.
  35. 如权利要求34所述的制剂,其特征在于所述制剂还含有辅料,所述辅料包括但不局限于润滑剂和助溶剂,如硬脂酸镁、硬脂酸钙、硬脂酸锌、硬脂酸、山嵛酸甘油酯、十八烷基富马酸钠、滑石、硅胶;崩解剂,如淀粉、环糊精、羧甲基纤维素、交联羧甲基纤维素、交联聚乙烯吡咯烷酮、明胶;稀释剂或压缩剂,如乳糖、蔗糖、甘露醇木糖醇、赤藻糖醇、山梨糖醇、微晶纤维素;调味料或其它成分,如草莓、桔子、香蕉、薄荷、蜂蜜;助溶剂,如酒类(啤酒、果酒、米酒、白酒)、植物油、动物油、乙醇、异丙醇、甘油、丙二醇、苯甲醇、二甲基亚风、聚乙二醇、阿拉伯胶、卵磷脂、吡咯酮、油酸、氮酮。The formulation of claim 34, wherein said formulation further comprises adjuvants including, but not limited to, lubricants and co-solvents such as magnesium stearate, calcium stearate, zinc stearate, hard Fatty acid, glyceryl behenate, sodium stearyl fumarate, talc, silica gel; disintegrants such as starch, cyclodextrin, carboxymethylcellulose, croscarmellose, cross-linked poly Vinyl pyrrolidone, gelatin; diluent or compressive agent such as lactose, sucrose, mannitol xylitol, erythritol, sorbitol, microcrystalline cellulose; seasonings or other ingredients such as strawberries, oranges, bananas, mint , honey; cosolvents, such as alcohol (beer, fruit wine, rice wine, white wine), vegetable oil, animal oil, ethanol, isopropanol, glycerin, propylene glycol, benzyl alcohol, dimethyl sulfite, polyethylene glycol, gum arabic, Lecithin, pyrrolidone, oleic acid, azone.
  36. 如权利要求34或35所述的制剂,其特征在于所述的剂型为控释制剂。A formulation according to claim 34 or 35, wherein the dosage form is a controlled release formulation.
  37. 如权利要求36所述的制剂,其特征在于所述控释制剂包括核芯和包裹核芯的控释层。The formulation of claim 36 wherein said controlled release formulation comprises a core and a controlled release layer that encapsulates the core.
  38. 含有权利要求34-37任一项所述的制剂,其中所述核芯含有至少两种活性成分,其中至少一种活性成分为淫羊藿苷及其衍生物。A formulation according to any one of claims 34 to 37, wherein the core comprises at least two active ingredients, at least one of which is icariin and a derivative thereof.
  39. 如权利要求34-38任一项所述的制剂,其中所述核芯还可以包含至少另外一种活性成分为干扰素(Interferon)、胸腺肽α(Thymosinα)、病毒唑(Ribavirin)、金刚烷胺(Amantadine)、金刚乙胺(Rimantadine)奥司他韦和扎那米韦。The preparation according to any one of claims 34 to 38, wherein the core may further comprise at least one other active ingredient: Interferon, Thymosin alpha, Ribavirin, Amantadine (Amantadine), Rimantadine oseltamivir and zanamivir.
  40. 如权利要求34-38任一项所述的制剂,其中所述核芯还可以含有至少另外一种中药活性成分为板蓝根、大青叶、连翘、金银花、柴胡、穿心莲等中药成分的一种或多种组合。The preparation according to any one of claims 34 to 38, wherein the core may further comprise at least one other traditional Chinese medicine active ingredient which is a traditional Chinese medicine ingredient such as Radix, Radix, Forsythia, Honeysuckle, Bupleurum, Andrographis paniculata One or more combinations.
  41. 如权利要求34-40任一项所述的控释制剂,其特征在于所述所述控释层含有甲基丙烯酸和醋酸乙酯共聚物、甲基丙烯酸和甲基丙烯酸甲酯共聚物、乙基纤维素、柠檬酸三乙酯。 The controlled release preparation according to any one of claims 34 to 40, wherein said controlled release layer contains a copolymer of methacrylic acid and ethyl acetate, a copolymer of methacrylic acid and methyl methacrylate, and Cellulose, triethyl citrate.
  42. 如权利要求34-41任一项所述的控释制剂,其特征在于:如权利要求1所述的控释制剂,其特征在于,按照重量比,所述核芯和包裹核芯的控释层的比例为1:0.01-100。The controlled release preparation according to any one of claims 1 to 4, wherein the controlled release preparation according to claim 1, wherein the core and the coated core are controlled release according to a weight ratio The ratio of the layers is 1:0.01-100.
  43. 如权利要求34-42任一项权利要求所述的控释制剂,其特征在于:所述制剂在pH 1至10.0范围内提供与pH无关的体外释放特。A controlled release formulation according to any one of claims 34 to 42 wherein the formulation provides a pH independent release in vitro in the range of pH 1 to 10.0.
  44. 如权利要求34-43任一项任一项所述的制剂在制备用于预防和/或治疗流感病毒引起的人或动物病理症状或疾病的药物、食品、保健品或化妆品中的用途。Use of a formulation according to any one of claims 34-43 for the manufacture of a medicament, food, nutraceutical or cosmetic for the prevention and/or treatment of pathological conditions or diseases of human or animal caused by influenza virus.
  45. 如权利要求44所述的用途,其特征在于所述流感病毒包括流感病毒甲型、乙型和丙型及其各种亚型、血清型、基因型、基因突变体病毒。The use according to claim 44, characterized in that the influenza virus comprises influenza virus A, B and C and various subtypes thereof, serotypes, genotypes, gene mutant viruses.
  46. 如权利要求45所述的用途,其特征在于所述流感病毒为甲型流感病毒。The use according to claim 45, characterized in that the influenza virus is an influenza A virus.
  47. 如权利要求44-46任一项所述的用途,其特征在于所述的病理症状或疾病包括咳嗽、发烧、喷嚏、鼻塞、流清水样鼻涕、畏寒、头痛、乏力、全身酸痛、肺炎、扁桃体炎、咽炎、喉炎、流泪、声音嘶哑、呼吸综合症、心肺症、四肢冰冷,恶心呕吐、干咳、口干舌燥、头昏欲睡、腹泻、感冒流产、心肌炎、高血压、心肌梗塞、脑中风、鼻腔黏膜充血、水肿、有分泌物、咽部轻度充血、颌下淋巴结肿大、咽部充血水肿、感冒综合征组成的组的一种或多种的疾病。The use according to any one of claims 44 to 46, wherein the pathological symptom or disease comprises cough, fever, sneezing, nasal congestion, runny nose, chills, headache, fatigue, body aches, pneumonia, Tonsillitis, pharyngitis, laryngitis, tearing, hoarseness, respiratory syndrome, cardiopulmonary disease, cold limbs, nausea and vomiting, dry cough, dry mouth, drowsiness, diarrhea, cold abortion, myocarditis, hypertension, myocardial infarction One or more diseases of the group consisting of brain stroke, nasal mucosal congestion, edema, secretion, mild hyperemia of the pharynx, enlarged submandibular lymph nodes, pharyngeal congestion and edema, and cold syndrome.
  48. 如权利要求47所述的用途,其特征在于所述的病理症状或疾病包括咳嗽、发烧、鼻塞、畏寒、头痛、乏力、全身酸痛、流泪、四肢冰冷,恶心呕吐、头昏欲睡、水肿、感冒综合征组成的组的一种或多种的疾病。The use according to claim 47, characterized in that the pathological symptoms or diseases include cough, fever, stuffy nose, chills, headache, fatigue, body aches, tears, cold limbs, nausea and vomiting, drowsiness, edema One or more diseases of the group consisting of cold syndrome.
  49. 如权利要求44-48任一项所述的用途,其特征在于对所述的受试者每日施用1次所述控释制剂。The use according to any one of claims 44 to 48, wherein said controlled release preparation is administered to said subject once a day.
  50. 如权利要求49所述的用途,其特征在于所述受试者包括人、农场动物、宠物动物、实验室动物在内的动物。The use according to claim 49, wherein the subject comprises an animal such as a human, a farm animal, a pet animal, or a laboratory animal.
  51. 如权利要求50所述的用途,其特征在于所述受试者为人。The use according to claim 50, characterized in that the subject is a human.
  52. 如权利要求44-51任一项所述的用途,其特征在于所述受试者已经患有或正在患有或处于流感病毒感染的病理症状或疾病的风险。Use according to any of claims 44-51, characterized in that the subject already has or is at risk of having or being at a pathological condition or disease of influenza virus infection.
  53. 如权利要求52所述的用途,其特征在于人已经患有流感病毒感染的病理症状或疾病。The use according to claim 52, characterized in that the human has already suffered from a pathological symptom or disease of influenza virus infection.
  54. 一种制备控释制剂的方法,其特征在于,所述的制备方法包括如下至少一个步骤:制备核芯步骤和制备至少一层包衣的缓释层步骤。 A method of preparing a controlled release preparation, characterized in that the preparation method comprises at least one of the steps of: preparing a core step and preparing a sustained release layer of at least one layer of coating.
PCT/CN2015/097322 2015-01-08 2015-12-15 New inhibitor for influenza virus neuraminidase and uses thereof WO2016110173A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510012661 2015-01-08
CN201510012661.0 2015-01-08

Publications (1)

Publication Number Publication Date
WO2016110173A1 true WO2016110173A1 (en) 2016-07-14

Family

ID=53903476

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/097322 WO2016110173A1 (en) 2015-01-08 2015-12-15 New inhibitor for influenza virus neuraminidase and uses thereof

Country Status (2)

Country Link
CN (1) CN104857007A (en)
WO (1) WO2016110173A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112022810A (en) * 2019-06-03 2020-12-04 鲁南制药集团股份有限公司 Icaritin pharmaceutical composition and preparation method thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105727269B (en) * 2016-03-22 2018-10-02 中国药科大学 The application of neuraminidase and its inhibitor in myocardial ischemia and myocardial infarction

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1732925A (en) * 2005-08-02 2006-02-15 天津药物研究院 Sustained release minipills of lovastatin and its preparation process
CN1969870A (en) * 2005-11-24 2007-05-30 北京奇源益德药物研究所 Antivirus compound formulation, preparation process, quality control method and use thereof
CN103621567A (en) * 2013-11-19 2014-03-12 青岛茂丰有机蔬菜有限公司 Drug for preventing and treating gummosis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100404049C (en) * 2004-12-31 2008-07-23 中国医学科学院药用植物研究所 Medicine for preventing and treating bird flu and its preparation process
CN103083670A (en) * 2012-10-26 2013-05-08 通化新海生物产业有限公司 Application of inhibiting agent of cathepsin K
CN103265591B (en) * 2013-05-24 2016-04-20 四川大学 Two derivatives from icariin and investigation and its production and use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1732925A (en) * 2005-08-02 2006-02-15 天津药物研究院 Sustained release minipills of lovastatin and its preparation process
CN1969870A (en) * 2005-11-24 2007-05-30 北京奇源益德药物研究所 Antivirus compound formulation, preparation process, quality control method and use thereof
CN103621567A (en) * 2013-11-19 2014-03-12 青岛茂丰有机蔬菜有限公司 Drug for preventing and treating gummosis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
XIE, JUANPING ET AL.: "Advances in Studies on Chemical Constituents and Pharmaceuticals in Species of Epimedium", STRAIT PHARMACEUTICAL JOURNAL, vol. 18, no. 5, 31 May 2006 (2006-05-31), pages 19 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112022810A (en) * 2019-06-03 2020-12-04 鲁南制药集团股份有限公司 Icaritin pharmaceutical composition and preparation method thereof
CN112022810B (en) * 2019-06-03 2022-05-24 鲁南制药集团股份有限公司 Icaritin pharmaceutical composition and preparation method thereof

Also Published As

Publication number Publication date
CN104857007A (en) 2015-08-26

Similar Documents

Publication Publication Date Title
JP7019727B2 (en) How to treat the flu
Ge et al. Anti-influenza agents from traditional Chinese medicine
US20190307722A1 (en) Antiviral compositions for the treatment of infections linked to coronaviruses
JP2008523153A (en) Compositions and methods for reducing disease transmission
CN107281210A (en) Application of the azithromycin in anti infection of coronavirus
WO2021179950A1 (en) Use of pharmaceutical composition in preparing anti-viral drug
RU2505306C2 (en) Composition for preventing and treating viral infections
US20230321126A1 (en) Micronutrient combination to inhibit coronavirus cell infection
Shin et al. Comparison of anti-influenza virus activity and pharmacokinetics of oseltamivir free base and oseltamivir phosphate
TWI453026B (en) Use of anisomeles indica (l.) kuntze extract and purified products thereof against influenza virus
WO2016110173A1 (en) New inhibitor for influenza virus neuraminidase and uses thereof
Chakraborty et al. Fighting the flu: a brief review on anti-influenza agents
Wang et al. Effect of Echinacea purpurea (L.) Moench and its extracts on the immunization outcome of avian influenza vaccine in broilers
CN105246484B (en) Usage of mycophenolate mofetil or salt thereof in preparing drug for resisting against influenza virus
CN107663158B (en) Preparation method of oseltamivir carboxylate and atomizing agent thereof
CA2465062C (en) Preventive and/or therapeutic agent for viral infection
EP4260862A1 (en) Antiviral composition containing fucosyllactose as active ingredient
US20210346324A1 (en) Niclosamide Formulations and Methods of Use
RU2740657C1 (en) Antiviral composition
WO2022088037A1 (en) Application of sirtinol in preparation of drug for preventing and treating coronavirus
US6475531B1 (en) Safe botanical drug for treatment and prevention of influenza and increasing immune function
CN104248654B (en) The application of karanjin or Indian beech extract in anti-influenza virus medicament
CN108014102A (en) The micromolecular inhibitor of Ebola's pseudovirus
CN106265684A (en) The application of Lupenyl acetate
CN106943416B (en) Pharmaceutical composition and application thereof in preparing medicine for treating pneumonia or/and enhancing natural killer cell activity in upper respiratory tract infection

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15876673

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205 DATED 01-12-17)

122 Ep: pct application non-entry in european phase

Ref document number: 15876673

Country of ref document: EP

Kind code of ref document: A1