WO2023116397A1 - 固有免疫激活药物及其用途 - Google Patents

固有免疫激活药物及其用途 Download PDF

Info

Publication number
WO2023116397A1
WO2023116397A1 PCT/CN2022/136399 CN2022136399W WO2023116397A1 WO 2023116397 A1 WO2023116397 A1 WO 2023116397A1 CN 2022136399 W CN2022136399 W CN 2022136399W WO 2023116397 A1 WO2023116397 A1 WO 2023116397A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
pharmaceutically acceptable
drugs
immune
Prior art date
Application number
PCT/CN2022/136399
Other languages
English (en)
French (fr)
Inventor
侯法建
Original Assignee
珞达生物医药(苏州)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 珞达生物医药(苏州)有限公司 filed Critical 珞达生物医药(苏州)有限公司
Priority to CN202280004982.4A priority Critical patent/CN116157126A/zh
Publication of WO2023116397A1 publication Critical patent/WO2023116397A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D219/00Heterocyclic compounds containing acridine or hydrogenated acridine ring systems
    • C07D219/04Heterocyclic compounds containing acridine or hydrogenated acridine ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • C07D219/08Nitrogen atoms
    • C07D219/10Nitrogen atoms attached in position 9
    • C07D219/12Amino-alkylamino radicals attached in position 9

Definitions

  • the invention belongs to the field of medicine, in particular, the invention relates to an inherent immune activation drug with broad-spectrum antiviral activity, used for immunotherapy, anti-tumor and immune adjuvant and its application.
  • Viral infection can cause a variety of diseases, and innate immunity is the first natural defense line of the host's immune system against pathogenic microorganisms.
  • the antiviral innate immune response induces host cells to produce interferon to inhibit the reproduction and amplification of the virus, and the virus will escape the innate immune response during the evolution process. Therefore, producing interferon by inducing innate immune response provides a new idea for antiviral therapy.
  • recombinantly expressed interferon is widely used in the treatment of viral infection, inflammation, immune dysfunction, etc., but it is a protein polypeptide, lacks post-translational modification, has an unstable structure, cannot be taken orally, and requires high storage conditions. It can only be administered by injection, and the method of administration is single. In addition, its half-life is short and its high concentration can cause severe toxic and side effects.
  • the outstanding disadvantage of recombinantly expressed interferon in Escherichia coli is the lack of glycosylation modification and limited activity; however, the cost of expressing and purifying interferon containing glycosylation using eukaryotic system is high, and the price of finished medicine is expensive.
  • the body's immune system can exert its immune surveillance function against tumors.
  • the immune system can identify and specifically eliminate malignant cells through immune mechanisms to resist the occurrence and development of tumors; Immunosurveillance, rapid proliferation in the body, forming tumors. The occurrence and outcome of tumors depend on the results of these two aspects.
  • antibodies As a drug for treating tumors, antibodies have excellent therapeutic effects due to their strong specificity.
  • the amount of antibodies produced is small, which limits its application, and requires the use of immune adjuvants.
  • Immunological adjuvants refer to auxiliary substances that are injected into the body before or at the same time as the antigen, which can enhance the body's immune response to the antigen or change the type of immune response.
  • the immunobiological effects of adjuvants include prolonging the retention time of antigens in the body, enhancing the processing and presentation of antigens, and enhancing and amplifying the immune response. Because immune adjuvants can enhance the immune response, they are widely used.
  • the immune adjuvant should have the following characteristics: for a specific animal, it should have less side effects; the effect of the adjuvant should be long-lasting and stable; the production cost should be as low as possible; the immune response should be appropriate, and it is best to induce the body's cellular or humoral immunity The strength must be able to meet the protection requirements. Therefore, the development of new immune adjuvants with high efficiency and low toxicity, which can improve the body's immune level, cytokine secretion and immune memory is one of the current research hotspots.
  • the present invention solves the technical problem of lack of new drugs that can effectively improve the innate immunity of the human body, and provides a drug for innate immunity activation. adjuvant.
  • R1, R2, R3, R4, R5, R6, R7, R8, R9, R10 are each independently selected from hydrogen, halogen, alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, alkenyl , alkynyl, amino, hydroxyl, mercapto, carboxyl, alkoxy, cycloalkoxy, haloalkyl, cyano, sulfanyl, sulfo, sulfone, sulfoxide, phosphoric acid, the alkyl, ring Alkyl, alkoxy, cycloalkoxy, amino, heterocyclyl, aryl or heteroaryl may be substituted by one or more substituents selected from hydroxy, halogen, alkyl, alkoxy radical, cycloalkyl, cycloalkoxy, heterocyclyl, aryl, haloaryl, haloalkyl, heteroaryl, alkenyl, alkynyl,
  • substitution means that one or more (preferably 1, 2, 3, 4, 5, 6, 7 or 8) hydrogen atoms on the ring or group are replaced by a substituent selected from the following group Substitution: hydroxy, halogen, alkyl, alkoxy, cycloalkyl, cycloalkoxy, heterocyclyl, aryl, haloaryl, haloalkyl, heteroaryl, alkenyl, alkynyl, amino, mercapto , carboxyl, ester, alkoxycarbonyl, acyloxy, amide, ureido, alkylsulfonyl, aromatic sulfonyl, cyano, nitro, nitroso, thiocyano, isothiocyano, sulfur Alkyl, sulfo, phosphoric acid, phosphonic acid, alkyl phosphoric acid, alkyl phosphonic acid, aryl phosphoric acid, aryl phosphonic acid.
  • the heterocyclic ring of the heteroaryl group has 1-4 (preferably 1, 2, 3 or 4) heteroatoms selected from N, O and S.
  • R1, R2, R5, R7, R10 are each independently hydrogen, substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C3-C12 cycloalkyl.
  • R1, R2, R5, R7, R10 are each independently hydrogen, substituted or unsubstituted C1-C8 alkyl, substituted or unsubstituted C3-C8 cycloalkyl.
  • R1, R2, R5, R7, R10 are each independently hydrogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C3-C8 cycloalkyl.
  • R1, R2, R5, R7, R10 are each independently hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl.
  • R1, R2, R5, R7 and R10 are each independently hydrogen, methyl, ethyl, propyl or butyl.
  • R3 and R4 are each independently hydrogen, substituted or unsubstituted C1-C8 alkoxy, substituted or unsubstituted C1-C8 alkylthio.
  • R3 and R4 are each independently hydrogen, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C1-C6 alkylthio.
  • R3 and R4 are each independently hydrogen, substituted or unsubstituted C1-C4 alkoxy, substituted or unsubstituted C1-C4 alkylthio.
  • R3 and R4 are each independently hydrogen, substituted or unsubstituted C1-C2 alkoxy, substituted or unsubstituted C1-C2 alkylthio.
  • R6 is substituted or unsubstituted amino, substituted or unsubstituted heterocyclyl, heterocyclylalkyl or alkylheterocyclyl.
  • R8 is haloalkyl
  • R9 is hydrogen, halogen, amino.
  • the compound of formula I includes:
  • the pharmaceutically acceptable salts of the compound of formula I include:
  • the innate immune activating drugs include:
  • the cytokine is selected from the group consisting of interferon, tumor necrosis factor, interleukin, or a combination thereof.
  • the interferon is selected from the group consisting of type I interferon, type II interferon, type III interferon, or a combination thereof.
  • the interferon is selected from the group consisting of IFN ⁇ (interferon- ⁇ ), IFN ⁇ (interferon- ⁇ ), TNF ⁇ (tumor necrosis factor- ⁇ ), or a combination thereof.
  • the interleukin is selected from the group consisting of IL6 (interleukin-6), IL1 ⁇ (interleukin-1 ⁇ ), or a combination thereof.
  • the cytokines include cytokines in blood, serum or plasma.
  • the cytokine inducer increases the level or content of the cytokine.
  • the blood includes peripheral blood.
  • the inducer includes;
  • the inducer includes inducing the increase of cytokines in peripheral blood.
  • the adjuvant includes an antibody adjuvant.
  • the immune adjuvant is used to enhance the immune titer of the antibody.
  • the immune adjuvant is used to promote antibody production or as an antibody production accelerator.
  • the antibodies include serum antibodies.
  • the antigens targeted by the antibodies include protein antigens.
  • the protein antigen includes albumin antigen.
  • the albumin antigen includes ovalbumin antigen.
  • the immune adjuvant is used to prepare antibodies, vaccines, immunotherapeutic drugs and/or immune activators.
  • the antibody is selected from the group consisting of IgG, IgM, or a combination thereof.
  • the IgG includes IgG1.
  • the vaccine includes a tumor vaccine.
  • the type I interferon-related disease refers to a disease that is improved, treated or prevented by increasing the level of type I interferon in the body.
  • the type I interferon (IFN) related diseases include: viral infection, multiple sclerosis and/or chronic myeloid leukemia.
  • the viruses include DNA viruses and RNA viruses.
  • the DNA virus includes: HSV (herpes simplex virus), HBV (hepatitis B virus), HAV (hepatitis A virus), HPV (human papillomavirus) and/or EBV (Epstein-Barr virus).
  • HSV herpes simplex virus
  • HBV hepatitis B virus
  • HAV hepatitis A virus
  • HPV human papillomavirus
  • EBV Epstein-Barr virus
  • the RNA virus includes: VSV (vesicular stomatitis virus), HCV (hepatitis C virus), EMCV (encephalomyocarditis virus), Ebola (Ebola virus), HIV (AIDS virus) and and/or Zika virus.
  • VSV vesicular stomatitis virus
  • HCV hepatitis C virus
  • EMCV encephalomyocarditis virus
  • Ebola Ebola virus
  • HIV AIDS virus
  • Zika virus Zika virus
  • a method for inducing a subject to produce type I interferon comprising the step of: contacting the subject with a compound of formula I, thereby inducing the subject to produce type I interferon.
  • the object is a cell.
  • the cells include: Vero cells, THP-1 cells, HEK293 cells, HEK293T cells, and/or A549 cells.
  • the method is non-therapeutic in vitro.
  • the method includes the step of: cultivating the subject in a system containing the compound of formula I, thereby inducing the subject to produce interferon.
  • the concentration of the compound of formula I is ⁇ 1 ⁇ M; preferably, ⁇ 3 ⁇ M.
  • the incubation time is 10 minutes to 48 hours.
  • the type I interferon includes IFN ⁇ .
  • a method for suppressing viral infection which includes the steps of:
  • a subject is contacted with a compound of formula I, thereby inhibiting viral infection of the subject.
  • the object is a cell.
  • the cells include: Vero cells, THP-1 cells, HEK293 cells, HEK293T cells, and/or A549 cells.
  • the viruses include DNA viruses and RNA viruses.
  • the DNA virus includes: HSV (herpes simplex virus), HBV (hepatitis B virus), HAV (hepatitis A virus), HPV (human papillomavirus) and/or EBV (Epstein-Barr virus).
  • HSV herpes simplex virus
  • HBV hepatitis B virus
  • HAV hepatitis A virus
  • HPV human papillomavirus
  • EBV Epstein-Barr virus
  • the RNA virus includes: VSV (vesicular stomatitis virus), HCV (hepatitis C virus), EMCV (encephalomyocarditis virus), Ebola (Ebola virus), HIV (AIDS virus) and / or Zika virus (Zika virus), etc.
  • VSV vesicular stomatitis virus
  • HCV hepatitis C virus
  • EMCV encephalomyocarditis virus
  • Ebola Ebola virus
  • HIV AIDS virus
  • Zika virus Zika virus
  • the method is non-therapeutic in vitro.
  • the method includes the step of: cultivating the subject in a system containing the compound of formula I, thereby inducing the subject to produce interferon.
  • a compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered to a subject in need thereof.
  • the subject is a mammal; preferably, it is selected from the group consisting of human, mouse, and rat.
  • a broad-spectrum antiviral pharmaceutical composition which comprises: a compound of formula I or its tautomer, mesoform, racemate, Enantiomers, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • an immune composition which comprises: a compound of formula I or its tautomer, mesoform, racemate, enantiomer isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof; and antigens.
  • the antigen includes a protein antigen.
  • the protein antigen includes albumin antigen.
  • the albumin antigen includes ovalbumin antigen.
  • an anti-tumor pharmaceutical composition comprising: a compound of formula I or its tautomer, mesoform, racemate, enantiomer Isomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the first active ingredient is a compound shown in formula I, or its tautomer, meso, racemate, enantiomer, diastereoisomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a prodrug thereof;
  • a therapeutically effective amount of a second active ingredient comprising: an immunotherapy activator, a type I interferon modulator, or an antiviral agent.
  • a kit which includes:
  • the first preparation containing the first active ingredient is a compound shown in formula I, or its tautomer, meso, racemate, enantiomer, diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof;
  • a second formulation comprising a second active ingredient comprising: an immunotherapy activator, a type I interferon modulator, or an antiviral agent.
  • the immunotherapy activator includes PD-1 or PD-L1 antibody, and the combination of the compound represented by formula I of the present invention and PD-1 or PD-L1 antibody can exert a synergistic effect and significantly increase Inhibitory effect on tumor growth, significantly increased survival.
  • the use of the above-mentioned composition or the above-mentioned kit is also provided for the preparation of an innate immune activation drug, and the drug includes:
  • the present invention finds that the compound of formula I exhibits a strong ability to induce the expression of interferon (such as IFN ⁇ ).
  • the present invention finds that the compound of formula I can efficiently and safely induce the production of cytokines, thereby being used for treating diseases related to cytokines, such as tumors and other diseases.
  • the present invention finds that the compound of formula I can be used as an immune adjuvant, significantly improve the immune titer of antibodies, and promote the production of antibodies, thereby being used to prepare antibodies, vaccines, immunotherapeutic drugs and immune activators, and improve the application value of antibodies .
  • the compound of the present invention has high drug efficacy, good druggability and low side effects.
  • Figure 1 is a graph showing the results of transcriptional activation of type I interferon and interleukin-6 in HEK293T cells treated with compounds LD001-LD004 at a final concentration of 10 ⁇ M in Example 1 of the present invention for 12 hours;
  • Fig. 2 is a graph showing the results of transcriptional activation of type I interferon and interleukin-6 in HeLa cells after treatment with compounds LD001-LD004 at a final concentration of 10 ⁇ M in Example 1 of the present invention for 12 hours;
  • Fig. 3 is a graph showing the results of transcriptional activation of type I interferon and interleukin-6 in A549 cells after treatment with compounds LD001-LD004 at a final concentration of 10 ⁇ M in Example 1 of the present invention for 12 hours;
  • Fig. 4 is a graph showing the results of transcriptional activation of type I interferon and interleukin-6 in HEK293T cells treated with compounds LD005-LD011 at a final concentration of 10 ⁇ M in Example 1 of the present invention for 12 hours;
  • Fig. 5 is a graph showing the results of transcriptional activation of type I interferon and interleukin-6 in PEM cells after treatment with compounds LD005-LD011 at a final concentration of 10 ⁇ M in Example 1 of the present invention for 12 hours;
  • FIG. 6 is a graph showing the inhibition of VSV-GFP by LD002 of Example 2 of the present invention.
  • Fig. 7 is the inhibition curve graph of LD002 of the embodiment 2 of the present invention to H1N1-IAV;
  • Fig. 8 is the inhibition curve diagram of LD002 of the embodiment of the present invention 3 to HSV;
  • Figure 9 is a graph showing the results of an ELISA test in which the compound LD002 of Example 4 of the present invention induces the production of interferon in mice;
  • Figure 10 is a graph showing the experimental results of the compound LD002 of Example 5 of the present invention inhibiting the growth of melanoma when administered alone;
  • Figure 11 is a graph showing the experimental results of compound LD002 in Example 5 of the present invention combined with another immunotherapy activator anti-PD-L1 to inhibit the growth of melanoma;
  • Fig. 12 is a diagram showing the result of detecting that the compound of the present invention has immune adjuvant activity by ELISA method in Example 6 of the present invention.
  • a class of three-membered heterocyclic compounds can activate host innate immunity, induce different cells to express or produce cytokines such as type I interferon IFN ⁇ .
  • this class of compounds has broad-spectrum inhibitory effects (dose-related) on infection by RNA viruses (such as VSV) and DNA viruses (such as HSV). Therefore, a new drug capable of inducing cells to produce IFN ⁇ and having a potential broad-spectrum antiviral effect in clinical treatment is provided.
  • the compound of formula I described in the present invention has an excellent inducing effect on cytokines and is used for the treatment of diseases such as tumors.
  • the compound of formula I described in the present invention can also be used as an immune adjuvant to significantly improve the immune titer of antibodies and promote the production of antibodies, thereby being used to prepare antibodies, vaccines, immunotherapeutic drugs and immune activators, and to improve the immune titer of antibodies. Value.
  • the host's innate immunity is the first line of defense of the immune system against pathogenic microorganisms.
  • Viruses are pathogens that can cause various diseases in nature.
  • the innate immune response of host cells against viruses can induce the production of interferon to inhibit the reproduction and amplification of viruses.
  • the antiviral innate immune response is mediated by signal transduction pathways.
  • Previous studies have revealed how the innate immune system recognizes virus invasion and triggers immune defense functions quickly, thereby effectively clearing viral infections. This is an important research in the field of life science and medicine, which provides a theoretical basis for the design of related drugs and brings new hope for the prevention and treatment of viral infections.
  • host cells After host cells detect pathogenic microorganisms, they induce the production of interferons, pro-inflammatory factors, and chemokines through innate immune signaling pathways. On the one hand, these effectors can inhibit the replication of pathogens, thereby rapidly controlling the proliferation of pathogens; Cytokines can mobilize other immune cells of the host, including antigen-presenting cells and T and B lymphocytes in the adaptive immune system, to activate adaptive immunity. In addition, the induction and activation of the innate immune response also has anti-tumor effects and can enhance the effects of radiotherapy and chemotherapy.
  • the innate immune signaling pathway starts with the recognition of the relevant molecular patterns of pathogenic microorganisms by the pattern recognition receptors of host cells.
  • the innate immune signaling pathways related to viral infection that have been discovered so far mainly include the RLR-MAVS signaling pathway that recognizes RNA viruses in the cytoplasm, the cGAS-STING signaling pathway that recognizes DNA viruses, and the TLRs signaling pathway.
  • RLRs and cGAS both recruit and activate TBK1 through the downstream transduction protein, TBK1 phosphorylates the transcription factor IRF3 and enters the nucleus to induce the production of type I interferon, and finally achieves the effect of inhibiting the virus.
  • some viruses have also acquired some mechanisms to escape the innate immune response.
  • innate immunity plays an important role in all aspects of the body's life activities, innate immunity has important guiding significance for the development of vaccines and drugs.
  • the term "compound of the present invention” refers to a compound represented by formula I.
  • the term also includes various crystalline forms, pharmaceutically acceptable salts, isomers, hydrates, solvates or prodrugs of the compounds of formula I.
  • the term "pharmaceutically acceptable salt” or “pharmaceutically acceptable salt” refers to a salt formed by the compound of the present invention with an acid or a base which is suitable for use as a medicine.
  • Pharmaceutically acceptable salts include inorganic salts and organic salts.
  • a preferred class of salts are the salts of the compounds of the invention with acids.
  • Acids suitable for forming salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and other organic acids; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid
  • salts of the compounds of the present invention with bases, such as alkali metal salts (such as sodium or potassium salts), alkaline earth metal salts (such as magnesium or calcium salts), ammonium salts (such as lower alkanolammonium salts and other pharmaceutically acceptable amine salts), such as methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butyl amine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, trihydroxyethylamine salts, and amine salts formed from morpholine, piperazine, and lysine, respectively.
  • bases such as alkali metal salts (such as sodium or potassium salts), alkaline earth metal salts (such as magnesium or calcium salts), ammonium salts (such as lower alkanolammonium salts
  • the compound of formula I described in the present invention can be converted into its pharmaceutically acceptable salt by conventional methods, for example, the solution of the corresponding acid can be added to the solution of the above compound, and the solvent is removed after the salt formation is complete to obtain the present invention.
  • the corresponding salts of said compounds can be converted into its pharmaceutically acceptable salt by conventional methods, for example, the solution of the corresponding acid can be added to the solution of the above compound, and the solvent is removed after the salt formation is complete to obtain the present invention.
  • the corresponding salts of said compounds can be converted into its pharmaceutically acceptable salt by conventional methods, for example, the solution of the corresponding acid can be added to the solution of the above compound, and the solvent is removed after the salt formation is complete to obtain the present invention.
  • the corresponding salts of said compounds can be converted into its pharmaceutically acceptable salt by conventional methods, for example, the solution of the corresponding acid can be added to the solution of the above compound, and the solvent is removed after the salt formation is complete to obtain the present invention.
  • the corresponding salts of said compounds
  • prodrug includes prodrugs which themselves may be biologically active or inactive, which undergo metabolism or chemical reactions in the human body to convert to a compound of formula I when administered in an appropriate manner.
  • the prodrugs include (but are not limited to) carboxylates, carbonates, phosphates, nitrates, sulfates, sulfone esters, sulfoxide esters, amino compounds, carbamates, azo compounds of the compounds , phosphoramide, glucoside, ether, acetal and other forms.
  • the first active ingredient of the present invention is the compound of formula I, or its pharmaceutically acceptable salt, or its optical isomer, or its racemate, or its solvate, or its prodrug.
  • compounds of the present invention are used interchangeably to refer to compounds of formula I, or their tautomers, mesomers, racemates isomers, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof. It should be understood that the term also includes mixtures of the aforementioned components.
  • R1, R2, R3, R4, R5, R6, R7, R8, R9, R10 are each independently selected from hydrogen, halogen, alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, Alkenyl, alkynyl, amino, hydroxyl, mercapto, carboxyl, alkoxy, cycloalkoxy, haloalkyl, cyano, sulfanyl, sulfo, sulfone, sulfoxide, phosphoric acid, the alkyl , cycloalkyl, alkoxy, cycloalkoxy, amino, heterocyclyl, aryl or heteroaryl may be substituted by one or more substituents selected from hydroxyl, halogen, alkyl, Alkoxy, cycloalkyl, cycloalkoxy, heterocyclyl, aryl, haloaryl, haloalkyl, heteroaryl, alkenyl, alkyny
  • Preferred compounds of the invention include:
  • Preferred pharmaceutically acceptable salts of the compounds of the invention include:
  • the second active ingredient of the present invention is an anti-virus, anti-tumor, immune regulation and immune adjuvant drug, which can be used in combination with PD-1 or PD-L1 antibody for immunotherapy.
  • PD-L1 is called programmed cell death-ligand 1, which is an important immunosuppressive molecule. PD-L1 is expressed on the surface of tumors and interacts with the surface of T cells. PD-1 binds, inhibits T cell proliferation and cytokine secretion, negatively regulates lymphocyte activation, and participates in tumor immune escape.
  • compositions and methods of administration are provided.
  • the compound of the present invention has an excellent ability to induce the production of type I interferon
  • the compound of the present invention and various crystal forms thereof, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and compounds containing the present invention are
  • the pharmaceutical composition of the main active ingredients can be used to treat, prevent and alleviate diseases related to type I interferon (especially those diseases that can be improved, treated or prevented by regulating the level of interferon in the body, such as increasing the level of interferon).
  • the compound of the present invention can be used to treat the following diseases: viral infection, multiple sclerosis, systemic lupus erythematosus, chronic myeloid leukemia and the like.
  • the terms “inducing the production of interferon”, “inducing the production of type I interferon”, “inducing the production of type I interferon” or “inducing the production of interferon” are used interchangeably and are intended to include: The expression, transcription or protein content level of interferon or type I interferon-related proteins, cytokines, etc.
  • the pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof within a safe and effective amount range and a pharmaceutically acceptable excipient or carrier.
  • safe and effective dose refers to: the amount of the compound is sufficient to obviously improve the condition without causing severe side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention per dose, more preferably 10-500 mg of the compound of the present invention per dose.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and low enough toxicity. "Compatibility” herein means that the components of the composition can be blended with the compound of the present invention and with each other without significantly reducing the efficacy of the compound.
  • Examples of pharmaceutically acceptable carrier parts include cellulose and derivatives thereof (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween ), wetting agent (such as sodium lauryl sulfate), coloring agent, flavoring agent, stabilizer, antioxidant, preservative, pyrogen-free water, etc.
  • cellulose and derivatives thereof such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as talc
  • solid lubricants such as stearic acid , magnesium stearate
  • the mode of administration of the compound or pharmaceutical composition of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration .
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with (a) fillers or extenders, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow agents, such as paraffin; (f) Absorption accelerators such as quaternary ammonium compounds; (g) wetting agents such as cetyl alcohol and glyceryl monostea, or
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shell materials, such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • coatings and shell materials such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • Examples of usable embedding components are polymeric substances and waxy substances.
  • the active compounds can also be in microencapsulated form, if desired, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • liquid dosage forms may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, etc.
  • inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and
  • compositions can also contain adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
  • Dosage forms for topical administration of a compound of this invention include ointments, powders, patches, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required, if necessary.
  • the compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage is a pharmaceutically effective dosage when administered, for a person with a body weight of 60kg, the daily
  • the dosage is usually 1-2000 mg, preferably 20-500 mg.
  • factors such as the route of administration and the health status of the patient should also be considered for the specific dosage, which are within the skill of skilled physicians.
  • compositions or formulations, combinations and kits of active ingredients and methods of administration are provided.
  • the present invention also provides a composition or preparation, a combination of active ingredients and a kit, and the composition or preparation, a combination of active ingredients and a kit can be used to prepare an innate immune activating drug, and the drug includes: (a) Broad-spectrum antiviral drugs; (b) antineoplastic drugs; (c) drugs for the prevention and/or treatment of cancer; (d) drugs for the treatment or prevention of type I interferon-related diseases; (e) immune adjuvants; and/or (f) Cytokine inducers.
  • the drug includes: (a) Broad-spectrum antiviral drugs; (b) antineoplastic drugs; (c) drugs for the prevention and/or treatment of cancer; (d) drugs for the treatment or prevention of type I interferon-related diseases; (e) immune adjuvants; and/or (f) Cytokine inducers.
  • composition of the present invention is preferably a pharmaceutical composition.
  • compositions of the present invention may include a pharmaceutically acceptable carrier.
  • the pharmaceutical preparation should match the mode of administration, and the preferred mode of administration is oral administration, injection administration (such as intratumoral injection), and when used, the drug of a therapeutically effective amount is applied to the required object (such as human or non-tumor injection). human mammal).
  • the term "therapeutically effective amount” refers to an amount that produces functions or activities on humans and/or animals and is acceptable to humans and/or animals. Those of ordinary skill in the art should understand that the "therapeutically effective amount” may vary depending on the form of the pharmaceutical composition, the route of administration, the adjuvant of the drug used, the severity of the disease, and the combination with other drugs. different.
  • a safe and effective daily dose of the first active ingredient will generally be at least about 0.1 mg, and in most cases will not exceed about 2500 mg.
  • the dose is 1 mg to 500 mg; a safe and effective amount of the second active ingredient is usually at least about 0.01 mg, and in most cases not more than 2500 mg.
  • the dosage range is 0.1 mg to 2500 mg.
  • factors such as the route of administration and the health status of the patient should also be considered for the specific dosage, all of which are within the skill of skilled physicians.
  • Example 1 Compounds of the present invention induce cells to secrete interferon
  • the 293T cell line, HeLa cell line and A549 cell line used were all purchased from the Cell Bank of Shanghai Institute of Biological Sciences, Chinese Academy of Sciences.
  • the monolayer was cultured in an incubator at 37°C and 5% CO 2 , and the cells were grown in DMEM medium containing 10% FBS, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • PEM cells are macrophages isolated from the peritoneal cavity of C57BL/6 mice.
  • HEK293T, HeLa, A549 and PEM in the logarithmic growth phase on 12-well cell culture plates at a seeding density of 80-90%. After the cells adhere to the wall for 6 hours, replace the cell culture supernatant. A culture medium containing various small molecule compounds of LD001-LD011 above at a final concentration of 10 ⁇ M was prepared and cultured for 12 hours, and finally the cells were collected and total RNA was extracted from the cells.
  • Embodiment 2 Compound LD002 of the present invention inhibits RNA viruses (VSV and H1N1-IAV)
  • Example 1 The results of Example 1 above have proved that LD002 can significantly induce the expression of type I interferon in cells, and then use the virus cell infection model to detect the inhibitory effect of LD002 on virus amplification.
  • VSV Vesicular stomatitis virus
  • IAV influenza virus
  • the A549 cells in the logarithmic growth phase were spread on a 12-well cell culture plate at a seeding density of 80-90%. After the cells adhered to the wall for 6 hours, a dilution of VSV virus with a multiplicity of infection (MOI) of 0.5 was added. One hour after virus infection, they were treated with different concentrations of the compound LD002 of the present invention, and 12 hours later, whole-cell RNA was extracted and virus amplification was detected.
  • MOI multiplicity of infection
  • the EC50 (half effective concentration) of LD002 on the inhibition of VSV-GFP infection was calculated to be 1.789, and the SI (selection index) was 135.1 (Fig. 6). It shows that the compound of the present invention has excellent anti-VSV virus activity and safety.
  • the A549 cells in the logarithmic growth phase were spread on a 12-well cell culture plate at a seeding density of 80 to 90%. After the cells were attached to the wall for 6 hours, the multiplicity of infection (MOI) was added to the H1N1-IAV virus of 0.5. Diluent. One hour after virus infection, they were treated with different concentrations of the compound LD002 of the present invention, and 12 hours later, whole-cell RNA was extracted and virus amplification was detected.
  • MOI multiplicity of infection
  • the EC50 (half effective concentration) of LD002 for the inhibition of H1N1-IAV infection was calculated to be 4.735, and the SI (selection index) was 52.79 (Figure 7). The results show that the compound of the present invention has better anti-IAV virus activity and safety.
  • Example 3 The inhibitory effect of compound LD002 of the present invention on DNA virus (such as HSV) infection
  • the herpes simplex virus HSV is selected as the representative of the DNA virus; specifically, the HSV-1-GFP virus strain is used, which can express the green fluorescent protein GFP itself, which can indicate the proliferation and load of the virus.
  • A549 cells in the logarithmic growth phase were spread on 12-well cell culture plates at a seeding density of 80-90%. After the cells had adhered to the wall for 6 hours, a dilution of HSV virus with a multiplicity of infection (MOI) of 0.5 was added. One hour after virus infection, they were treated with different concentrations of the compound LD002 of the present invention, and 12 hours later, whole-cell RNA was extracted and virus amplification was detected.
  • MOI multiplicity of infection
  • the EC50 (half effective concentration) of LD002 for the inhibition of HSV-GFP infection was calculated to be 1.948, and the SI (selection index) was 123.92 (Fig. 8).
  • the results show that the compound of the present invention has excellent anti-HSV virus activity and safety.
  • Example 4 The compound LD002 of the present invention induces the production of interferon in mice
  • mice used in all experiments were 6-8 week old male C57BL/6 mice provided by the animal experiment technology platform of the Molecular Cell Excellence Center.
  • ELISA was used to detect the content changes of various cytokines in serum.
  • the results show that the compound LD002 experimental group of the present invention can significantly increase the levels of type I interferon IFN ⁇ (interferon- ⁇ ), IL1 ⁇ (interleukin-1 ⁇ ) in serum, and can slightly increase the level of IL-6 (interleukin-6).
  • TNF ⁇ tumor necrosis factor- ⁇
  • type III interferon IFN ⁇ interferon- ⁇ expression levels
  • Example 5 The compound LD002 of the present invention has anti-tumor effect and the function of promoting tumor immunotherapy
  • the B16-F10 melanoma cell line used in this experiment was purchased from the Cell Bank of the Chinese Academy of Sciences in Shanghai, routinely cultured in DMEM medium containing 10% FBS, and cultured in a 5% CO 2 incubator at 37°C.
  • Cells in the exponential proliferation phase were digested with 0.25% trypsin, centrifuged and resuspended in PBS for counting. The rate of viable cells was over 95%. After counting, the concentration of the cell suspension was adjusted to 2*10 ⁇ 6 mL -1 .
  • the resuspended tumor cells were divided into sterile EP tubes and placed on ice for later use.
  • mice The hair on the mouse's back was removed with an electric shaver, and the dehaired area was about 9 cm 2 .
  • the B16-F10 cells prepared above were taken, and 100 ⁇ L of tumor cell suspension was subcutaneously injected into the back of each mouse. The tumor growth of the mice was observed every day. On the 4th day, the formation of melanoma was visible to the naked eye. The mice without tumors were excluded, and the remaining mice were randomly divided into 7 groups, and the number of mice in each experimental group was 5.
  • the first experiment group was the control group, the 100 ⁇ g compound LD002 group of the present invention, and the 300 ⁇ g compound LD002 group of the present invention; : 10F.9G2) group, 100 ⁇ g compound LD002 of the present invention + 200 ⁇ g anti-PD-L1 antibody (B7-H1) (Clone: 10F.9G2) group, 6-8 rats in each group.
  • the experimental method is as follows: on the fifth day after the tumor cell injection, the tumor-bearing mice were administered according to the groups, and LD002 was injected intraperitoneally, and two dosage groups were set up: 100 ⁇ g/mouse and 300 ⁇ g/mouse.
  • Anti-PD-L1 antibody was administered by intraperitoneal injection, and one dose group was set up: 200 ⁇ g/mouse.
  • the solvent buffer system for administration is PBS buffer solution.
  • the control group was intragastrically administered the same volume of PBS buffer solution.
  • each mouse was administered once every 2 days, and administered 3 times in total.
  • the administration volume was 100 ⁇ L.
  • For the anti-PD-L1 antibody each mouse was administered once every 2 days, for a total of 3 administrations.
  • the dosing volume was 50 ⁇ L. Specifically, the administration was carried out on the 5th, 7th, and 9th day respectively, with an interval of 2 days, and a total of 3 administrations.
  • the tumor size was measured every other day, the survival period of the mice was observed and relevant data were recorded, and the tumor volume was measured using an electronic caliper.
  • the volume calculation formula was: ⁇ /6xlengthxwidthxheight. Tumor-bearing mice were sacrificed when the tumor volume was greater than 2000 mm 3 .
  • the compound LD002 of the present invention inhibits the growth of subcutaneous melanoma in mice, and improves the survival period of tumor-bearing mice.
  • the compounds LD002 and LD002+anti-PD-L1 of the present invention were detected to inhibit tumor growth and improve the survival of tumor-bearing mice.
  • the results showed that, compared with the control group, single administration
  • the compound of the present invention has a significant dose-dependent inhibitory effect on the growth of melanoma, and increases the survival period of tumor-bearing mice ( FIG. 10 ).
  • anti-PD-L1 can significantly enhance the inhibitory effect of the compound of the present invention on the growth of melanoma and improve the survival period of mice. It can be seen from the result data that the compound LD002 of the present invention and anti-PD-L1 can jointly play a synergistic effect, and the inhibitory effect on the growth of melanoma and the improvement of the survival period of mice are more significant (Figure 11).
  • the compound LD002 of the present invention can induce the increase of various cytokines such as interferon in peripheral blood, has an excellent inhibitory effect on tumors such as melanoma and improves survival.
  • the combination of the compound LD002 of the present invention and anti-PD-L1 has a synergistic effect in inhibiting tumor growth and improving survival. Therefore, the compound of the present invention can be used as a drug for inducing various cytokines such as interferon, and has anti-tumor effect and function of promoting tumor immunotherapy.
  • Example 6 Compound LD002 of the present invention has immune adjuvant activity
  • the experiments were divided into blank group (injection of the same volume of PBS), antigen group and antigen/compound of the present invention group (including different concentration gradients), with 5 mice in each group.
  • the antigen ovalbumin OVA and the compound LD002 of the present invention were respectively dissolved in PBS to obtain the antigen solution and the compound solution of the present invention, and were formulated into solutions containing the antigen and the compound of the present invention with different concentration gradients according to the concentration requirements.
  • Each mouse in the experimental group was immunized with 100 ⁇ l of antigen solution or antigen solution and LD002 solution each time, and the immunization method was intramuscular injection.
  • the mice in the blank group were injected with the same volume of PBS solution as that in the experimental group.
  • the first immunization was set as day 0, and the booster immunization was performed on the 7th and 14th days respectively.
  • blood was collected from the orbit of the mice, and the serum of the mice was collected. After dilution, the serum of the mice was detected by ELISA antibody content.
  • protective humoral immune response could not be produced after OVA immunization alone, when OVA combined with the compound of the present invention immunized mice could significantly increase the total amount of OVA-specific IgG, IgG1 and IgG2b antibodies, indicating that the compound of the present invention It has obvious immune adjuvant activity (Figure 12).

Landscapes

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

Abstract

公开了一种固有免疫激活药物,该药物具有广谱抗病毒活性、免疫佐剂活性、促进免疫疗法和抗肿瘤活性。具体地,公开了式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药在制备固有免疫激活药物中的用途。所述化合物能够激活固有免疫,诱导产生I型干扰素,显著抑制病毒感染以及促进免疫功能和抗肿瘤。

Description

固有免疫激活药物及其用途 技术领域
本发明属于医药领域,具体地,本发明涉及一种具有广谱抗病毒活性、用于免疫疗法、抗肿瘤和免疫佐剂的固有免疫激活药物及其用途。
背景技术
病毒感染能够引起多种疾病,而固有免疫是宿主的免疫系统抵抗病原微生物的第一道天然防线。抗病毒的固有免疫反应诱导宿主细胞产生干扰素来抑制病毒的繁殖和扩增,病毒在进化过程中会逃逸固有免疫反应。因此,通过诱导固有免疫反应产生干扰素为抗病毒治疗提供了一种新思路。
目前利用重组表达的干扰素广泛应用于病毒性感染、炎症、免疫功能障碍等的治疗,但其为蛋白多肽,缺乏翻译后修饰,结构不稳定,不能口服且保存条件要求高。只能依靠注射给药,给药方式单一。另外其半衰期短且高浓度会引起严重的毒副作用。大肠杆菌重组表达的干扰素突出缺点是缺少糖基化修饰,活力受限;而利用真核系统表达纯化的包含糖基化的干扰素成本较高,成药价格贵。
综上所述,本领域迫切需要开发具有诱导产生干扰素能力的新药物。
另外,机体免疫系统能够发挥免疫监视功能对抗肿瘤,一方面,免疫系统能够识别并通过免疫机制特异地清除恶变细胞,抵御肿瘤的发生发展;另一方面,恶变细胞可能通过多种机制逃避机体的免疫监视,在体内迅速增殖,形成肿瘤。肿瘤的发生及转归与否取决于这两方面的作用结果。抗体作为肿瘤治疗药物,由于其特异性强,具有优异的治疗作用。另外,动物产生抗体的过程中,由于机体对抗原的免疫应答慢和差等因素,抗体的产生量少,从而限制了其的应用,需要借助免疫佐剂的作用。免疫佐剂简称佐剂,指先于抗原或与抗原同时注入机体内,可增强机体对抗原的免疫应答或改变免疫应答类型的辅助物质。佐剂的免疫生物学作用包括延长抗原在体内的潴留时间、增强对抗原的加工与提呈、增强和放大免疫反应。由于免疫佐剂具有增强免疫应答的作用,故其应用广泛。免疫佐剂应具有以下特点:对于特定的动物,要具有较小的副作用;佐剂作用要持久稳定;生产成本要尽量低;产生的免疫应答反应要适当,最好能够诱导机体细胞或体液免疫的强度要能达到保护要求等。因此,研发高效低毒,能够提高机体免疫水平、细胞因子分泌和免疫记忆的新型免疫佐剂是目前研究的热点之一。
因此,本领域需要开发促进肿瘤免疫治疗和用作免疫佐剂的药物。
发明内容
本发明要解决目前缺乏能有效提高人体固有免疫的新药物的技术问题,提供一种 固有免疫激活药物,该药物不仅可以诱导产生干扰素对抗病毒感染,而且能促进肿瘤免疫治疗,及用作免疫佐剂。
在本发明的一个方面,提供一种式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药的用途,用于制备固有免疫激活药物,
Figure PCTCN2022136399-appb-000001
其中,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10各自独立地选自氢、卤素、烷基、环烷基、芳基、杂芳基、杂环基、烯基、炔基、氨基、羟基、巯基、羧基、烷氧基、环烷氧基、卤代烷基、氰基、硫烷基、磺基、砜基、亚砜基、磷酸基,所述烷基、环烷基、烷氧基、环烷氧基、氨基、杂环基、芳基或杂芳基可被一个或多个取代基所取代,所述取代基选自羟基、卤素、烷基、烷氧基、环烷基、环烷氧基、杂环基、芳基、卤代芳基、卤代烷基、杂芳基、烯基、炔基、氨基、巯基、羧基、酯基、烷氧基羰基、酰基氧基、酰胺基、脲基、烷基磺酰基、芳香磺酰基、氰基、硝基、亚硝基、硫氰基、异硫氰基、硫烷基、磺基、磷酸基、膦酸基、烷基磷酸基、烷基膦酸基、芳基磷酸基、芳基膦酸基中的一个或多个取代基,或为空;所述杂环基包含至少一个N原子,或者包含1个或2个或3个任选于N、S和O的杂原子。
其中,所述的“取代”是指环或基团上的一个或多个(优选为1、2、3、4、5、6、7或8个)氢原子被选自下组的取代基所取代:羟基、卤素、烷基、烷氧基、环烷基、环烷氧基、杂环基、芳基、卤代芳基、卤代烷基、杂芳基、烯基、炔基、氨基、巯基、羧基、酯基、烷氧基羰基、酰基氧基、酰胺基、脲基、烷基磺酰基、芳香磺酰基、氰基、硝基、亚硝基、硫氰基、异硫氰基、硫烷基、磺基、磷酸基、膦酸基、烷基磷酸基、烷基膦酸基、芳基磷酸基、芳基膦酸基。
所述杂芳基的杂环上具有1-4个(优选为1、2、3个或4个)选自N、O和S的杂原子。
在另一优选例中,R1、R2、R5、R7、R10各自独立地为氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C12环烷基。
在另一优选例中,R1、R2、R5、R7、R10各自独立地为氢、取代或未取代的C1-C8烷基、取代或未取代的C3-C8环烷基。
在另一优选例中,R1、R2、R5、R7、R10各自独立地为氢、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基。
在另一优选例中,R1、R2、R5、R7、R10各自独立地为氢、取代或未取代的C1- C4烷基、取代或未取代的C3-C8环烷基。
在另一优选例中,R1、R2、R5、R7、R10各自独立地为氢、甲基、乙基、丙基或丁基。
在另一优选例中,R3、R4各自独立地为氢、取代或未取代的C1-C8烷氧基、取代或未取代的C1-C8烷硫基。
在另一优选例中,R3、R4各自独立地为氢、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷硫基。
在另一优选例中,R3、R4各自独立地为氢、取代或未取代的C1-C4烷氧基、取代或未取代的C1-C4烷硫基。
在另一优选例中,R3、R4各自独立地为氢、取代或未取代的C1-C2烷氧基、取代或未取代的C1-C2烷硫基。
在另一优选例中,R6为取代或未取代的氨基、取代或未取代的杂环基、杂环基烷基或烷基杂环基。
在另一优选例中,R8为卤代烷基。
在另一优选例中,R9为氢、卤素、氨基。
在另一优选例中,所述式I化合物包括:
Figure PCTCN2022136399-appb-000002
Figure PCTCN2022136399-appb-000003
优选的,所述式I化合物的可药用盐包括:
Figure PCTCN2022136399-appb-000004
优选的,所述固有免疫激活药物包括:
(a)广谱抗病毒药物;
(b)抗肿瘤药物;
(c)预防和/或治疗癌症的药物;
(d)治疗或预防I型干扰素相关疾病的药物;
(e)免疫佐剂;
和/或(f)细胞因子诱导剂。
在另一优选例中,所述的细胞因子选自下组:干扰素、肿瘤坏死因子、白细胞介 素,或其组合。
在另一优选例中,所述的干扰素选自下组:I型干扰素、II型干扰素、III型干扰素,或其组合。
在另一优选例中,所述的干扰素选自下组:IFNβ(干扰素-β)、IFNλ(干扰素-λ)、TNFα(肿瘤坏死因子-α),或其组合。
在另一优选例中,所述的白细胞介素选自下组:IL6(白细胞介素-6)、IL1α(白细胞介素-1α),或其组合。
在另一优选例中,所述的细胞因子包括血液、血清或血浆中的细胞因子。
在另一优选例中,所述的细胞因子的诱导剂提高细胞因子的水平或含量。
在另一优选例中,所述血包括外周血。
在另一优选例中,所述的诱导剂包括;
(i)诱导细胞因子的表达;和/或
(i)诱导细胞因子的含量升高。
在另一优选例中,所述的诱导剂包括诱导外周血中细胞因子的升高。
在另一优选例中,所述的佐剂包括抗体佐剂。
在另一优选例中,所述免疫佐剂用于增强抗体的免疫滴度。
在另一优选例中,所述免疫佐剂用于促进抗体的产生或作为抗体生成促进剂。
在另一优选例中,所述的抗体包括血清抗体。
在另一优选例中,所述抗体针对的抗原包括蛋白抗原。
在另一优选例中,所述的蛋白抗原包括白蛋白抗原。
在另一优选例中,所述的白蛋白抗原包括卵白蛋白抗原。
在另一优选例中,所述的免疫佐剂用于制备抗体、疫苗、免疫治疗药物和/或免疫激活剂。
在另一优选例中,所述的抗体选自下组:IgG、IgM,或其组合。
在另一优选例中,所述的IgG包括IgG1。
在另一优选例中,所述的疫苗包括肿瘤疫苗。
在另一优选例中,所述I型干扰素相关疾病是指通过是体内I型干扰素水平增加而被改善、治疗或预防的疾病。
在另一优选例中,所述I型干扰素(IFN)相关疾病包括:病毒感染、和多发性硬化症和/或慢性粒细胞白血病。
在另一优选例中,所述的病毒包括DNA病毒和RNA病毒。
在另一优选例中,所述的DNA病毒包括:HSV(单纯疱疹病毒)、HBV(乙肝病毒)、HAV(甲肝病毒)、HPV(人乳头瘤病毒)和/或EBV(EB病毒)。
在另一优选例中,所述的RNA病毒包括:VSV(水疱性口炎病毒)、HCV(丙肝病毒)、EMCV(脑心肌炎病毒)、Ebola(埃博拉病毒)、HIV(艾滋病毒)和/或寨卡病毒(Zika virus)。
在本发明的另一方面,还提供了一种诱导对象产生I型干扰素的方法,所述的方法包括步骤:使对象与式I化合物接触,从而诱导所述对象产生I型干扰素。
在另一优选例中,所述的对象是细胞。
在另一优选例中,所述的细胞包括:Vero细胞、THP-1细胞、HEK293细胞、HEK293T细胞、和/或A549细胞。
在另一优选例中,所述的方法是体外非治疗性的。
在另一优选例中,所述的方法包括步骤:在含式I化合物的体系中培育所述对象,从而诱导所述对象产生干扰素。
在另一优选例中,所述含式I化合物的体系中,式I化合物的浓度≥1μM;较佳地,≥3μM。
在另一优选例中,培育时间为10min~48h。
在另一优选例中,所述的I型干扰素包括IFNβ。
在本发明的另一方面,还提供了一种抑制病毒感染的方法,其中,包括步骤:
使对象与式I化合物接触,从而抑制病毒对所述对象的感染。
在另一优选例中,所述的对象是细胞。
在另一优选例中,所述的细胞包括:Vero细胞、THP-1细胞、HEK293细胞、HEK293T细胞、和/或A549细胞。
在另一优选例中,所述的病毒包括DNA病毒和RNA病毒。
在另一优选例中,所述的DNA病毒包括:HSV(单纯疱疹病毒)、HBV(乙肝病毒)、HAV(甲肝病毒)、HPV(人乳头瘤病毒)和/或EBV(EB病毒)等。
在另一优选例中,所述的RNA病毒包括:VSV(水疱性口炎病毒)、HCV(丙肝病毒)、EMCV(脑心肌炎病毒)、Ebola(埃博拉病毒)、HIV(艾滋病毒)和/或Zika virus(寨卡病毒)等。
在另一优选例中,所述的方法是体外非治疗性的。
在另一优选例中,所述的方法包括步骤:在含式I化合物的体系中培育所述对象,从而诱导所述对象产生干扰素。
在本发明的另一方面,还提供了一种提高体内I型干扰素水平的方法,所述的方法包括步骤:
向需要的对象施用式I化合物,或其药学上可接受的盐。
在另一优选例中,所述的对象是哺乳动物;较佳地,选自下组:人、小鼠、和大鼠。
在本发明的另一方面,还提供了一种广谱抗病毒药物组合物,所述的药物组合物包含:式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
在本发明的另一方面,还提供了一种免疫组合物,所述的免疫组合物包含:式I化 合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和抗原。
在另一优选例中,所述抗原包括蛋白抗原。
在另一优选例中,所述的蛋白抗原包括白蛋白抗原。
在另一优选例中,所述的白蛋白抗原包括卵白蛋白抗原。
在本发明的另一方面,还提供了一种抗肿瘤药物组合物,所述的药物组合物包含:式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
在本发明的另一方面,还提供了一种组合物,所述组合物包括
(a)第一活性成分,所述第一活性成分为式I所示化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和
(b)治疗有效量的第二活性成分,所述第二活性成分包括:免疫治疗激活剂、I型干扰素调节剂、或抗病毒剂。
在本发明的另一方面,还提供了一种药盒,所述药盒包括:
(A)含有第一活性成分的第一制剂,所述第一活性成分为式I所示化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和
(B)含有第二活性成分的第二制剂,所述第二活性成分包括:免疫治疗激活剂、I型干扰素调节剂、或抗病毒剂。
在另一优选例中,所述的免疫治疗激活剂包括PD-1或者PD-L1抗体,本发明式I所示化合物和PD-1或者PD-L1抗体联合使用,能发挥协同作用,显著增加对肿瘤生长的抑制作用,明显提高生存期。
在本发明的另一方面,还提供了上述的组合物或上述药盒的用途,用于制备固有免疫激活药物,所述药物包括:
(a)广谱抗病毒药物;
(b)抗肿瘤药物;
(c)预防和/或治疗癌症的药物;
(d)治疗或预防I型干扰素相关疾病的药物;
(e)免疫佐剂;
和/或(f)细胞因子诱导剂。
本发明固有免疫激活药物的主要优点包括:
(a)本发明发现式I化合物表现出很强的诱导干扰素(如IFNβ)表达的能力。
(b)本发明发现式I化合物具有优异的抑制病毒感染能力以及抗病毒活性。
(c)本发明发现式I化合物能够高效且安全诱导细胞因子的产生,从而用于治疗与细胞因子相关的疾病例如肿瘤等疾病。
(d)本发明发现式I化合物能够用作免疫佐剂,显著提高抗体的免疫滴度,促进抗体的产生,从而用于制备抗体、疫苗、免疫治疗药物和免疫激活剂,提高抗体的应用价值。
(e)本发明所述化合物药效高、成药性好和副作用低。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
下面结合附图和具体实施方式对本发明作进一步详细的说明。
图1是本发明实施例1的分别使用终浓度为10μM的化合物LD001-LD004处理12小时后,HEK293T细胞中I型干扰素及白介素-6的转录激活结果图;
图2是本发明实施例1的分别使用终浓度为10μM的化合物LD001-LD004处理12小时后,HeLa细胞中I型干扰素及白介素-6的转录激活结果图;
图3是本发明实施例1的分别使用终浓度为10μM的化合物LD001-LD004处理12小时后,A549细胞中I型干扰素及白介素-6的转录激活结果图;
图4是本发明实施例1的分别使用终浓度为10μM的化合物LD005-LD011处理12小时后,HEK293T细胞中I型干扰素及白介素-6的转录激活结果图;
图5是本发明实施例1的分别使用终浓度为10μM的化合物LD005-LD011处理12小时后,PEM细胞中I型干扰素及白介素-6的转录激活结果图;
图6是本发明实施例2的LD002对VSV-GFP的抑制曲线图;
图7是本发明实施例2的LD002对H1N1-IAV的抑制曲线图;
图8是本发明实施例3的LD002对HSV的抑制曲线图;
图9是本发明实施例4的化合物LD002在小鼠体内诱导产生干扰素的ELISA试验结果图;
图10是本发明实施例5的化合物LD002单独给药抑制黑色素瘤生长的实验结果图;
图11是本发明实施例5的化合物LD002与另一免疫治疗激活剂anti-PD-L1联合给药抑制黑色素瘤生长的实验结果图;
图12是本发明实施例6的ELISA法检测本发明化合物具有免疫佐剂活性的结果图。
具体实施方式
发明人经过广泛而深入地研究,发现一类三元杂环化合物能够激活宿主固有免疫,诱导不同的细胞表达或产生I型干扰素IFNβ等细胞因子。此外,这类化合物对RNA病毒(如VSV)和DNA病毒(如HSV)感染具有广谱的抑制作用(剂量相关)。从而提供了一种具有诱导细胞产生IFNβ,且在临床治疗中具有潜在的广谱抗病毒作用的新药物。意外 发现本发明所述的式I化合物对细胞因子具有优异的诱导作用,用于治疗例如肿瘤等疾病。同时,本发明所述的式I化合物还能够用作免疫佐剂显著提高提高抗体的免疫滴度,促进抗体的产生,从而用于制备抗体、疫苗、免疫治疗药物和免疫激活剂,提高抗体的应用价值。
固有免疫
宿主的固有免疫是免疫系统防御病原微生物的第一道防线。自然界中病毒是一种能够引起多种疾病的病原体,宿主细胞抗病毒的固有免疫反应能够诱导产生干扰素来抑制病毒的繁殖和扩增。抗病毒固有免疫反应由信号转导通路来介导,前期研究揭示了固有免疫系统如何识别病毒入侵进而迅速地触发免疫防御功能,从而有效地清除病毒感染。这是生命科学与医学领域中的重要研究,为相关药物的设计提供理论依据,为病毒感染的防治带来新的希望。宿主细胞检测到病原微生物后通过固有免疫信号通路诱导产生干扰素、促炎因子以及细胞趋化因子,这些效应因子一方面可以抑制病原体复制,从而起到快速控制病原体增殖的作用;另一方面这些细胞因子可以动员宿主的其他免疫细胞包括适应性免疫系统中的抗原呈递细胞及T、B淋巴细胞,激活适应性免疫。除此之外,固有免疫反应的诱导激活还具有抗肿瘤的作用,能够强化放疗和化疗的效果。
固有免疫信号通路起始于宿主细胞的模式识别受体对病原微生物的相关分子模式的识别。目前已经发现的与病毒感染相关的固有免疫信号通路主要包括细胞质中识别RNA病毒的RLR-MAVS信号通路和识别DNA病毒的cGAS-STING信号通路,以及TLRs信号通路。RLRs和cGAS这两条信号通路激活后都通过下游的转导蛋白招募并激活TBK1,TBK1使转录因子IRF3发生磷酸化入核诱导I型干扰素的产生,最终达到抑制病毒的效果。在长期进化过程中,有些病毒也获得了一些逃逸固有免疫反应的机制。
由于固有免疫在机体生命活动的各个环节都扮演着重要的角色,固有免疫对于疫苗的研制和药物的开发都具有重要的指导意义。
术语
活性成分
如本文所用,术语“本发明化合物”指式I所示的化合物。该术语还包括式I化合物的各种晶型形式、药学上可接受的盐、异构体、水合物、溶剂合物或前药。
其中,术语“可药用盐”或“药学上可接受的盐”指本发明化合物与酸或碱所形成的适合用作药物的盐。药学上可接受的盐包括无机盐和有机盐。一类优选的盐是本发明化合物与酸形成的盐。适合形成盐的酸包括但并不限于:盐酸、氢溴酸、氢氟酸、硫酸、硝酸、磷酸等无机酸;甲酸、乙酸、三氟乙酸、丙酸、草酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、苯甲酸、甲磺酸、乙磺酸、对甲苯磺酸、苯磺酸、萘磺酸等有机酸;以及脯氨酸、苯丙氨酸、天冬氨酸、谷氨酸等氨基酸。另一类优选的盐是本发明化合物与碱形成的盐,例如碱金属盐(例如钠盐或钾盐)、碱土金属盐(例如镁盐或钙盐)、 铵盐(如低级的烷醇铵盐以及其它药学上可接受的胺盐),例如甲胺盐、乙胺盐、丙胺盐、二甲基胺盐、三甲基胺盐、二乙基胺盐、三乙基胺盐、叔丁基胺盐、乙二胺盐、羟乙胺盐、二羟乙胺盐、三羟乙胺盐,以及分别由吗啉、哌嗪、赖氨酸形成的胺盐。
本发明所述的如式I化合物可通过常规方法转化为其药学上可接受的盐,例如,可将相应的酸的溶液加入到上述化合物的溶液中,成盐完全后除去溶剂即得本发明所述化合物的相应的盐。
如本文所用,术语“前药”包括其本身可以是具有生物学活性的或非活性的前药,当用适当的方法服用后,其在人体内进行代谢或化学反应而转变成式I的一类化合物,或式I的一个化合物所组成的盐或溶液。所述的前药包括(但不局限于)所述化合物的羧酸酯、碳酸酯、磷酸酯、硝酸酯、硫酸酯、砜酯、亚砜酯、氨基化合物、氨基甲酸盐、偶氮化合物、磷酰胺、葡萄糖苷、醚、乙缩醛等形式。
第一活性成分
本发明的第一活性成分为式I化合物,或其药学上可接受的盐、或其光学异构体、或其外消旋体、或其溶剂化物、或其前药。
如本文所用,“本发明化合物”、“本发明式I化合物”、或“式I化合物”可互换使用,指式I化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药。应理解,该术语还包括上述组分的混合物。
Figure PCTCN2022136399-appb-000005
式I中,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10各自独立地选自氢、卤素、烷基、环烷基、芳基、杂芳基、杂环基、烯基、炔基、氨基、羟基、巯基、羧基、烷氧基、环烷氧基、卤代烷基、氰基、硫烷基、磺基、砜基、亚砜基、磷酸基,所述烷基、环烷基、烷氧基、环烷氧基、氨基、杂环基、芳基或杂芳基可被一个或多个取代基所取代,所述取代基选自羟基、卤素、烷基、烷氧基、环烷基、环烷氧基、杂环基、芳基、卤代芳基、卤代烷基、杂芳基、烯基、炔基、氨基、巯基、羧基、酯基、烷氧基羰基、酰基氧基、酰胺基、脲基、烷基磺酰基、芳香磺酰基、氰基、硝基、亚硝基、硫氰基、异硫氰基、硫烷基、磺基、磷酸基、膦酸基、烷基磷酸基、烷基膦酸基、芳基磷酸基、芳基膦酸基中的一个或多个取代基,或为空;所述杂环基包含至少一个N原子,或者包含1个或2个或3个任选于N、S和O的杂原子。
本发明优选的化合物包括:
Figure PCTCN2022136399-appb-000006
本发明优选的化合物药用盐包括:
Figure PCTCN2022136399-appb-000007
Figure PCTCN2022136399-appb-000008
第二活性成分
本发明的第二活性成分为抗病毒、抗肿瘤、免疫调节和免疫佐剂药物,可以和PD-1或者PD-L1抗体联用进行免疫治疗。
如本文所用,术语“PD-L1”称为细胞程序性死亡配体1,programmed cell death-ligand 1,是一种重要的免疫抑制分子,PD-L1会表达在肿瘤表面,与T细胞表面的PD-1结合,抑制T细胞增殖以及细胞因子分泌,负调控淋巴细胞的激活,参与肿瘤的免疫逃逸。
药物组合物和施用方法
由于本发明化合物具有优异的诱导I型干扰素产生的能力,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于治疗、预防以及缓解与I型干扰素相关的疾病(尤其是那些能够通过调节体内干扰素水平如使干扰素水平增加从而被改善、治疗或预防的疾病)。根据现有技术,本发明化合物可用于治疗以下疾病:病毒感染、多发性硬化症、系统性红斑狼疮和慢性粒细胞白血病等。
如本文所用,术语“诱导产生干扰素”、“诱导产生I型干扰素”、“诱导I型干扰素的产生”或“诱导干扰素的产生”可以互换使用,意在包括:提升I型干扰素或与I型干扰素相关的蛋白、细胞因子等的表达、转录或蛋白含量水平等。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-500mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组 合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温
Figure PCTCN2022136399-appb-000009
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。 活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物联合给药。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
组合物或制剂、活性成分的组合和药盒和施用方法
本发明还提供一种组合物或制剂、活性成分的组合和药盒,所述的组合物或制剂、活性成分的组合和药盒可用于制备固有免疫激活药物,所述药物包括:(a)广谱抗病毒药物;(b)抗肿瘤药物;(c)预防和/或治疗癌症的药物;(d)治疗或预防I型干扰素相关疾病的药物;(e)免疫佐剂;和/或(f)细胞因子诱导剂。
本发明所述的组合物优选为药物组合物。本发明所述的组合物可以包括药学上可接受的载体。
药物制剂应与给药方式相匹配,优选的给药方式为口服给药、注射给药(如瘤内注射),使用时,是将治疗有效量的药物施用于所需对象(如人或非人哺乳动物)。如本文所用,术语“治疗有效量”,是指对人和/或动物产生功能或活性的且可被人和/或动物所接受的量。本领域的普通技术人员应该理解,所述的“治疗有效量”可随着药物组合物的形式、给药途径、所用药物的辅料、疾病的严重程度以及与其他药物联合用药等情况的不同而有所不同。
在一个施用方式中,第一活性成分的安全有效日使用剂量通常至少约0.1mg,而且在大多数情况下不超过约2500mg。较佳地,该剂量是1mg-500mg;第二活性成分的安全有效量通常至少约0.01mg,而且在大多数情况下不超过2500mg。较佳地,该剂量范围是0.1mg至2500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是在熟练医师技能范围之内的。
实施例1 本发明化合物诱导细胞分泌干扰素
首先,在HEK293T、HeLa、A549及PEM等不同类型的细胞里,检测下述11种新合成的小分子化合物(LD001-LD011)诱导产生I型干扰素的活性。
Figure PCTCN2022136399-appb-000010
Figure PCTCN2022136399-appb-000011
所用293T细胞系,HeLa细胞系以及A549细胞系均购于中科院上海生科院细胞库。单细胞层在37℃和5%CO 2培养箱中培养,细胞生长在含10%FBS及100U/ml青霉素和100μg/ml链霉素的DMEM培养基中。PEM细胞为从C57BL/6小鼠腹腔中分离得到的巨噬细胞。
将处于对数生长期的HEK293T、HeLa、A549及PEM等各种细胞以80~90%的接 种密度分别铺于12孔细胞培养板,待细胞贴壁6小时后,将细胞培养上清换置成含有终浓度为10μM的上述LD001-LD011各种小分子化合物的培养基,并继续培养12小时,最终收集细胞,提取细胞总RNA。
接着利用qPCR检测各种细胞在药物处理下诱导I型干扰素及白介素-6的转录激活,结果表明LD002可以较为显著地促进HEK293T、HeLa、A549等三种细胞中I型干扰素转录,同时白介素-6的转录水平也有一定程度升高(图1,图2和图3)。另外,结果表明LD007和LD010可以在HEK293T和PEM细胞中明显诱导I型干扰素表达,LD005可以在PEM细胞中显著诱导I型干扰素的表达(图4和图5)。总之,由图1-5可知,化合物LD001-LD011在HEK293T、HeLa、A549或PEM细胞中都具有诱导I型干扰素或白介素-6表达的活性。考虑到LD007和LD010对细胞的生长具有抑制作用,表现出较强的细胞毒性,而LD005在PBS中的溶解度较低,因此本发明选择化合物LD002作为代表性化合物进行后续实施例的研究。
实施例2 本发明化合物LD002对RNA病毒(VSV和H1N1-IAV)的抑制
前面实施例1的结果已证明LD002可以较为显著地诱导细胞中I型干扰素的表达,接着利用病毒的细胞感染模型,检测LD002对于病毒扩增的抑制作用。选用水泡性口炎病毒(VSV)和流感病毒(IAV)作为RNA病毒的代表;具体使用VSV-GFP和H1N1病毒株,VSV-GFP自身能够表达绿色荧光蛋白GFP,有利于指示病毒的增殖和载量。
将处于对数生长期的A549细胞以80~90%的接种密度铺于12孔细胞培养板,待细胞贴壁6小时后,加入感染复数(MOI)为0.5的VSV病毒的稀释液。病毒感染一小时后,用不同浓度本发明化合物LD002处理,12小时后进行全细胞RNA的提取并检测病毒扩增情况。根据不同浓度LD002处理A549细胞后VSV-GFP感染受到抑制的程度计算出LD002对VSV-GFP感染抑制作用的EC 50(半数有效浓度)为1.789,SI(选择指数)为135.1(图6),结果表明本发明化合物具有优异的抗VSV病毒的活性以及安全性。
同样的,将处于对数生长期的A549细胞以80~90%的接种密度铺于12孔细胞培养板,待细胞贴壁6小时后,加入感染复数(MOI)为0.5的H1N1-IAV病毒的稀释液。病毒感染一小时后,用不同浓度本发明化合物LD002处理,12小时后进行全细胞RNA的提取并检测病毒扩增情况。根据不同浓度LD002处理A549细胞后H1N1-IAV感染受到抑制的程度计算出了LD002对H1N1-IAV感染抑制作用的EC 50(半数有效浓度)为4.735,SI(选择指数)为52.79(图7),结果表明本发明化合物具有较好的抗IAV病毒的活性以及安全性。
实施例3 本发明化合物LD002对DNA病毒(如HSV)感染的抑制作用
选用单纯疱疹病毒HSV作为DNA病毒的代表;具体使用HSV-1-GFP病毒株,自身能够表达绿色荧光蛋白GFP,可指示病毒的增殖和载量。
将处于对数生长期的A549细胞以80~90%的接种密度铺于12孔细胞培养板,待细胞贴壁6小时后,加入感染复数(MOI)为0.5的HSV病毒的稀释液。病毒感染一小时后,用不同浓度本发明化合物LD002处理,12小时后进行全细胞RNA的提取并检测病毒扩增情况。根据不同浓度LD002处理A549细胞后HSV-GFP感染受到抑制的程度计算出了LD002对HSV-GFP感染抑制作用的EC 50(半数有效浓度)为1.948,SI(选择指数)为123.92(图8),结果表明本发明化合物具有优异的抗HSV病毒的活性以及安全性。
实施例4 本发明化合物LD002在小鼠体内诱导产生干扰素
所有实验所用小鼠均为分子细胞卓越中心动物实验技术平台提供的6-8周龄的雄性C57BL/6小鼠。给小鼠通过腹腔注射不同剂量(100μg或300μg)的本发明化合物LD002,6小时后,利用ELISA检测血清中各种细胞因子的含量变化。结果显示,本发明化合物LD002实验组可明显提高血清中I型干扰素IFNβ(干扰素-β)、IL1β(白细胞介素-1β)的水平,可轻微提高IL-6(白细胞介素-6)、TNFα(肿瘤坏死因子-α)及III型干扰素IFNλ(干扰素-λ)的表达水平(图9)。图9中,Ctrl为腹腔注射PBS的对照组,注射剂量为100μL/每只。
实施例5 本发明化合物LD002具有抗肿瘤作用以及促进肿瘤免疫治疗的功能
1.方法
本实验所用B16-F10黑色素瘤细胞株购买于上海中科院细胞库,常规培养于含10%FBS的DMEM培养基中,置于5%CO 2培养箱中37℃培养。指数增殖期的细胞经0.25%胰酶消化、离心后用PBS重悬计数,检测活细胞率在95%以上,计数后调节细胞悬液浓度为2*10^ 6mL -1。将重悬的肿瘤细胞分装到无菌EP管中,置于冰上待用。
用电动剃毛器将小鼠背部的毛发进行去除,去毛面积约为9cm 2。取上述准备好的B16-F10细胞,每只老鼠背部皮下注射100μL肿瘤细胞悬液。每天观察小鼠肿瘤生长情况,第4天肉眼可见黑色素瘤的形成,剔除未成瘤老鼠,对剩余老鼠进行随机分组,分为7组,每个实验组小鼠数量为5只。
实验一分组分别为对照组,100μg本发明化合物LD002组,300μg本发明化合物LD002组;实验二分组分别为对照组,100μg本发明化合物LD002组,200μg抗PD-L1抗体(B7-H1)(Clone:10F.9G2)组,100μg本发明化合物LD002+200μg抗PD-L1抗体(B7-H1)(Clone:10F.9G2)组,每组6-8只。实验方法为:在肿瘤细胞注射后第五天,根据分组对于荷瘤小鼠进行给药,通过经腹腔注射LD002,设置2个剂量组:100μg/只,300μg/只。Anti-PD-L1抗体经腹腔注射给药,设置1个剂量组:200μg/只。给药的溶剂缓冲体系为PBS缓冲溶液。对照组灌胃给予同体积的PBS缓冲溶液。对于本发明化合物药物,每只小鼠每2天给药1次,总共给药3次。给药体积均为100μL。对于anti-PD-L1抗体,每只小鼠每2天给药1次,总共给药3次。给药体积为 50μL。具体分别为第5、7、9天进行给药,2天为间隔,共给药3次。隔天测量肿瘤大小,观测小鼠生存期并且记录相关数据,使用电子卡尺对于肿瘤体积进行测量,体积计算公式为:π/6x长x宽x高。肿瘤体积大于2000mm 3时处死荷瘤小鼠。
2.实验结果
本发明化合物LD002抑制小鼠皮下黑色素瘤的生长,提高荷瘤小鼠的生存期限。
通过建立皮下黑色素瘤荷瘤小鼠模型,检测本发明化合物LD002以及LD002+anti-PD-L1对于肿瘤生长的抑制作用及提高荷瘤小鼠生存期,结果显示,相比对照组,单独给药本发明化合物对黑色素瘤的生长具有显著的剂量依赖性抑制作用,且提高荷瘤小鼠的生存期(图10)。
相比对照组和单独给药本发明化合物LD002,anti-PD-L1能够显著增强本发明化合物对黑色素瘤生长的抑制作用和提高小鼠的生存期。从结果数据可以看出,本发明化合物LD002和anti-PD-L1能够联合发挥协同作用,对黑色素瘤生长的抑制作用和提高小鼠的生存期更佳显著(图11)。
从实施例4、5可以看出,本发明化合物LD002能够诱导外周血中干扰素等多种细胞因子的升高,对肿瘤如黑色素瘤具有优异的抑制作用且提高生存期。本发明化合物LD002和anti-PD-L1联用在对肿瘤生长的抑制作用和提高生存期方面具有协同作用。因此,本发明化合物能够作为一种诱导干扰素等多种细胞因子的药物,具有抗肿瘤作用以及促进肿瘤免疫治疗的功能。
实施例6 本发明化合物LD002具有免疫佐剂活性
实验分为空白组(注射相同体积的PBS),抗原组和抗原/本发明化合物组(包含不同浓度梯度),每组5只小鼠。分别将抗原卵清蛋白OVA和本发明化合物LD002用PBS溶解,得到抗原溶液和本发明化合物溶液,并按照浓度需求配制成包含抗原及不同浓度梯度本发明化合物的溶液。实验组每只小鼠每次用100μl抗原溶液或抗原溶液和LD002溶液免疫小鼠,免疫方式为肌肉注射。空白组小鼠注射与实验组相同体积的PBS溶液。第一次免疫设为第0天,分别于第7天和第14天加强免疫一次,第21天免疫结束后,小鼠眼眶取血,收集小鼠血清,稀释后用ELISA法检测小鼠血清中抗体含量。结果显示,与抗原组相比,单纯OVA免疫后并不能产生保护性体液免疫反应,当OVA联合本发明化合物免疫小鼠可显著提高OVA特异性IgG、IgG1以及IgG2b抗体总量,表明本发明化合物具有明显的免疫佐剂活性(图12)。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。

Claims (14)

  1. 一种式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药的用途,用于制备固有免疫激活药物,
    Figure PCTCN2022136399-appb-100001
    其中,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10各自独立地选自氢、卤素、烷基、环烷基、芳基、杂芳基、杂环基、烯基、炔基、氨基、羟基、巯基、羧基、烷氧基、环烷氧基、卤代烷基、氰基、硫烷基、磺基、砜基、亚砜基、磷酸基,所述烷基、环烷基、烷氧基、环烷氧基、氨基、杂环基、芳基或杂芳基可被一个或多个取代基所取代,所述取代基选自羟基、卤素、烷基、烷氧基、环烷基、环烷氧基、杂环基、芳基、卤代芳基、卤代烷基、杂芳基、烯基、炔基、氨基、巯基、羧基、酯基、烷氧基羰基、酰基氧基、酰胺基、脲基、烷基磺酰基、芳香磺酰基、氰基、硝基、亚硝基、硫氰基、异硫氰基、硫烷基、磺基、磷酸基、膦酸基、烷基磷酸基、烷基膦酸基、芳基磷酸基、芳基膦酸基中的一个或多个取代基,或为空;所述杂环基包含至少一个N原子,或者包含1个或2个或3个任选于N、S和O的杂原子。
  2. 如权利要求1所述的用途,其特征在于,所述式I化合物包括:
    Figure PCTCN2022136399-appb-100002
    Figure PCTCN2022136399-appb-100003
  3. 如权利要求1或2所述的用途,其特征在于,所述式I化合物的可药用盐包括:
    Figure PCTCN2022136399-appb-100004
    Figure PCTCN2022136399-appb-100005
  4. 如权利要求1所述的用途,其特征在于,所述固有免疫激活药物包括:
    (a)广谱抗病毒药物;
    (b)抗肿瘤药物;
    (c)预防和/或治疗癌症的药物;
    (d)治疗或预防I型干扰素相关疾病的药物;
    (e)免疫佐剂;
    和/或(f)细胞因子诱导剂。
  5. 如权利要求4所述的用途,其特征在于,所述的免疫佐剂用于制备抗体、疫苗、免疫治疗药物和/或免疫激活剂。
  6. 一种广谱抗病毒药物组合物,其特征在于,所述的药物组合物包含:权利要求1或2所述的式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  7. 一种免疫组合物,其特征在于,所述的免疫组合物包含:权利要求1或2所述的式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和抗原。
  8. 一种抗肿瘤药物组合物,其特征在于,所述的药物组合物包含:权利要求1或2所述的式I化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  9. 一种组合物,其特征在于,所述组合物包括
    (a)第一活性成分,所述第一活性成分为式I所示化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和
    (b)治疗有效量的第二活性成分,所述第二活性成分包括:免疫治疗激活剂、I型干扰素调节剂、或抗病毒剂。
  10. 一种药盒,其特征在于,所述药盒包括:
    (A)含有第一活性成分的第一制剂,所述第一活性成分为式I所示化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药;和
    (B)含有第二活性成分的第二制剂,所述第二活性成分包括:免疫治疗激活剂、I型干扰素调节剂、或抗病毒剂。
  11. 权利要求9所述的组合物或权利要求10所述药盒的用途,其特征在于,用于制备固有免疫激活药物,所述药物包括:
    (a)广谱抗病毒药物;
    (b)抗肿瘤药物;
    (c)预防和/或治疗癌症的药物;
    (d)治疗或预防I型干扰素相关疾病的药物;
    (e)免疫佐剂;
    和/或(f)细胞因子诱导剂。
  12. 一种式I化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药,
    Figure PCTCN2022136399-appb-100006
    其中,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10各自独立地选自氢、卤素、烷基、环烷基、芳基、杂芳基、杂环基、烯基、炔基、氨基、羟基、巯基、羧基、烷氧基、环烷氧基、卤代烷基、氰基、硫烷基、磺基、砜基、亚砜基、磷酸基,所述烷基、环烷基、烷氧基、环烷氧基、氨基、杂环基、芳基或杂芳基可被一个或多个取代基所取代,所述取代基选自羟基、卤素、烷基、烷氧基、环烷基、环烷氧基、杂环基、芳基、卤代芳基、卤代烷基、杂芳基、烯基、炔基、氨基、巯基、羧基、酯基、烷氧基羰基、酰基氧基、酰胺基、脲基、烷基磺酰基、芳香磺酰基、氰基、硝基、亚硝基、硫氰基、异硫氰基、硫烷基、磺基、磷酸基、膦酸基、烷基磷酸基、烷基膦酸基、芳基磷酸基、芳基膦酸基中的一个或多个取代基,或为空;所述杂环基包含至少一个N原子,或者包含1个或2个或3个任选于N、S和O的杂原子。
  13. 如权利要求12所述的化合物,其特征在于,所述式I化合物包括:
    Figure PCTCN2022136399-appb-100007
    Figure PCTCN2022136399-appb-100008
  14. 如权利要求12所述的化合物,其特征在于,所述式I化合物的可药用盐包括:
    Figure PCTCN2022136399-appb-100009
PCT/CN2022/136399 2021-12-25 2022-12-02 固有免疫激活药物及其用途 WO2023116397A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280004982.4A CN116157126A (zh) 2021-12-25 2022-12-02 固有免疫激活药物及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111605896.2 2021-12-25
CN202111605896 2021-12-25

Publications (1)

Publication Number Publication Date
WO2023116397A1 true WO2023116397A1 (zh) 2023-06-29

Family

ID=86901203

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/136399 WO2023116397A1 (zh) 2021-12-25 2022-12-02 固有免疫激活药物及其用途

Country Status (1)

Country Link
WO (1) WO2023116397A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991002529A2 (en) * 1989-08-14 1991-03-07 John Bennett Kizer Product and method for killing abnormal vertebrate cells
CN101856355A (zh) * 2009-04-12 2010-10-13 蔡荣 阿的平及其代用品对乙型病毒性肝炎的治疗作用
WO2015153535A1 (en) * 2014-03-31 2015-10-08 MiRx Pharmaceuticals, LLC Novel hdmx inhibitors and their use for cancer treatment
CN113842386A (zh) * 2020-06-28 2021-12-28 中国科学院分子细胞科学卓越创新中心 具有广谱抗病毒活性的药物
CN115212209A (zh) * 2021-04-19 2022-10-21 中国科学院分子细胞科学卓越创新中心 一种具有免疫佐剂活性和促进免疫疗法的抗肿瘤药物

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991002529A2 (en) * 1989-08-14 1991-03-07 John Bennett Kizer Product and method for killing abnormal vertebrate cells
CN101856355A (zh) * 2009-04-12 2010-10-13 蔡荣 阿的平及其代用品对乙型病毒性肝炎的治疗作用
WO2015153535A1 (en) * 2014-03-31 2015-10-08 MiRx Pharmaceuticals, LLC Novel hdmx inhibitors and their use for cancer treatment
CN113842386A (zh) * 2020-06-28 2021-12-28 中国科学院分子细胞科学卓越创新中心 具有广谱抗病毒活性的药物
CN115212209A (zh) * 2021-04-19 2022-10-21 中国科学院分子细胞科学卓越创新中心 一种具有免疫佐剂活性和促进免疫疗法的抗肿瘤药物

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GLAZ E. T., SZOLGAY ERIKA, STOGER IVANA, TALAS ' MARGARITA: "Antiviral Activity and Induction of Interferon-Like Substance by Quinacrine and Acranil", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, 1 May 1973 (1973-05-01), pages 537 - 541, XP055893190, DOI: 10.1128/AAC.3.5.537 *
ISHIGAMI-YUASA MARI; WATANABE YUKO; MORI TAKAYASU; MASUNO HIROYUKI; FUJII SHINYA; KIKUCHI ERIKO; UCHIDA SHINICHI; KAGECHIKA HIROYU: "Development of WNK signaling inhibitors as a new class of antihypertensive drugs", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 25, no. 14, 19 May 2017 (2017-05-19), AMSTERDAM, NL, pages 3845 - 3852, XP085062457, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2017.05.034 *
MICHELE TONELLI; GEROLAMO VETTORETTI; BRUNO TASSO; FEDERICA NOVELLI; VITO BOIDO; FABIO SPARATORE; BERNARDETTA BUSONERA; AICHA OUHT: "Acridine derivatives as anti-BVDV agents", ANTIVIRAL RESEARCH, vol. 91, no. 2, 8 May 2011 (2011-05-08), NL , pages 133 - 141, XP028236207, ISSN: 0166-3542, DOI: 10.1016/j.antiviral.2011.05.005 *
MILLICH FRANK, OSTER GERALD: "Photoreduction of Acridine Dyes", J. AM. CHEM. SOC., 20 March 1959 (1959-03-20), pages 1357 - 1363, XP093074579, DOI: 10.1021/ja01515a023 *

Similar Documents

Publication Publication Date Title
JP7050165B2 (ja) Hbv複製阻害剤としての置換ピロリジン化合物
US20170342068A1 (en) Compounds for the treatment of hepatitis b virus infection
Nain et al. Tumor necrosis factor-alpha production of influenza A virus-infected macrophages and potentiating effect of lipopolysaccharides.
CA2997955A1 (en) Modulators of toll-like receptors for the treatment of hiv
AU2018372851B2 (en) Bisdiazabicyclo compound for treating and/or preventing hepatitis virus-related diseases or disorders
JP2008255121A (ja) 抗b型肝炎ウイルス活性を有するヌクレオシド
JP2017537166A (ja) 癌免疫療法におけるアジュバントとしてのブロモドメイン阻害剤
JP2022009683A (ja) テノホビルアラフェナミドの結晶形態
US20220289763A1 (en) Macrocyclic Compounds as Sting Agonists and Methods and Uses Thereof
JP2020503344A (ja) 腫瘍溶解性ワクシニアウイルスおよびnk細胞を含む治療剤、ならびに腫瘍および/またはがんの治療のための薬物のためのその使用
US20240065987A1 (en) Anti-cancer activity of adamantane derivatives
WO2023116397A1 (zh) 固有免疫激活药物及其用途
CN113842386A (zh) 具有广谱抗病毒活性的药物
CN112121044A (zh) 氨来占诺用于制备抗肝炎病毒药物的用途
US20230165870A1 (en) 6,8-dioxa-3-azabicyclo[3.2.l]Octane Carboxylic Acids And Their Derivatives For Use In The Treatment Of Inflammations
CN116157126A (zh) 固有免疫激活药物及其用途
US20160279236A1 (en) Use of rapamycin as vaccine adjuvant and preparation method therefor
CN115212209A (zh) 一种具有免疫佐剂活性和促进免疫疗法的抗肿瘤药物
EP4265268A2 (en) Cigb-300 for use in the the induction of antitumor and antiviral immunty
JP2023551018A (ja) エンベロープウィルス感染の治療のための化合物
EP0471057A4 (en) Methods and compositions for the prophylaxis and treatment of cytomegalovirus infections
TW202406544A (zh) 哌類化合物和pd-1抑制劑或pd-l1抑制劑的組合物以及其在製備抗腫瘤藥物中的用途
CN113940933A (zh) 抗新型冠状病毒的药物
JPS6136728B2 (zh)
JPWO2021014380A5 (zh)

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: 22909712

Country of ref document: EP

Kind code of ref document: A1