CN116966147A - Novel STING agonist delivery system and immunotherapeutic application - Google Patents

Novel STING agonist delivery system and immunotherapeutic application Download PDF

Info

Publication number
CN116966147A
CN116966147A CN202210432161.2A CN202210432161A CN116966147A CN 116966147 A CN116966147 A CN 116966147A CN 202210432161 A CN202210432161 A CN 202210432161A CN 116966147 A CN116966147 A CN 116966147A
Authority
CN
China
Prior art keywords
compound
optionally
pharmaceutical composition
branched alkyl
linear
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210432161.2A
Other languages
Chinese (zh)
Inventor
李艳梅
郭东升
陈永湘
吴军军
陈方远
李娟娟
韩贝贝
岳宇昕
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tsinghua University
Nankai University
Original Assignee
Tsinghua University
Nankai University
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 Tsinghua University, Nankai University filed Critical Tsinghua University
Priority to CN202210432161.2A priority Critical patent/CN116966147A/en
Publication of CN116966147A publication Critical patent/CN116966147A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1273Polymersomes; Liposomes with polymerisable or polymerised bilayer-forming substances
    • 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
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Dispersion Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention relates to the technical field of drug delivery, in particular to a novel STING agonist delivery system and an application of immunotherapy. The present invention provides a pharmaceutical composition comprising: a drug to be delivered comprising a STING agonist, and a delivery vehicle comprising compound (i) or a pharmaceutically acceptable salt thereofThe invention provides a macrocyclic molecule nano delivery system based on amphiphilic calixarene, which is used for loading and delivering STING agonist CDN. Selection of chemically derived cyclic dinucleotide analogues CDG SF Delivery efficacy and application validation was performed as representative of STING agonists. By means of the ultra-strong interaction force of host and guest between the cyclic dinucleotide and the calixarene, the calixarene system can efficiently load the cyclic dinucleotide and can respond to tumor tissues and intracellular high-concentration ATP to rapidly release the cyclic dinucleotide.

Description

Novel STING agonist delivery system and immunotherapeutic application
Technical Field
The invention relates to the technical field of drug delivery, in particular to a novel STING agonist delivery system and an application of immunotherapy.
Background
Tumor immunotherapy has now gradually evolved into an important and powerful direction for cancer treatment. In immunotherapy, how to effectively enhance the tumor microenvironment immune response and alleviate immune tolerance is a core problem related to the therapeutic effect. As a recently discovered novel pattern recognition receptor, the interferon gene stimulatory protein (stimulator of interferon genes, STING) pathway has attracted the development interest of numerous large pharmaceutical enterprises. The company such as Nohua, merck and Bai-shi Guibao invests in succession to develop the STING channel agonist for clinical immunotherapy of tumor. STING proteins are located on the endoplasmic reticulum and are highly expressed in T cells and antigen presenting cells and can be activated by their natural agonist cyclic dinucleotides (cyclic dinucleotides, CDNs) to facilitate the transcriptional translation of downstream host defense genes including type I interferons and other pro-inflammatory cytokines. Based on this, STING agonists such as cyclic dinucleotides can be ideal choices as therapeutic agents or vaccine adjuvants. However, the characteristics of STING agonists such as poor stability and poor transmembrane property greatly hinder the clinical application value. Therefore, the design and development of suitable methods to improve STING agonist stability and transmembrane efficiency is of great research value.
Disclosure of Invention
The present invention aims to solve at least one of the technical problems in the related art to some extent. It is therefore an object of the present invention to provide an amphiphilic calixarene-based macrocyclic molecular nanodelivery system for loading and delivering STING agonist CDN (cyclic dinucleotide) based compounds. The invention selects the CDG of the chemically derived cyclic dinucleotide analogue SF Delivery efficacy and application validation was performed as representative of STING agonists. By means of the ultra-strong interaction force of host and guest between the cyclic dinucleotide and the calixarene, the calixarene system can efficiently load the cyclic dinucleotide and can respond to tumor tissues and intracellular high-concentration ATP to rapidly release the cyclic dinucleotide.
To this end, a first aspect of the invention provides a pharmaceutical composition. According to an embodiment of the present invention, the pharmaceutical composition comprises:
the drug to be delivered and the delivery vehicle,
wherein the drug to be delivered comprises a STING agonist,
the delivery vehicle comprises a hydrated vesicle comprising a compound (I) or a pharmaceutically acceptable salt thereof,
wherein n=4-20;
R 1 selected from C 4 -C 20 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
R 2 selected from amino groups,
In order to solve the problems of poor stability and poor transmembrane property of STING agonists and influencing clinical efficacy, the inventor discovers that by long-time exploration, amphiphilic calixarene macrocyclic molecules are used as delivery vehicles to form hydration vesicles, STING agonists (such as CDN compounds) are loaded, and the calixarene system can efficiently load cyclic dinucleotides by means of super-strong guest interaction force between the STING agonists and the calixarene and can respond to tumor tissues and intracellular high-concentration ATP to rapidly release the cyclic dinucleotides. In vitro and in vivo experiments prove that the calixarene delivery system can effectively improve the transmembrane efficiency of STING agonists, promote enrichment of STING agonists in tumors and tumor-infiltrating lymph nodes, induce an immunogenic tumor microenvironment, and further effectively inhibit tumor growth and improve the survival rate of mice.
According to an embodiment of the invention, n=4-10 in the compound (i). When n in the compound (I) is 4 to 10, the hydrated vesicle structure formed by the compound (I) is more stable.
According to an embodiment of the invention, n=4-8 in the compound (i). When n in the compound (I) is 4, 5, 6 or 8, the stability of the hydrated vesicle structure formed by the compound (I) can be further improved.
According to an embodiment of the invention, R in the compound (I) 1 Selected from C 4 -C 15 A straight or branched alkyl group, said straight or branched alkyl group being unsubstituted or substituted with at least one halogen. R in the compound (I) 1 Can maintain the stability of vesicles in the hydrated vesicles formed from the compound (i) to increase the loading rate of STING agonists.
According to an embodiment of the invention, the halogen is selected from fluorine, chlorine, bromine, iodine.
According to an embodiment of the invention, the STING agonist comprises a compound selected from the CDN class.
According to the embodiment of the invention, the CDN compound has the structural general formula:
wherein B is 1 And B 2 Each independently selected from the group consisting of natural bases A, T,C. G, U and any one of unnatural bases;
Y 1 and Y 2 Each independently selected from any one of-OH and-SH;
X 1 and X 2 Each independently selected from any one of-H, -OH and-F.
According to an embodiment of the invention, the non-natural base is selected from an artificial base or a modified natural base.
According to an embodiment of the invention, the modified natural base is I (hypoxanthine) or mC (5-methylcytosine).
According to an embodiment of the invention, in the pharmaceutical composition, the molar ratio of the CDN-based compound to the compound (I) is 1:1-1:10. The molar ratio of the CDN compound to the compound (I) is in the range of 1:1-1:10, so that the efficient load rate of calixarene in the hydration vesicle to the CDN compound can be maintained, the transmembrane efficiency of the STING agonist is improved, and the enrichment of the STING agonist in tumors and tumor-infiltrating lymph nodes is promoted.
According to the embodiment of the invention, the molar ratio of the CDN compound to the compound (I) is 1:1-1:2. The molar ratio of the CDN compound to the compound (I) is in the range of 1:1-1:2, so that the efficient load rate of calixarene in the hydration vesicle to the CDN compound can be maintained, the transmembrane efficiency of the STING agonist is further improved, and the enrichment of the STING agonist in tumors and tumor-infiltrating lymph nodes is promoted.
According to an embodiment of the invention, the pharmaceutical composition further comprises a stabilizer for extending the half-life of the drug. The stabilizers are useful for increasing the metabolic half-life and stability of drug-loaded delivery vehicles.
According to an embodiment of the invention, the stabilizer comprises a stabilizer selected from the group consisting of C 4 -C 20 A linear or branched alkyl-linked PEG, said linear or branched alkyl being unsubstituted or substituted with at least one halogen. The halogen is selected from fluorine, chlorine, bromine and iodine. The stabilizer contained in the pharmaceutical composition provided by the invention is not limited to C 4 -C 20 The PEG linked to the linear or branched alkyl group of (2) may also be other stabilizers known in the art, as long as they can also beCan function to increase the metabolic half-life and stability of the drug-loaded delivery vehicle, and are included within the scope of the present invention.
According to an embodiment of the present invention, the degree of polymerization of the PEG is 1000 to 5000. Thereby, the metabolic half-life and stability of the STING agonist-loaded hydrated vesicles can be further improved.
According to an embodiment of the present invention, in the pharmaceutical composition, the compound (i) or a pharmaceutically acceptable salt thereof and the and C 4 -C 20 The molar ratio of the PEG connected with the straight chain or branched chain alkyl is 1 (0.02-10). Thereby, the metabolic half-life and stability of the STING agonist-loaded hydrated vesicles can be further improved.
According to an embodiment of the present invention, in the pharmaceutical composition, the compound (i) or a pharmaceutically acceptable salt thereof and the and C 4 -C 20 The molar ratio of PEG attached to the linear or branched alkyl group is 1: (0.1-1). Thus, the metabolic half-life and stability of the STING agonist-loaded hydrated vesicles can be further improved.
According to an embodiment of the invention, the pharmaceutical composition further comprises an immune checkpoint inhibitor. The inventor finds that the vesicle structure loaded with STING agonist (such as CDN compound) and the immune checkpoint inhibitor are combined, so that the therapeutic effect rate of the immune checkpoint inhibitor can be effectively improved, and immune memory is induced to be generated, so that tumor recurrence is inhibited.
According to an embodiment of the invention, the immune checkpoint inhibitor comprises at least one selected from the group consisting of anti-PD-1, anti-PD-L1, anti-CTLA-4, anti-LAG-3, anti-TIM-3, anti-TIGIT. The inventor finds that the hydration vesicle structure loaded with CDN compounds and the immune checkpoint inhibitor anti-PD-1 are combined, so that the treatment effect rate of the immune checkpoint inhibitor can be effectively improved, and immune memory is induced to be generated, so that tumor recurrence is inhibited. Other immune checkpoint inhibitors, such as anti-PD-L1, anti-CTLA-4, anti-LAG-3, anti-TIM-3, anti-TIGIT or other immune checkpoint inhibitors known in the art, can achieve a comparable clinical effect when used in combination with hydrated vesicle structures loaded with CDN-type compounds. The inventors found that the macrocyclic molecule nanodelivery system, STING agonist, in combination with immune checkpoint therapy can increase the efficacy rate of immune checkpoint therapy, further synergistically inhibiting tumor growth. The combination of the delivery system and the STING agonist can be used as a general high-efficiency delivery platform of the STING agonist, and can be widely applied to single or combined immunotherapy (anti-tumor, antiviral and antibacterial) strategies of targeting STING channels.
According to an embodiment of the invention, the pharmaceutical composition further comprises pharmaceutically acceptable excipients.
In a second aspect, the invention provides a pharmaceutical formulation. According to an embodiment of the present invention, the pharmaceutical formulation comprises the pharmaceutical composition according to the first aspect.
In a third aspect the invention provides the use of a delivery vehicle for the delivery of STING agonists. According to an embodiment of the invention, the delivery vehicle comprises a hydrated vesicle comprising compound (I) or a pharmaceutically acceptable salt thereof,
wherein n=4-20;
R 1 a linear or branched alkyl group selected from C4 to C20 unsubstituted or substituted with at least one halogen;
R 2 selected from amino groups,
According to an embodiment of the invention, n=4-10 in the compound (i).
According to an embodiment of the invention, n=4-8 in the compound (i).
According to an embodiment of the invention, R in the compound (I) 1 Selected from C 4 -C 15 A straight or branched alkyl group, said straight or branched alkyl group being unsubstituted or substituted with at least one halogen. According to the inventionIn embodiments, the halogen is selected from fluorine, chlorine, bromine, iodine.
According to an embodiment of the invention, the STING agonist comprises a compound selected from the CDN class.
According to the embodiment of the invention, the CDN compound has the structural general formula:
wherein B is 1 And B 2 Each independently selected from any one of natural base A, T, C, G, U and non-natural base;
Y 1 and Y 2 Each independently selected from any one of-OH and-SH;
X 1 and X 2 Each independently selected from any one of-H, -OH and-F.
According to an embodiment of the invention, the non-natural base is selected from an artificial base or a modified natural base.
According to an embodiment of the invention, the modified natural base is I or mC.
According to an embodiment of the invention, in the pharmaceutical composition, the molar ratio of the CDN-based compound to the compound (I) is 1:1-1:10.
According to the embodiment of the invention, the molar ratio of the CDN compound to the compound (I) is 1:1-1:2.
In a fourth aspect, the present invention provides the use of a pharmaceutical composition according to the first aspect for the preparation of an antitumor drug.
According to an embodiment of the invention, the tumor comprises a tumor selected from the group consisting of melanoma, colon cancer, breast cancer, lymphoma, lung cancer.
Additional aspects and advantages of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention.
Drawings
The foregoing and/or additional aspects and advantages of the invention will become apparent and may be better understood from the following description of embodiments taken in conjunction with the accompanying drawings in which:
FIG. 1 shows a CDG according to one embodiment of the invention SF Structure of @12C system, preparation scheme, GC5A-12C and representative STING agonist CDG SF Chemical structure of (a);
FIG. 2 shows the general structural formula of a STING agonist CDN-type compound according to one embodiment of the present invention, wherein B 1 And B 2 Each independently selected from any one of natural base A, T, C, G, U and non-natural base; y is Y 1 And Y 2 Each independently selected from any one of-OH and-SH; x is X 1 And X 2 Each independently selected from any one of-H, -OH, and-F;
FIG. 3 shows hydrated vesicles GC5A-12C and CDG according to one embodiment of the invention SF Particle size characterization of @12C, wherein (a) shows hydrated vesicles GC5A-12C and CDG SF Particle size characterization of @ 12C; (b) The figure shows CDG SF TEM image of @12C vesicles (scale = 200 nm);
FIG. 4 shows a CDG of one embodiment of the invention SF The expression of CD86 and CD40 by RAW264.7 macrophages and BMDC cells was stimulated by the @12C and control, respectively, wherein panel (a) shows CDG SF Results of flow analysis of CD86 expression of control stimulated RAW264.7 macrophages at 12C (CDG) SF Concentration 3 μm); (b) The figure shows CDG SF Results of flow analysis of CD40 expression from control stimulated BMDC cells at 12C (CDG) SF Concentration 0.5 μm); (c) The figure shows CDG SF Results of flow analysis of CD86 expression from control stimulated BMDC cells at 12C (CDG) SF Concentration 0.5 μm), x: p is p<0.001,*:p<0.05, ns: no significant differences;
FIG. 5 shows a CDG of one embodiment of the invention SF Effect of intratumoral injection on mice in @12C and control group, wherein
(a) The figure shows the construction and administration flow of melanoma tumor-bearing mice; (b) The figure shows CDG SF Day 82 tumor-free mouse picture @12C group; (c) The graph shows 8 μg CDG SF Intratumorally injecting tumor volume of mice; (d) The figure shows 8. Mu.g CDG SF Dosage forms of tumorSurvival curves of mice injected internally; (e) The figure shows melanoma and lymph node pictures taken after treatment of tumor-bearing mice on day 17: p is p<0.001。
FIG. 6 shows a CDG of one embodiment of the invention SF Effects of subcutaneous injections in mice in the @12C and control groups, wherein
(a) The graph shows 8 μg CDG SF Subcutaneous injection of tumor volume in mice at doses; (b) The graph shows 8 μg CDG SF Dose subcutaneous mice survival curve; (c) The graph shows 20 μg CDG SF Subcutaneous injection of tumor volume in mice at doses; (d) The graph shows 20 μg CDG SF Dose subcutaneous mice survival curve, x: p is p<0.001,*:p<0.05;
FIG. 7 shows a CDG of one embodiment of the invention SF Effect of combination of the @12C system with anti-PD-1 antibody and control group on mouse tumor, wherein (a) panel shows melanoma tumor-bearing mouse construction and CDG SF Schematic of the dosing flow of the @12C system in combination with the anti-PD-1 antibody; (b) The figure shows tumor volume curves of single mice after dosing (lower right number: number of tumor free mice/total number of mice);
FIG. 8 shows a CDG of one embodiment of the invention SF Effect of the @12C system in combination with anti-PD-1 antibody and control group on mouse tumor, wherein panel (a) shows CDG SF Mice tumor volume curves of each group were combined with anti-PD-1 antibody in the @12C system; (b) The figure shows CDG SF Mice survival curves for each group combined with anti-PD-1 antibody for the @12C system; (c) The figure shows the day 40 CDG SF Surviving mouse pictures (gray circle-tumor, white circle-no tumor) of the group and control group combined with anti-PD-1 antibody of the @12C system;
FIG. 9 shows the flow results of one embodiment of the invention, wherein (a) shows IFN-gamma + CD8 + T cell population representative flow scattergrams; (b) The figure shows IFN-gamma + CD8 + Counting the proportion of T cell groups; (c) The figure shows CD69 + CD8 + Counting the proportion of T cell groups; (d) The figure shows the statistics of the proportion of regulatory T cell (Treg) populations: p is p<0.001,**:p<0.01,*:p<0.05, ns: no significant differences.
Detailed Description
Embodiments of the present invention are described in detail below. The following examples are illustrative only and are not to be construed as limiting the invention.
It should be noted that the terms "first," "second," and "second" are used for descriptive purposes only and are not to be construed as indicating or implying a relative importance or implying a number of technical features being indicated. Thus, a feature defining "a first" or "a second" may explicitly or implicitly include one or more such feature. Further, in the description of the present invention, unless otherwise indicated, the meaning of "a plurality" is two or more.
The endpoints and any values of the ranges disclosed herein are not limited to the precise range or value, and are understood to encompass values approaching those ranges or values. For numerical ranges, one or more new numerical ranges may be found between the endpoints of each range, between the endpoint of each range and the individual point value, and between the individual point value, in combination with each other, and are to be considered as specifically disclosed herein.
In order that the invention may be more readily understood, certain technical and scientific terms are defined below. Unless clearly defined otherwise herein in this document, all other 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.
In this document, the terms "comprise" or "include" are used in an open-ended fashion, i.e., to include what is indicated by the present invention, but not to exclude other aspects.
In this document, the terms "optionally," "optional," or "optionally" generally refer to the subsequently described event or condition may, but need not, occur, and the description includes instances in which the event or condition occurs, as well as instances in which the event or condition does not.
The term "pharmaceutical composition" as used herein generally refers to unit dosage forms and may be prepared by any of the methods well known in the pharmaceutical arts. All methods include the step of combining the active ingredient with adjuvants that constitute one or more adjunct ingredients. Generally, the compositions are prepared by uniformly and sufficiently combining the active compound with a liquid adjuvant, a finely divided solid adjuvant, or both.
As used herein, the term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. Preferably, the term "pharmaceutically acceptable" as used herein refers to use in animals, particularly humans, approved by the federal regulatory agency or a state government or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia.
As used herein, the term "pharmaceutically acceptable excipient" or "pharmaceutically acceptable carrier" may include any solvent, solid excipient, diluent or other liquid excipient, and the like, as appropriate for the particular dosage form of interest. In addition to the extent to which any conventional adjuvant is incompatible with the compounds of the present invention, such as any adverse biological effects produced or interactions with any other component of the pharmaceutically acceptable composition in a deleterious manner, their use is also contemplated by the present invention.
For other pharmaceutically acceptable excipients mentioned herein, and processes therefor, reference is made to the extensive literature on this subject, see in particular Handbook of Pharmaceutical Excipients, 3 rd edition, arthur h.kibbe edit American Pharmaceutical Association, washington, USA and Pharmaceutical Press, london; and Lexikon der Hilfsstoffe f ur Pharmazie, kosmetik and angrenzende Gebiete, h.p. fiedler edit, 4 th edition, edit Cantor, aulendorf and early version.
As used herein, the term "administering" refers to introducing a predetermined amount of a substance into a patient by some suitable means. The fusion protein or pharmaceutical composition of the invention may be administered by any common route, provided that it reaches the desired tissue. Various modes of administration are contemplated, including intravenous injection, intramuscular injection, subcutaneous injection, and the like, but the invention is not limited to these illustrated modes of administration. Preferably, the compositions of the present invention are administered intravenously.
As used herein, the term "cancer" or "tumor" can be any unregulated cell growth. Illustratively, non-small cell lung cancer, papillary thyroid cancer, glioblastoma multiforme, colon cancer, rectal cancer, lung cancer, head and neck cancer, kidney cancer, bladder cancer, breast cancer, ovarian cancer, liver cancer, cholangiocarcinoma or sarcoma, acute myelogenous leukemia, large cell neuroendocrine cancer, neuroblastoma, prostate cancer, neuroblastoma, pancreatic cancer, melanoma, head and neck squamous cell carcinoma, cervical cancer, skin cancer, glioma, esophageal cancer, oral squamous cell carcinoma or gastric cancer, and the like.
In this context, the term "treatment" is intended to mean obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic in terms of completely or partially preventing the disease or symptoms thereof, and/or may be therapeutic in terms of partially or completely curing the disease and/or adverse effects caused by the disease. As used herein, "treating" encompasses diseases in mammals, particularly humans, including: (a) Preventing the occurrence of a disease or disorder in an individual susceptible to the disease but not yet diagnosed with the disease; (b) inhibiting disease, e.g., arresting disease progression; or (c) alleviating a disease, e.g., alleviating symptoms associated with a disease. As used herein, "treating" or "treatment" encompasses any administration of a drug or compound to an individual to treat, cure, alleviate, ameliorate, reduce or inhibit a disease in the individual, including, but not limited to, administration of a drug comprising a compound described herein to an individual in need thereof.
In this context, the term "antineoplastic agent" includes agents that reduce the volume of solid tumors, inhibit proliferation of tumor cells, alleviate clinical symptoms of tumors, inhibit recurrence of tumors, and the like.
Herein, "amphiphilic calixarene" refers to calixarene having hydrophilicity and lipophilicity. According to one embodiment of the present invention, there is provided a pharmaceutical composition comprising a drug to be delivered and a delivery vehicle,
wherein the drug to be delivered comprises a STING agonist,
the delivery vehicle comprises a hydrated vesicle comprising a compound (I) or a pharmaceutically acceptable salt thereof,
wherein n=4-20;
R 1 selected from C 4 -C 20 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
R 2 selected from amino groups,
According to a specific embodiment of the present invention, the delivery vehicle contained in the pharmaceutical composition may be in the form of a hydrated vesicle, such as calixarene vesicle 12C-NV of FIG. 1, containing a significant amount of compound (I) or a pharmaceutically acceptable salt thereof, wherein the longer alkyl chain contained in compound (I) (e.g., GC5A-12C of FIG. 1) or a pharmaceutically acceptable salt thereof forms a vesicle structure, which interacts with a STING agonist (e.g., CDN class CDG of FIG. 1) via a host guest SF ) Stable combination, high-efficiency loading of cyclic dinucleotide and rapid release of cyclic dinucleotide in response to tumor tissues and intracellular high-concentration ATP. In vitro and in vivo experiments prove that the calixarene delivery system can effectively improve the transmembrane efficiency of STING agonists, promote enrichment of STING agonists in tumors and tumor-infiltrating lymph nodes, induce an immunogenic tumor microenvironment, and further effectively inhibit tumor growth and improve the survival rate of mice.
According to a specific embodiment of the present invention, the STING agonist contained in the pharmaceutical composition is a CDN compound, and the structural general formula of the STING agonist is shown in fig. 2, wherein B 1 And B 2 Each independently selected from any one of natural base A, T, C, G, U and non-natural base; y is Y 1 And Y 2 Each independently selected from any of-OH and-SHOne of the two; x is X 1 And X 2 Each independently selected from any one of-H, -OH and-F. All CDN compounds can be loaded through the hydration vesicles so as to improve the stability and transmembrane efficiency of STING agonists.
According to a specific embodiment of the present invention, the compound (i) or a pharmaceutically acceptable salt thereof, which is not in the form of a hydrated vesicle, is capable of loading CDN compounds in the form of free single molecules, thereby improving stability and transmembrane efficiency of STING agonists, and the method is also within the scope of the present invention.
The aspects of the present disclosure will be explained below with reference to examples. Those skilled in the art will appreciate that the following examples are illustrative of the present disclosure and should not be construed as limiting the scope of the present disclosure. The examples are not to be construed as limiting the specific techniques or conditions described in the literature in this field or as per the specifications of the product. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
EXAMPLE 1CDG SF Preparation of the @12C delivery System
1. Preparation of CDG SF Amphiphilic calixarenes GC5A-12C (upper guanidine-modified, lower dodecane-modified pentabenzene calixarene) and polyethylene glycol PEG-12C of the @12C delivery system were synthesized according to routes known in the art. First, GC5A-12C and PEG-12C were dissolved in a 1/1 (volume ratio) of a mixed solution of methanol and chloroform at a molar ratio of 1/1. The solvent was then pumped out with an oil pump and the remainder was subjected to HEPES (10 mM, pH=7.4) buffer and sonicated at 80℃for 4h to complete the preparation of hydrated vesicle 12C-NV. Follow-up CDG taking SF (see FIG. 1 for structure) and 12C-NV solution were mixed in HEPES buffer (molar ratio 1:2) to complete loading. Obtaining a CDG-loaded product SF CDG of (2) SF @12C delivery system.
2、CDG SF Particle size characterization of @12C
GC5A-12C vesicles and CDG using Transmission Electron Microscopy (TEM) and Dynamic Light Scattering (DLS), respectively SF Particle size characterization of @12C (fig. 3). FIG. 3 shows that the particle size of GC5A-12C vesicles is 25.2nm and CDG SF Particle size @12C25.4nm. These characterizations illustrate the load CDG SF The GC5A-12C vesicle morphology is not obviously affected after the process.
EXAMPLE 2CDG SF Evaluation of activation of @12C antigen presenting cells
CDG using RAW264.7 (macrophage) and BMDC (bone marrow derived dendritic cell), respectively SF Evaluation of stimulation Activity at 12C.
The two cells were obtained separately and cultured in 24-well plates at 50 ten thousand/well, for 12 hours. The culture medium used in RAW264.7 cell stimulation experiment is Opti-MEM and CDG SF Active load at @12C (acquisition method same as example 1), stimulus concentration CDG SF 3. Mu.M. GC5A-12C vesicles were separated into two concentrations of 3. Mu.M and 6. Mu.M, 500. Mu.L per well volume. Stimulation time 18h, followed by digestion of cells with pancreatin for anti-CD86-PE (1/200 dilution) antibody staining and flow analysis (BD Calibur).
The culture medium used in BMDC stimulation experiment is Opti-MEM and CDG SF Active @12C group present load (acquisition method same as example 1), stimulus concentration CDG SF 0.5. Mu.M, 2. Mu.M for GC5A-12C vesicles, 500. Mu.L per well volume. Stimulation time 24h, after which cells were collected by direct centrifugation and antibody staining on ice (anti-CD 11c, anti-CD86 and anti-CD40, biolegend,1/200 dilution). Cells were washed and analyzed on a BD LSRFortessa instrument. The results indicated CDG SF @12C is effective in promoting CDG SF Transmembrane, and activates both antigen presenting cells (fig. 4).
EXAMPLE 3CDG SF Evaluation of antitumor Effect of @12C
Melanoma cell line B16-F10 cell culture: RPMI-1640+10% FBS+1% double antibody (penicillin and streptomycin), 5% CO 2 37 ℃. After the completion of the culture, cells were digested with pancreatin, resuspended in PBS buffer or serum-free RPMI-1640 medium, and finally injected subcutaneously on the right back with 15 ten thousand or 20 ten thousand B16-F10 cells (melanoma cells) per mouse (C57 BL/6, female). Groups of mice with uniform tumor volumes (5-10 per group) were picked from them by day 9 for subcutaneous and intratumoral injection treatment, once a day apart for four times. Injection CDG SF Triethylamine salt 8 μg/or 20 μg/,CDG SF GC5A-12C molar ratio = 1:2. Tumor volume calculation formula: length x width x 0.5cm 3 . Mice were euthanized when tumor growth diameter reached 15 mm. The anti-PD-1 antibody in the monoclonal antibody combination experiment is injected into the abdominal cavity with the dosage of 200 mug/patient.
FIG. 5 shows the experimental results of CDG SF The @12C system significantly increased CDG both intratumorally (fig. 5) and subcutaneously (fig. 6) by the two routes of administration SF The anti-tumor effect of the anti-tumor agent is used for inhibiting tumor growth and improving survival rate, and the anti-tumor effect of anti-PD-1 can be effectively improved by combining the anti-tumor agent with an anti-PD-1 antibody of immune checkpoint blocking therapy, and immune memory is induced to be generated at the same time, so that tumor recurrence is inhibited (figures 7 and 8).
EXAMPLE 4 tumor infiltrating lymphocyte assay
Tumor-bearing mice were obtained in the same manner as in example 3 until tumors of the mice grew to about 200mm 3 Intratumoral administration (CDG) SF Triethylamine salt 8 μg/dose), 1 injection at 1 day intervals, 2 total injections. At 24h after the second injection, mice were treated and tumors, tumor-infiltrating lymph nodes and spleen tissue were collected. The tumor tissue was removed by grinding with a syringe plunger, then filtering with a 70 μm cell strainer, adding into a siliconizing tube, centrifuging at 1700rpm for 5min, and sucking off the supernatant. Percoll cell isolate (40%) diluted with PBS buffer was added, vortexed, centrifuged at 2800rpm for 15min and the supernatant was slowly aspirated. Then, the cells were collected by incubating with RBC lysate (1 mL) for 5min, terminating with 4mL of PBS, and centrifuging at 1700rpm for 5 min. After the cell count, a portion of the cells were plated onto a 24-well plate, and T cell stimulating reagent Cell Activation Cocktail with Brefeldin A was added to the plate and incubated in an incubator for 4 hours. Antibody staining (anti-CD 45, anti-NK1.1, anti-CD3, anti-CD4, anti-CD8, anti-Foxp3 and anti-IFN-gamma) is carried out on the cells after stimulation and the unstimulated cells respectively, wherein the IFN-gamma belongs to intracellular cytokines, and the anti-IFN-gamma is stained after surface antibodies are stained first and then a penetrating fixing solution is used for perforation.
FIG. 9 flow chart shows CDG SF Injection of @12C can induce an immunogenic tumor microenvironment, promote T cell anti-tumor activity, and reduce the proportion of immunosuppressive cell tregs, fromThereby producing a high-efficiency tumor inhibition effect.
In the description of the present specification, the descriptions of the terms "one embodiment," "some embodiments," "examples," "particular examples," "some embodiments," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present invention. In this specification, schematic representations of the above terms are not necessarily directed to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, the different embodiments or examples described in this specification and the features of the different embodiments or examples may be combined and combined by those skilled in the art without contradiction.
While embodiments of the present invention have been shown and described above, it will be understood that the above embodiments are illustrative and not to be construed as limiting the invention, and that variations, modifications, alternatives and variations may be made to the above embodiments by one of ordinary skill in the art within the scope of the invention.

Claims (14)

1. A pharmaceutical composition comprising:
the drug to be delivered and the delivery vehicle,
wherein the drug to be delivered comprises a STING agonist,
the delivery vehicle comprises a hydrated vesicle comprising a compound (I) or a pharmaceutically acceptable salt thereof,
wherein n=4-20;
R 1 selected from C 4 -C 20 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
R 2 selected from amino groups,
2. The pharmaceutical composition according to claim 1, wherein n = 4-10 in compound (i);
optionally, n=4-8;
optionally R 1 Selected from C 4 -C 15 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
optionally, the halogen is selected from fluorine, chlorine, bromine, iodine.
3. The pharmaceutical composition of claim 1, wherein the STING agonist comprises a compound selected from the CDN class.
4. The pharmaceutical composition of claim 1, wherein the CDN compound has the structural formula:
wherein B is 1 And B 2 Each independently selected from any one of natural base A, T, C, G, U and non-natural base;
Y 1 and Y 2 Each independently selected from any one of-OH and-SH;
X 1 and X 2 Each independently selected from any one of-H, -OH and-F.
5. The pharmaceutical composition of claim 4, wherein the non-natural base is selected from an artificial base or a modified natural base;
optionally, the modified natural base is I or mC.
6. The pharmaceutical composition according to claim 4, wherein the molar ratio of CDN compound to compound (i) in the pharmaceutical composition is 1:1 to 1:10;
optionally, the molar ratio of the CDN compound to the compound (I) is 1:1-1:2.
7. The pharmaceutical composition of claim 4, further comprising a stabilizer for extending the half-life of the drug,
optionally, the stabilizer comprises a stabilizer selected from the group consisting of C 4 -C 20 A PEG linked to a linear or branched alkyl group, said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
optionally, the halogen is selected from fluorine, chlorine, bromine, iodine;
optionally, the degree of polymerization of the PEG is 1000-5000;
optionally, in the pharmaceutical composition, the compound (I) or a pharmaceutically acceptable salt thereof and the C 4 -C 20 The molar ratio of the linear or branched alkyl-linked PEG is 1 (0.02-10), preferably 1: (0.1-1).
8. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition further comprises an immune checkpoint inhibitor;
optionally, the immune checkpoint inhibitor comprises at least one selected from anti-PD-1, anti-PD-L1, anti-CTLA-4, anti-LAG-3, anti-TIM-3, anti-TIGIT.
9. The pharmaceutical composition of any one of claims 1-8, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.
10. A pharmaceutical formulation comprising a pharmaceutical composition according to any one of claims 1 to 9.
11. The use of a delivery vehicle for the delivery of a STING agonist, characterised in that the delivery vehicle comprises a hydrated vesicle comprising compound (i) or a pharmaceutically acceptable salt thereof,
wherein n=4-20;
R 1 selected from C 4 -C 20 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
R 2 selected from amino groups,
12. Use according to claim 11, characterized in that in compound (i) n = 4-10;
optionally, n=4-8;
optionally R 1 Selected from C 4 -C 15 A linear or branched alkyl group of (2), said linear or branched alkyl group being unsubstituted or substituted with at least one halogen;
optionally, the halogen is selected from fluorine, chlorine, bromine, iodine;
optionally, the STING agonist comprises a compound selected from CDN class;
optionally, the structural general formula of the CDN compound is as follows:
wherein B is 1 And B 2 Each independently selected from any one of natural base A, T, C, G, U and non-natural base;
Y 1 and Y 2 Each independently selected from any one of-OH and-SH;
X 1 and X 2 Each independently selected from any one of-H, -OH, and-F;
optionally, the non-natural base is selected from an artificial base or a modified natural base;
optionally, the modified natural base is I or mC;
optionally, in the pharmaceutical composition, the molar ratio of the CDN compound to the compound (i) is 1:1 to 1:10;
optionally, the molar ratio of the CDN compound to the compound (I) is 1:1-1:2.
13. Use of a pharmaceutical composition according to any one of claims 1-9 for the preparation of an antitumor drug.
14. The use according to claim 13, wherein the tumour comprises a member selected from melanoma, colon cancer, breast cancer, lymphoma, lung cancer.
CN202210432161.2A 2022-04-22 2022-04-22 Novel STING agonist delivery system and immunotherapeutic application Pending CN116966147A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210432161.2A CN116966147A (en) 2022-04-22 2022-04-22 Novel STING agonist delivery system and immunotherapeutic application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210432161.2A CN116966147A (en) 2022-04-22 2022-04-22 Novel STING agonist delivery system and immunotherapeutic application

Publications (1)

Publication Number Publication Date
CN116966147A true CN116966147A (en) 2023-10-31

Family

ID=88475387

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210432161.2A Pending CN116966147A (en) 2022-04-22 2022-04-22 Novel STING agonist delivery system and immunotherapeutic application

Country Status (1)

Country Link
CN (1) CN116966147A (en)

Similar Documents

Publication Publication Date Title
Siriwon et al. CAR-T cells surface-engineered with drug-encapsulated nanoparticles can ameliorate intratumoral T-cell hypofunction
Martins et al. Vaccine adjuvant uses of poly-IC and derivatives
TW202021982A (en) T cell receptor constructs and uses thereof
KR101323540B1 (en) Therapeutic agent for cancer
Zhou et al. Overcoming immune resistance by sequential prodrug nanovesicles for promoting chemoimmunotherapy of cancer
Wang et al. Toll-like receptor 3 agonist complexed with cationic liposome augments vaccine-elicited antitumor immunity by enhancing TLR3–IRF3 signaling and type I interferons in dendritic cells
Lu et al. Exploiting in situ antigen generation and immune modulation to enhance chemotherapy response in advanced melanoma: a combination nanomedicine approach
MXPA04004998A (en) Methods for treating cancer.
WO2018021400A1 (en) Immune adjuvant for cancer
Pi et al. Exosomes: powerful weapon for cancer nano-immunoengineering
Dai et al. Nanomedicines modulating myeloid-derived suppressor cells for improving cancer immunotherapy
US20230080443A1 (en) Immunogenic compounds for cancer therapy
JP2020520368A (en) Novel pharmaceutical composition containing particles containing complex of double-stranded polyribonucleotide and polyalkyleneimine
Mohammadian Haftcheshmeh et al. Immunoliposomes bearing lymphocyte activation gene 3 fusion protein and P5 peptide: A novel vaccine for breast cancer
JP2023036999A (en) Oxabicycloheptanes for modulating immune response
WO2016119308A1 (en) Antitumour preparation and preparation method thereof
Chakraborty et al. Application of toll-like receptors (TLRs) and their agonists in cancer vaccines and immunotherapy
Zhao et al. Targeting STING in cancer: Challenges and emerging opportunities
RU2709015C2 (en) Medicament
Yan et al. Nano-adjuvants and immune agonists promote antitumor immunity of peptide amphiphiles
Najar et al. Topical CpG enhances the response of murine malignant melanoma to dacarbazine
CN116966147A (en) Novel STING agonist delivery system and immunotherapeutic application
Cascinelli et al. Evaluation of clinical efficacy and tolerability of intravenous high dose thymopentin in advanced melanoma patients
Tseng et al. Induction of immune responses and phosphatidylserine exposure by TLR9 activation results in a cooperative antitumor effect with a phosphatidylserine-targeting prodrug
US20210379106A1 (en) Oxabicycloheptanes for enhancing car t cell function

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination