CN117015383A - Multifunctional cyclic dinucleotide and application thereof - Google Patents

Multifunctional cyclic dinucleotide and application thereof Download PDF

Info

Publication number
CN117015383A
CN117015383A CN202180071565.7A CN202180071565A CN117015383A CN 117015383 A CN117015383 A CN 117015383A CN 202180071565 A CN202180071565 A CN 202180071565A CN 117015383 A CN117015383 A CN 117015383A
Authority
CN
China
Prior art keywords
formula
compound
compounds
cancer
compound according
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
CN202180071565.7A
Other languages
Chinese (zh)
Inventor
张彦涛
屈粒
楼良
郑普吉
吕飞
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.)
Taili Biotechnology Shanghai Co ltd
Original Assignee
Taili Biotechnology Shanghai Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Taili Biotechnology Shanghai Co ltd filed Critical Taili Biotechnology Shanghai Co ltd
Priority claimed from PCT/CN2021/124704 external-priority patent/WO2022083584A1/en
Publication of CN117015383A publication Critical patent/CN117015383A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/213Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids containing cyclic phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Abstract

A multifunctional cyclic dinucleotide compound of formula (X) and derivatives thereof, which are useful as prodrugs of apoptosis inducers or cytotoxic agents for inducing apoptosis or antiviral, and for modulating immune pathways to produce therapeutically beneficial immune responses. Pharmaceutical compositions and pharmaceutical combinations of cyclic dinucleotide compounds, methods for the synthesis thereof, and medical uses thereof.

Description

Multifunctional cyclic dinucleotide and application thereof Technical Field
The present invention relates to multifunctional cyclic dinucleotide compounds and derivatives thereof, the functions of which include use as prodrugs of apoptosis inducers or cytotoxic agents for inducing apoptosis or antiviral in tumor cells, and as immunomodulators for modulating immune pathways to generate therapeutically beneficial immune responses, in particular for activating STING-mediated immune pathways. The present disclosure further relates to pharmaceutical compositions and pharmaceutical combinations comprising the cyclic dinucleotide compounds of the invention, methods for the synthesis thereof, and medical uses thereof.
Background
Cancer is a malignant disease characterized by abnormal cell differentiation and proliferation, loss of control of growth, infiltration and metastasis, has become one of the important causes of death in humans, and the incidence is still continuously growing worldwide; at the same time, viral infections have also led to death in millions of humans worldwide.
Antimetabolite nucleoside analogues are one of the main therapeutic approaches in anticancer/antitumor and antiviral infection treatment strategies. Specifically, antimetabolite nucleoside drugs themselves cannot directly act, must be converted into a triphosphorylated form in vivo through various cell kinases, become active substrates of polymerase as pseudo metabolites, are inserted into DNA or RNA through nucleic acid biosynthesis pathways, inhibit modification and extension of DNA or RNA, or inhibit reverse transcriptase related to DNA or RNA synthesis, thereby inducing apoptosis of tumor cells or preventing replication of viruses, exhibit cytotoxicity, and can be used for the treatment of cancer/tumor or viral infection; meanwhile, cell debris generated by cytotoxicity of the nucleoside drugs can cause immune response of host cells, so that growth and reproduction of tumor cells or viruses are further inhibited.
The anticancer nucleosides currently used clinically include the following:
however, the nucleoside medicine has a plurality of active groups such as hydroxyl, amino and the like on the molecular structure, which results in low film permeation efficiency, poor stability and poor drug substitution property, and special or frequent drug administration is often required, thus bringing a plurality of inconveniences to patients. Therefore, the search for new nucleoside drug improvements has been an active area of drug development.
Endogenous Cyclic Dinucleotide (CDN) cGAMP is an important element of the cGAS-STING (cyclic GMP-AMP synthase-interferon gene stimulator) signaling pathway, an important component of the innate immune system. Specifically, cGAS interacts with DNA from tumor cells, dying cells, viruses, bacteria, or mitochondria, catalyzing ATP and GTP synthesis of Cyclic Dinucleotide (CDN) cGAMP. Endogenous cGAMP produced further binds STING on the endoplasmic reticulum membrane (ER), STING bound to cGAMP is activated, undergoes conformational changes, translocates to Golgi, induces activation of key transcription factors IRF-3 and NF- κb, which enter the nucleus, induces expression of type I interferons and pro-inflammatory cytokines such as IL-6, TNF- α and IFN- γ (Jiang et al, cGAS-STING, an important pathway in cancer immunotherapy, journal of Hematology & Oncology,2020,13:81;Xiangling Cui et al, STING modulators: predictive significance in drug discovery, european Journal of Medicinal Chemistry 182 (2019) 111591).
It is well known in the art that type I interferons not only exhibit antiviral activity, but also directly inhibit proliferation of human tumor cells, significantly enhance anti-tumor immune responses by inducing adaptation and activation of innate immune cells, and inhibit tumor invasion by modulating enzyme expression associated with tissue remodeling, and thus are useful as anticancer agents.
Given that endogenous Cyclic Dinucleotide (CDN) cGAMP is a key mediator of the innate immune system described above in response to viruses and tumors, ultimately contributing to interferon or pro-inflammatory cytokine production and thereby achieving therapeutic benefit, a range of CDN STING agonists have been synthesized and validated for laboratory activity, examples of which are described, for example, in WO2014/189805, WO2017/027645 and WO 2018/060323. However, existing treatments based on CDN still lack sufficient clinical therapeutic efficacy, and thus there is still a need for improved CDN-based STING agonists to provide a safer and more potent antiviral or antitumor effect.
According to the consensus in the art, STING pathway can be activated by exogenous DNA (tumor or virus, etc.). If no mediation of protein neoantigen exists, the downstream pro-inflammatory factors driven by the STING channel through interferon and the like lack targeting, so that the self-immune response tolerance is poor and the treatment window is narrow. The first generation of single STING agonists in combination with PD-1 antibodies still focused on the activation of systemic immunity, and were not able to address the problem of adaptive immunity selectivity and evoke responses in tumor microenvironments.
On the one hand, the compound of the invention is used as a known high-activity STING agonist, activates a signal channel, releases interferon and other inflammatory factors and activates an immune system; secondly, its cytotoxic function is then activated, selectively killing the tumor, releasing a large amount of tumor neoantigen and tumor DNA to establish the recognition function of adaptive immunity, training the immune system with a certain vector; again, tumor neoantigens and tumor DNA continue to activate STING pathways and other immune systems, killing tumor cells; finally, the released tumor neoantigen is recognized by DC cells, and interacts with T cells to form immune memory, thereby achieving long-term control of remote tumor and cancer cell migration.
In particular, in the process of searching for new cytotoxic nucleoside drugs with improved properties, the application introduces the nucleoside drugs with cytotoxic effects into the molecular structure of CDN as a building unit, namely, introduces hidden cytotoxic pharmacophores at the molecular level of CDN. The novel CDN drug molecules formed by the method can activate STING to induce I-type interferon to generate, so that antiviral or antitumor immunotherapy effect is realized. More innovative, the product formed by decomposing the molecule in vivo, namely the nucleoside cytotoxic drug, can specifically interfere with the metabolism of nucleic acid, prevent cell division and reproduction, lead to death of tumor cells or prevent virus replication, release tumor DNA to continuously activate STING and release tumor neoantigen to establish the recognition function of adaptive immunity, train the vector of immune system, overcome the defects of low efficiency, poor stability, poor drug generation property and frequent need of special or frequent administration of the independent nucleoside drug, and simultaneously overcome the source problem of exogenous DNA (such as tumor DNA) needed to continuously activate STING channels. More importantly, the CDN structure skeleton of the molecule disclosed by the application can activate STING, so that the generation of I-type interferon is induced, and further, the antiviral or antitumor immunotherapy effect is realized. On the other hand, the hidden cytotoxicity pharmacophore in the novel CDN molecule can timely release and generate apoptosis fragments, provide antigens aiming at tumors or viruses for an immune system, generate antibody-antigen response under the cooperation of immune leukocyte subpopulations, and further provide the capability of 'immune memory' or lasting immunity aiming at the antigens.
Therefore, the invention provides a novel CDN compound, and through the combination of drugs at the sub-molecular level, the antimetabolite treatment and immunosuppression of virus infection or tumor are realized at the same time, and compared with single cytotoxic drugs or simple CDN STING agonists, the novel CDN compound can provide enhanced and even synergistic effects.
It should be noted that the above discussion of the background to the invention is provided merely to aid the reader in understanding the invention and is not an admission that the prior art describes or constitutes the invention.
Summary of The Invention
The object of the present invention is to provide a novel group of antiviral or antitumor compounds based on cyclic dinucleotide structures.
In one aspect, the invention provides a set of cyclic dinucleotide compounds having the general formula, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof,
for example
Wherein X is 1 And X 2 Each independently selected from-OH and-SH; and has a cytotoxic or antiviral effect on at least one of the two nucleosides of the two nucleotides that form the loop. On the one hand, the compound with the structure generates independent nucleoside cytotoxicity or antiviral compounds after in vivo decomposition, and after activation by intracellular glycoside kinase triphosphates, the cell division and propagation are prevented by specifically interfering with the metabolism of nucleic acid, so that the proliferation of tumor cells or the replication of viruses are inhibited. On the other hand, the molecule of the invention keeps the immune activation function of the cyclic dinucleotide, namely activates target site STING and finally induces the generation of I-type interferon through STING signal transduction cascade, thereby generating tumor immune activity, inhibiting the growth and the metastasis of tumor or exerting antiviral activity; furthermore, apoptotic tissue fragments provide the immune system with antigens that are not naturally expressed in the host, generating antigen-antibody responses, thereby providing the ability to "memory" or persist immunity against the antigen encountered.
In particular, the present invention provides in this aspect a cyclic dinucleotide compound of formula (Y),
wherein X is 1 、X 2 、B 1 、B 2 、R 1 、R 1 ’、R 2 、R 2 ' as defined herein; stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts, prodrugs or solvates thereof.
More specifically, the present invention provides in this aspect cyclic dinucleotide compounds of formulae (I), (II), (III) and (IV) and their respective sub-formulae; more specifically, the present invention provides in this aspect cyclic dinucleotide compounds of the respective sub-formulae of formulae (I), (II), (III) and (IV); and their respective specific embodiments, as described herein below.
In another aspect, the present invention provides a process for preparing the compounds of the invention described herein, and also provides the compounds of the invention described herein obtainable by the process described herein.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention described herein, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt, prodrug or solvate thereof, and one or more pharmaceutically acceptable excipients.
In a further aspect, the invention provides a compound of the invention as described herein or a pharmaceutical composition as described herein for use as an active agent for the treatment or prophylaxis of a disease associated with or mediated by an immune response, in particular for the treatment or prophylaxis of a disease associated with or mediated by STING, more in particular for the treatment or prophylaxis of an inflammatory, allergic or autoimmune disease, infectious disease or cancer, in particular for use as an antiviral or antitumor agent, or as a vaccine adjuvant.
In another aspect, the invention provides a compound of the invention described herein or a pharmaceutical composition described herein for use as a cytotoxic agent, particularly an antineoplastic agent, for the treatment or prophylaxis of hyperproliferative diseases.
In another aspect, the invention provides a compound of the invention described herein or a pharmaceutical composition described herein for use as a cytotoxic agent for the treatment or prophylaxis of a viral infection.
In another aspect, the invention provides the use of a compound of the invention described herein or a pharmaceutical composition described herein for the treatment or prophylaxis of a disease associated with or mediated by an immune response, for example as a STING agonist, in particular for the treatment or prophylaxis of a disease associated with or mediated by STING, more in particular for the treatment or prophylaxis of an inflammatory, allergic or autoimmune disease, infectious disease or cancer, in particular a tumour or viral infection; or as a vaccine adjuvant.
In another aspect, the invention provides the use of a compound of the invention described herein or a pharmaceutical composition described herein as a cytotoxic agent in the treatment or prophylaxis of hyperproliferative diseases, in particular tumours; or as a cytotoxic agent in the treatment or prevention of viral infections.
In another aspect, the invention provides a method of treating or preventing a disease associated with or mediated by an immune response, particularly STING, more particularly an inflammatory, allergic or autoimmune disease, an infectious disease or cancer, particularly a tumor or viral infection, in a subject, the method comprising administering to a human or animal a compound of the invention described herein or a pharmaceutical composition described herein.
In another aspect, the invention provides a method of treating or preventing a hyperproliferative disease, particularly a tumor, in a subject comprising administering to a human or animal a compound of the invention described herein or a pharmaceutical composition described herein.
In another aspect, the invention provides a method of treating or preventing a viral infection in a subject, the method comprising administering to a human or animal a compound of the invention described herein or a pharmaceutical composition described herein.
In another aspect, the invention provides the use of a compound of the invention described herein or a pharmaceutical composition described herein in the manufacture of a medicament for the treatment or prophylaxis of a disease associated with or mediated by an immune response, particularly a disease associated with or mediated by STING, more particularly an inflammatory, allergic or autoimmune disease, an infectious disease or cancer, particularly a tumour or viral infection, or as a vaccine adjuvant.
In another aspect, the invention provides the use of a compound of the invention described herein or a pharmaceutical composition described herein in the manufacture of a medicament for the treatment or prophylaxis of hyperproliferative diseases, in particular tumours.
In another aspect, the invention provides the use of a compound of the invention described herein or a pharmaceutical composition described herein in the manufacture of a medicament for the treatment or prophylaxis of a viral infection.
In another aspect, the invention provides a compound of the invention described herein or a pharmaceutical composition described herein for use as a multifunctional agent having both immunotherapeutic and cytotoxic therapeutic activities, including the ability to activate the immune system to function against tumor and antiviral replication by agonizing the STING signaling pathway, to cause tumor cell death or prevent viral replication by release of a cytotoxic agent, followed by sustained activation of STING by release of tumor DNA to kill tumor cells, and by release of tumor neoantigens to generate an antibody-antigen response to provide "immune memory" or durable immunity to tumors. In this regard, the invention also provides the use of a compound of the invention as described herein or a pharmaceutical composition as described herein for achieving the various functions described above, for example, in particular for the treatment or prophylaxis of a viral infection or tumor; a method of treating or preventing a disease associated with or mediated by an immune response, particularly STING, more particularly an inflammatory, allergic or autoimmune disease, an infectious disease or cancer, particularly a tumor or viral infection in a subject by the various functions described herein, comprising administering to a human or animal a compound of the invention described herein or a pharmaceutical composition described herein; and the use of a compound of the invention described herein or a pharmaceutical composition described herein in the manufacture of a medicament for performing the various functions described above, for example, in particular, for the treatment or prophylaxis of a viral infection or tumour.
In another aspect, the invention provides a pharmaceutical combination comprising a compound of the invention, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, and at least one other therapeutic agent,
in another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention described herein, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, at least one other therapeutic agent, and one or more pharmaceutically acceptable excipients.
In another aspect, the invention provides a pharmaceutical combination comprising a compound of the invention and at least one other therapeutic agent as described herein for use in the treatment or prophylaxis of hyperproliferative diseases, viral infections or diseases associated with or mediated by STING, more particularly inflammatory, allergic or autoimmune diseases, infectious diseases or cancers, in particular tumours or viral infections.
In a further aspect, the invention provides the use of a pharmaceutical combination comprising a compound of the invention and at least one other therapeutic agent as described herein for the treatment or prophylaxis of hyperproliferative diseases, viral infections or diseases associated with or mediated by STING, more particularly inflammatory, allergic or autoimmune diseases, infectious diseases or cancers, in particular tumours or viral infections.
In another aspect, the invention provides a method of treating or preventing a hyperproliferative disease, a viral infection or a disease associated with or mediated by STING, more particularly an inflammatory, allergic or autoimmune disease, an infectious disease or cancer, especially a tumor or viral infection, in a subject comprising administering to a human or animal a pharmaceutical combination comprising a compound of the invention and at least one other therapeutic agent as described herein.
Brief description of the drawings
FIG. 1 shows the interferon stimulating activity of representative compounds of the invention in THP-1 cells.
FIG. 2 shows the tumor growth inhibiting activity of representative compounds of the present invention in a mouse transplanted CT26 colon cancer model. 2A: tumor volume change of treated tumors; 2B: tumor volume change of untreated tumors; 2C: body weight change in mice; 2D: tumor volume change in immunized/non-immunized mice.
FIG. 3 shows the immunological memory of representative compounds of the present invention in mice with/without immunization. 3A: immune memory in immunocompetent mice; 3B: immune memory in non-immunized mice.
FIG. 4 shows the hepatocyte metabolic properties of representative compounds of the invention. 4A: stability of hepatocyte metabolism; 4B: and (5) identifying metabolites of the liver cells.
Disclosure of Invention
Definition of the definition
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this application belongs.
The nomenclature used in the present application is based on IUPAC system nomenclature, unless otherwise indicated. IUPAC chemical name was generated using OpenEye lexichem version 1.2.0, perkinElmer E-notebook for Chemistry or Insight for Excel 2017 R2.
Unless otherwise indicated, any open valency appearing on a carbon, oxygen, sulfur, or nitrogen atom in a structure herein indicates the presence of a hydrogen atom.
The term "immune system" has its ordinary meaning as understood by those skilled in the art and means the whole or any one or more components of molecules, substances (e.g., body fluids), anatomical structures (e.g., cells, tissues, or organs), and physiological processes that are involved in preventing infection in the body, protecting the body during infection or disease, and/or helping the body to recover health after infection or disease.
The term "disease associated with or mediated by an immune response" means a disease associated with or mediated by a defensive response of the immune system of the body against an abnormal component or a mutated autologous component. For the purposes of the present application, a "disorder associated with or mediated by an immune response" particularly refers to a condition of a human or animal in which the function of the immune system is impaired, inactivated or otherwise compromised, or one or more immune components are impaired, inactivated or otherwise compromised, or a disease state caused by such a condition, particularly a disorder in which induction of an immune response via the STING pathway may be ameliorated.
The term "STING" is an abbreviation for interferon gene stimulator (stimulator of interferon genes). STING is a transmembrane protein receptor in humans, and activation of STING by Cyclic Dinucleotides (CDNs) results in activation of IRF3 and NF- κb pathways, thus leading to induction of type I interferons and pro-inflammatory cytokines, respectively. The term "STING agonist" refers to any substance that activates STING to elicit a physiological response in vitro or in vivo.
The term "disease associated with or mediated by STING" means a disease in which induction of an immune response by STING pathway can be ameliorated, i.e., activation of STING will reduce the incidence of disease, reduce or eliminate disease conditions, including but not limited to inflammation, allergic or autoimmune diseases, infectious diseases or cancers, etc. For the purposes of the present invention, the "disease associated with or mediated by STING" is preferably selected from tumours or cancers.
The term "hyperproliferative disease," "tumor," or "cancer" refers to a physiological condition in a subject characterized by uncontrolled or deregulated cell growth or death, including solid tumors and hematological tumors, whether malignant or benign, including but not limited to brain, skin, bladder, ovary, breast, stomach, pancreas, prostate, colon, blood, lung, and bone cancers. Examples of the above-mentioned types of cancers include neuroblastoma, intestinal cancer such as rectal cancer, colon cancer, familial adenomatous polyposis cancer and hereditary nonlymphoid colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, peritoneal cancer, soft tissue sarcoma, urothelial cancer, sweat gland cancer, gastric cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, endometrial cancer, pancreatic cancer, prostate cancer, testicular cancer, breast cancer (including HER2 negative breast cancer), urinary cancer, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, neuroblastoma and peripheral nerve ectodermal tumor, hodgkin's lymphoma, non-hodgkin's lymphoma, burkitt's lymphoma, acute Lymphoblastic Leukemia (ALL), chronic Lymphocytic Leukemia (CLL), chronic myelogenous leukemia (CLL) and lymphocytic carcinoma, acute Myelogenous Leukemia (AML), myelogenous leukemia (chronic myelogenous leukemia (CML), adult T-cell lymphoma, diffuse lymphoma (DLBCL), hepatoma, multiple myeloma, seminoma, osteosarcoma, chondrosarcoma, anal canal carcinoma, adrenocortical carcinoma, chordoma, fallopian tube carcinoma, gastrointestinal stromal tumor, myeloproliferative disorders, mesothelioma, biliary tract carcinoma, ewing's sarcoma and other rare tumor types.
The term "therapeutic agent" refers to one or more substances administered to a human or animal to achieve a therapeutic effect, including substances that prevent, cure or ameliorate the effects of a disease, improve health. The therapeutic agents of the present invention include not only the provided CDN compounds themselves, but also therapeutic agents that may be used in combination with the provided CDN compounds, including but not limited to chemotherapeutic agents, immune agents (particularly immune tumor agents), vaccines, adjuvants, and radiation therapies.
The term "chemotherapeutic agent" refers to one or more chemicals that are administered to a human or animal to kill a tumor, or to slow or prevent the growth of a tumor, and/or to slow or prevent the division of cancer cells, and/or to prevent or slow metastasis.
The term "immunizing agent" refers to any endogenous or exogenous substance that can interact with any one or more components of the immune system, including antibodies, antigens, vaccines and components thereof, nucleic acids, synthetic drugs, natural or synthetic organic compounds, cytokines, natural or modified cells, synthetic analogs thereof, and/or fragments thereof.
The term "immunotherapy" refers to any medical treatment in which one or more components of the immune system of a human or animal are deliberately modulated in order to directly or indirectly obtain a certain therapeutic benefit, including systemic and/or local effects as well as prophylactic and/or therapeutic effects. Immunotherapy may be the administration of one or more immunizing agents to human and animal subjects by any route, e.g., oral, intravenous, dermal, injectable, inhaled, etc., alone or in any combination, whether systemic, local, or a combination of both.
The term "vaccine" refers to a biological agent that is administered to a human or animal to elicit or enhance a specific immune system response and/or protection against one or more antigens in the human or animal.
The term "adjuvant" refers to a secondary therapeutic substance that is administered with a primary therapeutic substance in any order to achieve some complementary, synergistic or other beneficial effect that is not achieved with the primary therapeutic substance alone. Adjuvants may be used with vaccines, chemotherapeutics, or other therapeutic substances that enhance the efficacy of the primary therapeutic substance, reduce the toxic side effects of the primary therapeutic substance, or provide some protection to the subject receiving the primary therapeutic substance, such as, but not limited to, improving the function of the immune system.
The term "cytotoxic agent" or "apoptosis-inducing agent" or similar expression as used herein refers to an active agent for the treatment of abnormal and uncontrolled, progressive growth of cells. For the purposes of the present invention, "cytotoxic agent" refers in particular to nucleoside antimetabolite cytotoxic or antiviral agents including, but not limited to, cytarabine, azacytidine, fluorouridine, deoxyuridine, enocitabine, deoxyfluorouridine, penstin, fludarabine, cladribine, gemcitabine, capecitabine, clofarabine, nelarabine, trifluorothymidine, 8-chloroadenosine, tricitabine, forodesine, 5-fluorodeoxycytidine, ribavirin or acibenzodine.
The term "multifunctional agent" as used herein refers to a compound molecule of the present invention, based on its unique structural design, is capable of performing multiple functions in a subject, having both immunotherapeutic and cytotoxic therapeutic activities, including but not limited to the ability to activate the immune system to perform anti-tumor and antiviral replication by agonizing the STING signaling pathway, to cause tumor cell death or prevent viral replication by releasing a cytotoxic agent, to continuously activate STING to kill tumor cells by releasing tumor DNA, to generate an antibody-antigen response by releasing tumor neoantigen, thereby providing "immune memory" or persistent immunity to tumors.
The term "treatment" of a disease includes inhibiting the disease state, i.e., preventing the development of the disease state or its clinical symptoms, or alleviating the disease state, i.e., causing temporary or permanent regression of the disease state or its clinical symptoms.
The term "prevention" or "prevention" of a disease means that the clinical symptoms of the disease state are not progressed in a subject who is likely to be exposed to or susceptible to the disease state but has not yet experienced or displayed symptoms of the disease state.
The term "therapeutically effective amount" means an amount of a compound or molecule of the invention that (i) treats or prevents a particular disease, condition, or disorder, (ii) reduces, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease being treated, the age and relative health of the subject, the route and form of administration, the judgment of the attending physician or veterinarian, and other factors.
The terms "subject", "individual" or "patient" as used herein refer to a vertebrate. In certain embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, farm animals (e.g., cattle), sports animals, pets (e.g., guinea pigs, cats, dogs, rabbits, and horses), primates, mice, and rats. In a preferred embodiment, the mammal is a human.
The terms "pharmaceutical composition" and "pharmaceutical formulation" (or "formulation") are used interchangeably and refer to a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient and pharmaceutically acceptable excipients to be administered to a mammal (e.g., a human) in need thereof.
The term "pharmaceutical combination" means that the compounds of the present invention may be combined with other active agents for the purpose of the present invention. The other active agent may be one or more additional compounds of the present invention, or may be a second or additional (e.g., third) compound that is compatible with, i.e., does not adversely affect each other, or has complementary activity to, the compounds of the present invention. Such agents are suitably present in combination in an amount effective to achieve the intended purpose. The other active agents may be co-administered with the compounds of the present invention in a single pharmaceutical composition or may be administered separately in separate discrete units from the compounds of the present invention, either simultaneously or sequentially when administered separately.
The term "pharmaceutically acceptable" refers to the property of materials useful in the preparation of pharmaceutical compositions, which are generally safe, non-toxic, and not biologically or otherwise undesirable, and acceptable for veterinary as well as human pharmaceutical use.
The terms "pharmaceutically acceptable excipient", "pharmaceutically acceptable carrier" and "therapeutically inert excipient" are used interchangeably and refer to any pharmaceutically acceptable ingredient in a pharmaceutical composition that is not therapeutically active and is non-toxic to the subject to which it is administered, such as disintegrants, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, carriers, diluents or lubricants used to formulate pharmaceutical products.
The term "pharmaceutically acceptable salt" as used herein means a salt of a compound of the invention which is pharmaceutically acceptable and which has the desired pharmacological activity of the parent compound. In particular, such salts are non-toxic and may be inorganic or organic acid addition salts and base addition salts, including but not limited to: (1) Acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or acid addition salts with organic acids such as acetic acid, propionic acid, caproic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptanoic acid, 3-phenylpropionic acid, trimethylacetic acid, t-butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) a salt formed when an acidic proton present in the parent compound is replaced with a metal ion such as an alkali metal ion, alkaline earth metal ion or aluminum ion, or coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, or the like. Those skilled in the art are aware of the general principles and techniques for preparing pharmaceutically acceptable salts, such as those described in Berge et al, pharm ScL,66,1-19 (1977).
The term "pharmaceutically acceptable prodrugs" as used herein means compounds of the invention which have cleavable groups and become pharmaceutically active in vivo by solvolysis or under physiological conditions, and in particular prodrugs include those compounds which can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, unhydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated or dephosphorylated to give the active compound, including derivatives of the compounds of the invention. Various forms of prodrugs are well known in the art, and suitable prodrug moieties are described, for example, in "Prodrugs and Targeted Delivery", j. Rautico, ed., john Wiley & Sons, 2011.
Prodrugs of the CDN compounds described herein may generally increase the activity, bioavailability, or stability of the compound. In general, alkylation, acylation, or other lipophilic modification of the phosphate moiety or other analogue using a nucleotide will help to increase the stability of the nucleotide.
The term "solvate" as used herein refers to a solvent addition form comprising a stoichiometric or non-stoichiometric solvent, including for example a solvate with water, such as a hydrate, or with an organic solvent, such as methanol, ethanol or acetonitrile, i.e. as a methanolate, ethanolate or acetonitrile, respectively; or in the form of any polymorph. It will be appreciated that such solvates of the compounds of the present invention also include solvates of pharmaceutically acceptable salts of the compounds of the present invention.
The term "isotopic variation" as used herein refers to a compound in which one or more of the atoms constituting the compound are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Can be incorporated into the compounds of the applicationExamples of isotopes on one or more atoms of an species include, for example 2 H、 3 H、 13 C、 14 C、 15 N、 17 O、 18 O、 31 P、 32 P、 35 S and 18 f, thereby forming isotopic variations of the compounds of the present application, whether radioactive or not, are intended to be encompassed within the scope of the present application. In some embodiments, the isotope incorporated is 2H (deuterium); in other embodiments, the isotope incorporated is 3H (tritium).
The term "stereoisomer" as used herein refers to an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., 1, 2, 3, or 4) asymmetric centers, they can produce racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Similarly, the compounds of the application may exist as a mixture of two or more different structural forms (commonly referred to as tautomers) in rapid equilibrium. It is to be understood that the scope of the present application encompasses all such isomers in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%) or mixtures thereof.
The compounds of the invention may have one or more asymmetric centers and may therefore be prepared as (R) -or (S) -stereoisomers or as mixtures thereof, respectively. As used in the structural formulae or structural fragments of the compounds hereinOr (b)Representing the configuration of the asymmetric center, i.e. chiral center. Accordingly, the configuration with respect to the chiral center is denoted by R and S in the nomenclature of the compounds or intermediates provided herein. In the artThe skilled artisan will appreciate that phosphorothioate linkages in the compounds of the present invention are inherently chiral and may each exist in either the R or S configuration, so that it is possible that two phosphorothioate linkages are in the R, R, S, S, S, R and R, S forms. The present invention encompasses compounds of the invention and embodiments thereof in substantially pure form or in the form of mixtures, the compounds containing two phosphorothioate linkages preferably being in substantially pure form as R, R, S, S, S, R and R, S stereoisomers, particularly preferably in substantially pure R, R stereoisomers, i.e. both phosphorus atoms having the R configuration. Their absolute configuration assignment can be carried out according to literature methods (Zhao et al Nucleosides, nucleotides and Nucleic Acids 2009,289,352-378;Knouse et al.Science 2018,361,1234). It is noted that errors in configuration based on literature process errors do not affect the actual configuration of the compounds of the present invention.
The term "substantially pure" as used herein with respect to a CDN refers to a form of a certain stereochemistry at least 75% pure relative to other possible stereochemistry at the chiral center indicated in the above figures. In preferred embodiments, a substantially pure CDN is at least 85% pure, at least 90% pure, at least 95% pure, at least 97% pure, and at least 99% pure. The substantially pure CDN formulation of the present invention is "stereochemically pure" meaning that all CDNs within the formulation have a particular stereochemical configuration at these chiral centers, and is not intended to indicate that all CDNs within the formulation having a particular stereochemical configuration at these chiral centers are otherwise identical. For example, a substantially pure CDN R, R cGAMP phosphorothioate formulation may comprise a combination of R, R c-di-GMP phosphorothioate and R, R c-di-AMP phosphorothioate, and still be a substantially pure cyclic purine dinucleotide formulation.
All the terms "protecting group" herein denote a group that selectively blocks a reactive site in a polyfunctional compound, such that a chemical reaction can proceed selectively at another unprotected reactive site in the meaning typically associated therewith in synthetic chemistry. The protecting group may be removed at an appropriate point in time. Exemplary protecting groups include amino protecting groups including, but not limited to, TBS (t-butyl) Dimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl), benzyl, benzyloxycarbonyl (carbonylbenzyloxy, CBZ), fmoc (9-fluorenylmethoxycarbonyl), p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, t-Butoxycarbonyl (BOC) and trifluoroacetyl; and hydroxy protecting groups including, but not limited to, ester and ether forming groups, particularly tetrahydropyranoxy, bz (benzoyl), i- BuCO (isobutyryl), DMTr (bis (4-methoxyphenyl) benzyl), acetoxy, carbamoyloxy, benzyl and silyl ethers such as TBS (t-butyldimethylsilyl), TBDPS (t-butyldiphenylsilyl). Other examples of these groups are found in T.W.Greene and P.G.M.Wuts, "Greene's protective groups in organic synthesis", 5 th edition. John Wiley&Sons.,Inc.,Hoboken,New Jersey,2014。
The term "deprotection" or "deprotection" refers to the process of removing a protecting group after the selective reaction is complete. The deprotection reagent comprises an acid, a base or hydrogen, in particular potassium carbonate or sodium carbonate, an alcoholic solution of lithium hydroxide, a methanolic solution of zinc, acetic acid, trifluoroacetic acid, a palladium catalyst or boron tribromide.
The term "alkyl" as used herein means a straight or branched chain aliphatic hydrocarbon group having the indicated number of carbon atoms. In particular, the alkyl group may have 1 to 14, 1 to 12, 1 to 10, 1 to 8, 1 to 7, 1 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 carbon atoms. Suitable C 1-14 Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, isohexyl, dimethyl methyl, dipropylmethyl, ethylbutyl methyl, diethyl methyl, methylethylmethyl, ethylpropyl methyl, diethyl ethyl, diethyl propyl, dipropyl ethyl, and the like. Specific alkyl groups have 1 to 7 carbon atoms, for example 1 to 6 carbon atoms, 1 to 4 carbon atoms.
The term "halo" or "halogen" as used herein means fluorine (F), chlorine (Cl), bromine (Br) and iodine (I). A particular halo is fluoro or chloro.
For clarity, the term "guanine" is used herein"can also be expressed asThe phosphorothioate groups used in the structural formulae of the compounds of the present application may be drawn as
Compounds of the application
The terms "compound described herein", "inventive compound", and "compound of the application", and the like, as used throughout the present application, unless otherwise indicated, encompass compounds of formula (X), formula (Y), and specific embodiments of formulae (I), (II), (III), and (IV), individual sub-formula embodiments thereof, and specific or preferred embodiments thereof, stereoisomers, tautomers, racemates, stable isotopic variants, pharmaceutically acceptable salts or solvates, and pharmaceutically acceptable prodrugs thereof, each as described in the definition section above. The compounds of the application may be isolated as mixtures of isomers or as individual isomers which may be prepared synthetically by resolution of racemates by, for example, chromatography or fractional crystallization or from optically active starting materials. Similarly, references herein to "intermediates", whether or not they are themselves claimed, are intended to encompass both their free form as well as each of the derivatives described above, if the context permits.
Preferably, the compounds of the present invention are in free form or a pharmaceutically acceptable salt or solvate thereof; most preferably in free form or a pharmaceutically acceptable salt thereof.
Certain compounds of the invention may exist in polymorphic or amorphous forms and are also within the scope of the present invention. When in solid crystalline form, the compounds of the present invention may be in the form of co-crystals with another chemical entity, and this specification includes all such co-crystals.
In particular, in one aspect, the invention provides cyclic dinucleotide compounds of the formula, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts, prodrugs or solvates thereof:
for example
Wherein X is 1 And X 2 Each independently selected from-OH and-SH; and has a cytotoxic or antiviral effect on at least one of the two nucleosides of the two nucleotides forming the loop, preferably those antimetabolites, i.e., nucleoside anticancer agents or derivatives thereof, including but not limited to, clinically used enocitabine (enocabine), fludarabine (fludarabine), cytarabine, nelarabine (nelarabine), deoxyfluorouridine (doxifluridine), capecitabine (capecitabine), azacytidine (azacitidine), penstatin (pentastatin), cladribine (cladribine), gemcitabine (gemcitabine) and clofarabine (clofarabine), more preferably cladribine, gemcitabine and clofarabine or derivatives thereof. The nucleoside anticancer derivatives described herein refer to compounds which are derived by substituting hydrogen atoms or atomic groups in the structures of the compounds with other atoms or atomic groups and retain anticancer cytotoxicity.
In a specific embodiment, the present invention provides a cyclic dinucleotide compound of formula (Y) below,
wherein X is 1 And X 2 Each independently selected from-OH and-SH;
B 1 is adenine substituted by XWherein X is selected from Cl, F or-NHC 1-6 An alkyl group; or optionally by R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group;
R 1 and R is 1 ' each independently selected from H, F or-OH;
B 2 selected from adenine optionally substituted with XWherein X is selected from H, F or Cl; optionally by R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group; or guanineWherein OH is optionally C 1-6 Alkyl substitution;
represents that the phosphate bond can be attached to the pentose at the 2 'or 3' position, and the site of the pentose, which is not cyclic with the phosphoric acid, is R 2 And R is 2 ' substitution; and
R 2 and R is 2 ' each independently selected from H, -OH or F;
provided that when B 1 Or B is a 2 One of which is optionally R a In the case of substituted cytosine, the carbon atom adjacent to the pentose ring to which it is attached is substituted with two F;
stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts, prodrugs or solvates thereof.
In particular embodiments, the compound of formula (Y) may be
Wherein B is 1 、B 2 、R 1 、R 1 ’、R 2 、R 2 ' has the meaning defined above for the compounds of formula (Y).
In particular embodiments, the two phosphorothioate linkages in the compounds of the present invention, when present, are in the R, R, S, S, S, R or R, S configuration or mixtures thereof. In a preferred embodiment, the two phosphorothioate linkages in the compounds of the present invention, when present, are present in substantially pure form in the R, R, S, S, S, R or R, S configuration, with substantially pure R, R configuration being particularly preferred.
In particular embodiments, a single phosphorothioate bond present in a compound of the present invention may be present in either the R or S configuration, preferably in the R configuration.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 1 Is adenine substituted by XWherein X is selected from Cl or F, preferably Cl, or optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 1 Is adenine substituted by XWherein X is-NHC 1-6 Alkyl radicals, e.g. -NHCH 3 、-NHCH 2 CH 3 、-NHCH 2 CH 2 CH 3 preferably-NHCH 3
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), R 1 And R is 1 ' are both H, or one is H and the other is F, or are both F.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 2 Is adenine optionally substituted with XWherein X is selected from H, F or Cl, preferably H or Cl.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 2 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), B 2 Is guanine
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), R 2 And R is 2 ' are both H, or one is H and the other is F, or one is H and the other is OH, or both are F.
In one embodiment of the compounds of the formula (I), (II), (III) or (IV), R a is-C (O) C 1-14 The alkyl radical may be, for example, -C (O) C 1-10 Alkyl, -C (O) C 1-9 Alkyl, -C (O) C 1-8 Alkyl, -C (O) C 1-7 Alkyl, -C (O) CH (C) 1-4 Alkyl group 2 、-C(O)CH(C 1-3 Alkyl group 2 、-C(O)CH 2 CH(C 1-4 Alkyl group 2 、-C(O)CH 2 CH(C 1-3 Alkyl group 2
The compounds of formula (I), (II), (III) or (IV) of the present invention also encompass any combination between the various embodiments described above and preferred or exemplary modes thereof.
In a further specific embodiment, the present invention provides a cyclic dinucleotide compound of formula (Y) having the following sub-formula,
wherein B is 1 、B 2 、R 1 、R 1 ’、R 2 、R 2 ' has the meaning as defined above for the compounds of formula (I), (II), (III) or (IV) and for the individual embodiments.
In one embodiment of the compounds of the formula (II-a) or (II-B), in particular of the compounds of the formula (II-a), B 1 Is thatAnd B is 2 Is thatIn a specific embodiment, R 1 And R is 1 ' are all H; in another specific embodiment, R 1 And R is 1 ' one is H and the other is F. Further, in each of the specific embodiments, R 2 And R is 2 ' are both H, or one is H and the other is F, or one is H and the other is OH.
In one embodiment of the compounds of the formula (II-a) or (II-B), in particular of the compounds of the formula (II-a), B 1 And B 2 Are all optionally R a Substituted cytosinesIn a specific such embodiment, R 1 、R 1 ’、R 2 、R 2 ' are both F.
In one embodiment of the compounds of the formula (II-a) or (II-B), in particular of the compounds of the formula (II-B), B 1 Is thatAnd B is 2 Is thatIn a specific such embodiment, R 1 And R is 1 ' are all H; in another specific embodiment, R 1 And R is 1 ' one is H and the other is F. Further, in each of the embodiments described, R 2 And R is 2 ' are both H, or one is H and the other is F, or one is H and the other is OH, preferably one is H and the other is OH.
In one embodiment of the compounds of the formula (II-a) or (II-B), in particular of the compounds of the formula (II-B), B 1 Is optionally R a Substituted cytosinesAnd B is 2 Is thatIn a specific such embodiment, R 1 And R is 1 ' are both F. Further, in each of the embodiments described, R 2 And R is 2 ' are both H, or one is H and the other is F, or one is H and the other is OH, preferably one is H and the other is OH.
In one embodiment of the compounds of formula (II-a) or (II-b), R a Is H, or-C (O) C 1-10 Alkyl radicals, e.g. -C (O) C 1-9 Alkyl, -C (O) C 1-8 Alkyl, -C (O) C 1-7 Alkyl, -C (O) CH (C) 1-4 Alkyl group 2 、-C(O)CH(C 1-3 Alkyl group 2 、-C(O)CH 2 CH(C 1-4 Alkyl group 2 、-C(O)CH 2 CH(C 1-3 Alkyl group 2
In one embodiment of the compounds of formula (II-a) or (II-B), B 1 Is thatR 1 And R is 1 ' are all H.
In one embodiment of the compounds of formula (II-a) or (II-B), B 1 Is thatR 1 And R is 1 ' one is H and the other is F; in a particular embodiment, F and B 1 Located on the same side as the ribose.
In one embodiment of the compounds of the formula (II-a) or (II-B), in particular of the compounds of the formula (II-B), B 1 Is thatPreferablyAnd R is 1 And R is 1 ' are H, or R 1 And R is 1 ' one is H and the other is F, preferably R 1 And R is 1 ' one is H and the other is F.
In one embodiment of the compounds of formula (II-a) or (II-B), B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H, R 1 And R is 1 ' are both F.
In one embodiment of the compounds of formula (II-a) or (II-B), B 1 Is optionally R a Substituted cytosinesWherein R is a is-C (O) C 1-10 Alkyl, preferably-C (O) CH (C) 1-4 Alkyl group 2 ,R 1 And R is 1 ' are both F.
In an exemplary embodiment, R a is-C (O) CH (CH) 3 ) 2 、-C(O)CH(CH 2 CH 3 ) 2 、-C(O)CH(CH 3 )(CH 2 CH 3 )、-C(O)CH(CH 2 CH 2 CH 3 ) 2 、-C(O)CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、-C(O)CH 2 CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、C(O)CH 2 CH(CH 2 CH 2 CH 3 ) 2 Preferably R a Is C (O) CH (CH) 2 CH 2 CH 3 ) 2
In one embodiment of the compounds of formula (II-a) or (II-B), B 2 Is guanineOr adenineAnd R is 2 And R is 2 ' one is H and the other is selected from-OH or F.
In one embodiment of the compounds of formula (II-a), B 2 Is guanineR 2 And R is 2 ' one is H and the other is selected from-OH or F.
In one embodiment of the compounds of formula (II-a), B 2 Is adenineR 2 And R is 2 ' one is H and the other is selected from-OH or F.
In one embodiment of the compounds of formula (II-a), B 2 Adenine substituted with halogen XWherein X is Cl; r is R 2 And R is 2 ' one is H and the other is F, preferably F and B 2 Located on the same side as the ribose.
In one embodiment of the compounds of formula (II-a), B 2 Adenine substituted with halogen XWherein X is Cl; r is R 2 And R is 2 ' are all H.
In one embodiment of the compounds of formula (II-a), B 2 Is optionally R a Substituted cytosinesWherein R is a Selected from H, R 1 And R is 1 ' are both F.
In one embodiment of the compounds of formula (II-a), B 2 Is R is a Substituted cytosinesWherein R is a is-C (O) C 1-10 Alkyl, preferably-C (O) CH (C) 1-4 Alkyl group 2 ,R 1 And R is 1 ' are both F.
In an exemplary embodiment, R a is-C (O) C (CH) 3 ) 2 、-C(O)CH(CH 2 CH 3 ) 2 、-C(O)CH(CH 3 )(CH 2 CH 3 )、-C(O)CH(CH 2 CH 2 CH 3 ) 2 、-C(O)CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、-C(O)CH 2 CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、C(O)CH 2 CH(CH 2 CH 2 CH 3 ) 2 Preferably R a Is C (O) CH (CH) 2 CH 2 CH 3 ) 2
In one embodiment of the compounds of formula (II-B), B 2 Is guanineOr adenineR 2 And R is 2 ' one is H and the other is-OH.
In the compounds of formula (II-a) or (II-b) above and in their various embodiments, the two phosphorothioate linkages are in the R, R, S, S, S, R or R, S configuration or mixtures thereof; preferably in substantially pure form, the R, R, S, S, S, R or R, S configuration, particularly preferably in substantially pure R, R configuration.
The embodiments given above for the compounds of the formula (II-a) or (II-b) and their preferred or exemplary forms, respectively, also apply to the compounds of the formula (I-a) or (I-b), the compounds of the formula (III-a) or (III-b), the compounds of the formula (IV-a) or (IV-b), i.e. the invention also covers the compounds of the formula (I-a) or (I-b), the compounds of the formula (III-a) or (III-b), the compounds of the formula (IV-a) or (IV-b), respectively, wherein the individual substituents take the particular definitions given above for the compounds of the formula (II-a) or (II-b) or their particular embodiments.
For example, in one embodiment of a compound of formula (I-a) or (I-B), a compound of formula (III-a) or (III-B), or a compound of formula (IV-a) or (IV-B), especially a compound of formula (I-B), formula (III-B) or (IV-B), B 1 Is thatAnd B is 2 Is thatPreferablyIn a specific embodiment, R 1 And R is 1 ' one is H and the other is F. Further, in each of the embodiments described, R 2 And R is 2 ' one is H and the other is OH.
The compounds of the invention of formula (I-a)/(I-b), (II-a)/(II-b), (III-a)/(III-b) or (IV-a)/(IV-b) also encompass any combination between the various embodiments described above and preferred or exemplary modes thereof.
In still further specific embodiments, the present invention provides cyclic dinucleotide compounds of the sub-formula,
wherein B is 1 、B 2 、R 1 、R 1 ’、R 2 、R 2 ' having the meaning as defined above for the compounds of formula (I), (II), (III) or (IV) or each of its embodiments; more specifically, there are defined the meanings defined above for the compounds of the formula (I-a)/(I-b), (II-a)/(II-b), (III-a)/(III-b) or (IV-a)/(IV-b) or the individual embodiments thereof.
In one embodiment of the compounds of the formula (II-a ') or (II-B '), in particular of the compounds of the formula (II-a '), B 1 Is thatAnd B is 2 Is thatIn a specific such embodiment, R 1 And R is 1 ' are all H; in another specific embodiment, R 1 Is F, R 1 ' is H, i.e., F is on the same side of the ribose as B1. Further, in each of the specific embodiments, R 2 And R is 2 ' are H, or R 2 Is H and R 2 ' is F, or R 2 Is F and R 2 ' is H, or R 2 Is OH and R 2 ' is H.
In a compound of the formula (II-a ') or (II-b'),In particular, in embodiments of the compounds of formula (II-a'), B 1 And B 2 Are all optionally R a Substituted cytosinesIn a specific such embodiment, R 1 、R 1 ’、R 2 、R 2 ' are both F.
In one embodiment of the compounds of the formula (II-a ') or (II-B '), in particular of the compounds of the formula (II-B '), B 1 Is thatAnd B is 2 Is thatIn a specific such embodiment, R 1 And R is 1 ' are all H; in another specific embodiment, R 1 Is F, R 1 ' is H, i.e., F is on the same side of the ribose as B1. Further, in each of the embodiments described, R 2 And R is 2 ' are H, or R 2 Is H and R 2 ' is F, or R 2 Is F and R 2 ' is H, or R 2 Is OH and R 2 ' is H; preferably R 2 Is OH and R 2 ' is H.
In one embodiment of the compounds of the formula (II-a ') or (II-B '), in particular of the compounds of the formula (II-B '), B 1 Is optionally R a Substituted cytosinesAnd B is 2 Is thatIn a specific such embodiment, R 1 And R is 1 ' are both F. Further, in each of the embodiments described, R 2 And R is 2 ' are H, or R 2 Is H and R 2 ' is F, or R 2 Is F and R 2 ' is H, or R 2 Is OH and R 2 ' is H; preferably R 2 Is OH and R 2 ' is H.
In one embodiment of the compounds of the formula (II-a ') or (II-b'), R a Is H, or-C (O) C 1-10 Alkyl radicals, e.g. -C (O) C 1-9 Alkyl, -C (O) C 1-8 Alkyl, -C (O) C 1-7 Alkyl, -C (O) CH (C) 1-4 Alkyl group 2 、-C(O)CH(C 1-3 Alkyl group 2 、-C(O)CH 2 CH(C 1-4 Alkyl group 2 、-C(O)CH 2 CH(C 1-3 Alkyl group 2
In one embodiment of the compounds of the formula (II-a ') or (II-B'), B 1 Is thatR 1 And R is 1 ' are all H, whereby B 1 Forms, together with the ribose to which it is attached, the nucleoside antineoplastic agent cladribine.
In one embodiment of the compounds of the formula (II-a ') or (II-B'), B 1 Is thatR 1 Is F, R 1 ' is H, i.e. F and B 1 On the same side as ribose, whereby the B 1 Forms nucleosides together with the ribose to which they are attachedThe antineoplastic agent clofarabine.
In one embodiment of the compounds of the formula (II-a ') or (II-B '), in particular of the compounds of the formula (II-B '), B 1 Is thatPreferablyAnd R is 1 And R is 1 ' are H, or R 1 Is F and R 1 ' is H, preferably R 1 Is F and R 1 ' is H.
In one embodiment of the compounds of the formula (II-a ') or (II-B'), B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H, R 1 And R is 1 ' are all F, whereby B 1 Together with the ribose to which it is attached, forms the nucleoside antineoplastic agent gemcitabine.
In one embodiment of the compounds of the formula (II-a ') or (II-B'), B 1 Is optionally R a Substituted cytosinesWherein R is a is-C (O) C 1-10 Alkyl, preferably-C (O) CH (C) 1-4 Alkyl group 2 ,R 1 And R is 1 ' are all F, whereby B 1 Together with the ribose to which it is attached, form an alkanoylated derivative of the nucleoside antineoplastic agent gemcitabine. In an exemplary embodiment, R a is-C (O) CH (CH) 3 ) 2 、-C(O)CH(CH 2 CH 3 ) 2 、-C(O)CH(CH 3 )(CH 2 CH 3 )、-C(O)CH(CH 2 CH 2 CH 3 ) 2 、-C(O)CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、-C(O)CH 2 CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、C(O)CH 2 CH(CH 2 CH 2 CH 3 ) 2 Preferably R a Is C (O) CH (CH) 2 CH 2 CH 3 ) 2
In one embodiment of the compounds of formula (II-a'), B 2 Is guanineR 2 ' is H, R 2 Selected from-OH or F.
In one embodiment of the compounds of formula (II-a'), B 2 Is adenineR 2 ' is H, R 2 Selected from-OH or F.
In one embodiment of the compounds of formula (II-a'), B 2 Adenine substituted with halogen XWherein X is Cl, R 2 Is H, R 2 ' is F, and F and B 2 On the same side as ribose, whereby the B 2 Together with the ribose to which it is attached, forms the nucleoside antineoplastic agent clofarabine.
In a formula (II-a')In an embodiment of the compound, B 2 Adenine substituted with halogen XWherein X is Cl, R 2 And R is 2 ' are all H, whereby B 2 Forms, together with the ribose to which it is attached, the nucleoside antineoplastic agent cladribine.
In one embodiment of the compounds of formula (II-a'), B 2 Is optionally R a Substituted cytosinesWherein R is a Selected from H, R 1 And R is 1 ' are all F, whereby B 2 Together with the ribose to which it is attached, forms the nucleoside antineoplastic agent gemcitabine.
In one embodiment of the compounds of formula (II-a'), B 2 Is R is a Substituted cytosinesWherein R is a is-C (O) C 1-10 Alkyl, preferably-C (O) CH (C) 1-4 Alkyl group 2 ,R 1 And R is 1 ' are all F, whereby B 2 Together with the ribose to which it is attached, form an alkanoylated derivative of the nucleoside antineoplastic agent gemcitabine. In an exemplary embodiment, R a is-C (O) CH (CH) 3 ) 2 、-C(O)CH(CH 2 CH 3 ) 2 、-C(O)CH(CH 3 )(CH 2 CH 3 )、-C(O)CH(CH 2 CH 2 CH 3 ) 2 、-C(O)CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、-C(O)CH 2 CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )、C(O)CH 2 CH(CH 2 CH 2 CH 3 ) 2 Preferably R a Is C (O) CH (CH) 2 CH 2 CH 3 ) 2
In one embodiment of the compounds of formula (II-B'), B 2 Is guanineOr adenineR 2 ' is H, R 2 is-OH.
The specific embodiments given above for the compounds of the formula (II-a ') or (II-b'), and the preferred or exemplary modes thereof, are also applicable to the compounds of the formula (I-a ') or (I-b'), the compounds of the formula (III-a ') or (III-b'), the compounds of the formula (IV-a ') or (IV-b'), respectively, i.e., the invention also covers the compounds of the formula (I-a ') or (I-b'), the compounds of the formula (III-a ') or (III-b'), the compounds of the formula (IV-a ') or (IV-b'), respectively, wherein each specific substituent is as defined above for the compounds of the formula (II-a ') or (II-b'), respectively, or the specific embodiments thereof.
For example, in one embodiment of a compound of formula (I-a ') or (I-B '), a compound of formula (III-a ') or (III-B '), or a compound of formula (IV-a ') or (IV-B '), especially a compound of formula (I-B '), formula (III-B ') or (IV-B ') 1 Is thatAnd B is 2 Is thatPreferablyIn a specific embodiment, R 1 Is F and R 1 ' is H. Further, in each of the embodiments described, R 2 Is OH and R 2 ' is H.
The compounds of the present invention encompass the above embodiments, as well as any combination or sub-combination of the above embodiments, and any preferred or exemplary combination of the above embodiments.
Preferred embodiments of the compounds of the present invention include compounds, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof,
pharmacological Activity and advantageous effects of Compounds of the invention
The research shows that the cyclic dinucleotide compound provided by the invention has the following pharmacological activities and beneficial effects:
● The method can effectively stimulate the THP-1 cells to secrete IFN-beta, thereby demonstrating that the THP-1 cells have high affinity to the STING receptor, can effectively activate STING and induce the generation of I-type interferon;
● Effectively inhibit the in vitro growth of a colorectal cancer cell line CT26 of mice, which shows that the colorectal cancer cell line CT has toxic effect on tumor cells and prevents the tumor cells from dividing and propagating;
● The anti-tumor activity in the double-side transplantation tumor model of the immune sound CT26 isogenic mouse is better than that in the immune deficiency mouse model, which shows that the anti-tumor activity and the cytotoxicity effect generated by activating STING are simultaneously generated, and the anti-tumor activity and the cytotoxicity effect generate additive and even synergistic anti-tumor effects;
● The immune memory function is shown in a CT26 isogenic mouse transplanted tumor model, so that the tumor recurrence can be effectively prevented;
● Shows good pharmacokinetic properties in hepatocyte metabolism studies, e.g. with a longer t 1/2 And low clearance rate, so that the dosing interval can be increased, resulting in better patient compliance; and
● The medicine is applied to the focus part locally, has accurate effect, can reduce the medicine consumption, is difficult to diffuse out of the focus due to high molecular polarity, has limited toxicity and has good safety.
Pharmaceutical composition
In another aspect, the invention provides pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients, and methods of making the compositions using the compounds of the invention.
The compositions or dosage forms are formulated, administered and administered in a manner consistent with good medical practice. Factors considered in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the timing of administration, and other factors known to the medical practitioner.
Typical pharmaceutical compositions or dosage forms are prepared by mixing a compound of the invention with a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail, for example, in Gennaro A.R. et al, remington: the Science and Practice of Pharmacy (2000) Lippincott, williams & Wilkins, philadelphia. The formulation may also include one or more buffers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, sweeteners, fragrances, flavoring agents, diluents and other known additives to provide an elegant appearance of the drug (i.e., the compound of the present invention or pharmaceutical composition thereof) or to aid in the preparation of a pharmaceutical product (i.e., a medicament).
The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal administration, and if desired topical treatment, intralesional administration. Parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration.
In a preferred embodiment, a pharmaceutical composition comprising one or more compounds of the invention is provided, wherein the pharmaceutical composition is suitable for intravenous, intratumoral, peritumoral or subcutaneous administration. Intratumoral (direct access to the tumor mass) or peritumoral (peritumoral) administration of the compounds of the invention directly activates locally hydrophilic dendritic cells, directly promotes apoptosis of tumor cells or sensitizes tumor cells to cytotoxic agents.
The compounds of the present invention may be administered in any convenient form of administration, such as tablets, powders, capsules, sterile injectable preparations, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches and the like. Such compositions may contain components conventional in pharmaceutical formulations, such as diluents, carriers, pH modifying agents, preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavoring agents, salts for varying the osmotic pressure, buffers, masking agents, antioxidants and other active agents. They may also contain other therapeutically valuable substances. The various formulations may be prepared according to methods conventional in the formulation art, see for example Gennaro A.R. et al, remington: the Science and Practice of Pharmacy (2000) Lippincott, williams & Wilkins, philadelphia or the pharmacopoeias of various countries.
In a preferred embodiment, pharmaceutical compositions comprising one or more compounds of the invention are provided in the form of parenteral formulations, in particular in the form of sterile injectable formulations, such as sterile injectable solutions or suspensions in a non-toxic and parenterally acceptable diluent or solvent, or as lyophilized powders. Among the acceptable vehicles or solvents, water, 1, 3-butanediol, ringer's solution, or isotonic sodium chloride solution may be employed, for example; in addition, sterile, fixed oils may be conventionally employed as a solvent or suspending medium, and any bland fixed oil may be employed therefor including, for example, synthetic mono-or diglycerides, fatty acids and the like.
The dosage of the compounds of the invention administered can vary within wide limits and, of course, can be adjusted to the individual needs in each particular case. Typically, an effective amount of a compound of the invention is administered systemically in an amount of about 0.1 μg/kg/day to about 50 mg/kg/day, e.g., 0.5 μg/kg/day to about 10 mg/kg/day, 1 μg/kg/day to about 1 mg/kg/day. Each dosage unit may conveniently contain from 0.001 μg to 10mg, for example from 0.01 μg to 1mg, for example from 50 μg to 500 μg. An effective amount may be given in one or more doses, i.e., may be 1, 2 or more administrations, and may be administered multiple times at equal or unequal intervals, including one or more times per day, one or more times per week, or on an administration schedule of every few days/week.
Use and method
In view of the ability of the compounds of the invention to activate STING, induce expression of type I interferons and pro-inflammatory cytokines such as IL-6, TNF- α and IFN- γ, etc., while having cytotoxic activity, another aspect of the invention provides therapeutic uses and methods of the compounds of the invention.
In one aspect, the compounds or pharmaceutical compositions described herein are useful as therapeutic substances for the treatment or prevention of diseases associated with or mediated by immune responses, in particular for the treatment or prevention of diseases associated with or mediated by STING, including inflammatory, allergic or autoimmune diseases, infectious diseases or cancers, or as vaccine adjuvants.
In a preferred embodiment, the compounds or compositions of the invention are used as cytotoxic agents for the treatment or prophylaxis of hyperproliferative diseases, in particular tumours. In further preferred embodiments, the compounds or compositions of the invention are used for the treatment of recurrent tumors, or for preventing tumor recurrence.
In a preferred embodiment, the compounds or compositions of the invention are used as cytotoxic agents for the treatment or prophylaxis of viral infections.
In another aspect, the invention thus accordingly provides a method of inducing, stimulating or aiding an immune response in an individual comprising administering to the individual a compound or pharmaceutical composition of the invention. In one embodiment, the compounds of the invention are administered to an individual as an immunotherapy to induce in vivo production of a variety of cytokines useful therapeutically in humans or animals, including type I interferons and pro-inflammatory cytokines such as IL-6, TNF- α, and IFN- γ, to modulate the immune system of the human or animal to achieve certain therapeutic benefits.
In a further aspect, the present invention accordingly provides a method of treating or preventing a disease associated with or mediated by an immune response, in particular STING, including inflammatory, allergic or autoimmune diseases, infectious diseases or cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a compound or pharmaceutical composition of the invention.
In a preferred embodiment, the present invention provides a method of treating or preventing a hyperproliferative disease, especially a tumor, comprising administering to a subject in need thereof a therapeutically effective amount of a compound or pharmaceutical composition of the present invention. In a further preferred embodiment, the present invention provides a method of treating recurrent tumors or preventing tumor recurrence comprising administering to a subject in need thereof a therapeutically effective amount of a compound or pharmaceutical composition of the present invention.
In a preferred embodiment, the present invention provides a method of treating or preventing a viral infection comprising administering to a subject in need thereof a therapeutically effective amount of a compound or pharmaceutical composition of the present invention.
In a further aspect, the present invention accordingly provides the use of a compound or a pharmaceutical composition of the invention for the manufacture of a medicament for the treatment or prophylaxis of a disease associated with or mediated by an immune response, particularly STING, including inflammatory, allergic or autoimmune diseases, infectious diseases or cancer.
In a further aspect, the present invention thus accordingly also provides the use of a compound or pharmaceutical composition of the invention in the preparation of a vaccine adjuvant.
In a preferred embodiment, the present invention provides the use of a compound or pharmaceutical composition of the invention in the manufacture of a medicament for the treatment or prophylaxis of hyperproliferative diseases, in particular tumours. In a further preferred embodiment, the present invention provides the use of a compound or pharmaceutical composition of the invention in the manufacture of a medicament for the treatment of recurrent tumours or for the prevention of tumour recurrence.
In a preferred embodiment, the present invention provides the use of a compound or pharmaceutical composition of the invention in the manufacture of a medicament for the treatment or prophylaxis of a viral infection.
Inflammation for the above uses and methods may be acute or chronic, involving inflammation of any organ or tissue of the body, including musculoskeletal inflammation, vascular inflammation, neuroinflammation, digestive system inflammation, ocular inflammation, reproductive system inflammation or other inflammation, but also autoimmune and allergic disorders of inflammatory nature such as contact dermatitis, urticaria and airway allergies.
Autoimmune diseases for the above uses and methods refer to diseases in which an organism immunoreacts with an autoantigen resulting in damage to the self tissue, including, but not limited to, systemic lupus erythematosus, psoriasis, insulin dependent diabetes mellitus, dermatomyositis, sjogren's syndrome, chronic fatigue syndrome, aplastic anemia, autoimmune hepatitis, multiple sclerosis, optic neuritis, pemphigus, rheumatoid arthritis, ulcerative colitis, crohn's disease, scleroderma, and the like.
Hyperproliferative diseases with respect to the above uses and methods refer to physiological conditions in a subject characterized by uncontrolled or deregulated cell growth or death, particularly tumors or cancers, including solid tumors and blood-borne tumors, including but not limited to brain, skin, bladder, ovary, breast, stomach, pancreas, prostate, colon, blood, lung and bone cancers. Examples of the above-mentioned types of cancers include neuroblastoma, intestinal cancer such as rectal cancer, colon cancer, familial adenomatous polyposis cancer and hereditary nonlymphoid colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, peritoneal cancer, soft tissue sarcoma, urothelial cancer, sweat gland cancer, gastric cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, endometrial cancer, pancreatic cancer, prostate cancer, testicular cancer, breast cancer (including HER2 negative breast cancer), urinary cancer, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, neuroblastoma and peripheral nerve ectodermal tumor, hodgkin's lymphoma, non-hodgkin's lymphoma, burkitt's lymphoma, acute Lymphoblastic Leukemia (ALL), chronic Lymphocytic Leukemia (CLL), chronic myelogenous leukemia (CLL) and lymphocytic carcinoma, acute Myelogenous Leukemia (AML), myelogenous leukemia (chronic myelogenous leukemia (CML), adult T-cell lymphoma, diffuse lymphoma (DLBCL), hepatoma, multiple myeloma, seminoma, osteosarcoma, chondrosarcoma, anal canal carcinoma, adrenocortical carcinoma, chordoma, fallopian tube carcinoma, gastrointestinal stromal tumor, myeloproliferative disorders, mesothelioma, biliary tract carcinoma, ewing's sarcoma and other rare tumor types, as well as recurrent forms of the foregoing tumors.
In a preferred embodiment, the hyperproliferative disease for the above uses and methods is small cell lung cancer, non-small cell lung cancer, colorectal cancer, liver cancer, breast cancer, ovarian cancer, gastric cancer, prostate cancer, melanoma, renal cell carcinoma, head and neck cancer, pancreatic cancer, hodgkin's lymphoma, leukemia or bladder cancer.
Viral infection for the above uses and methods refers to the process by which viruses invade the body and proliferate in susceptible host cells by a variety of routes, and the viruses involved include, but are not limited to, double-and single-stranded DNA viruses, single-plus-strand RNA viruses, single-minus-strand RNA viruses and double-stranded RNA viruses, and retroviruses, examples being hepatitis b virus, TTV virus, adenovirus, papilloma virus, herpes zoster virus, smallpox virus and vaccinia virus, influenza virus, swine fever virus, hepatitis a virus, hepatitis c virus, hepatitis delta virus, hepatitis e virus, hepatitis hept virus, rabies virus, ebola virus, enterovirus, and human immunodeficiency virus, among others. The therapeutic uses and methods provided by the present invention are useful for the viral infections and diseases caused thereby.
The use of the compounds or compositions of the invention as vaccine adjuvants and in the preparation of vaccine adjuvants as described above means that the compounds or compositions of the invention may be used as adjuvants in therapeutic or prophylactic strategies employing vaccines, i.e. the compounds or compositions of the invention are used together with one or more vaccines selected for stimulating an immune response to one or more predetermined antigens, said vaccines comprising inactivated or attenuated bacteria or viruses, e.g. comprising inactivated tumor cells expressing and secreting one or more of GM-CSF, CCL-20, CCL3, IL-12p70, FLT-3 ligand, cytokines.
Pharmaceutical combination
In view of the advantageous pharmacological activity of the compounds of the invention they may, in addition to being useful alone in the therapeutic uses or methods as mentioned above, be used in combination with at least one other therapeutic agent or therapy to provide further therapeutic benefit.
Thus, in another aspect the invention also provides a pharmaceutical combination comprising a cyclic dinucleotide compound as described herein, a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt, a prodrug or solvate thereof, or a pharmaceutical composition thereof, and at least one other therapeutic agent, or both.
In another aspect, the invention provides a pharmaceutical composition comprising a pharmaceutical combination as described herein and one or more pharmaceutically acceptable excipients.
In another aspect, the invention provides the use of a pharmaceutical composition comprising a pharmaceutical combination as described herein or comprising the same, for the treatment or prophylaxis of a disease, or in the manufacture of a medicament for the treatment or prophylaxis of a disease, including hyperproliferative diseases, viral infections or diseases associated with or mediated by STING, more particularly inflammatory, allergic or autoimmune diseases, infectious diseases or cancers. In a preferred embodiment, the disease to which the use relates is a tumour or a viral infection.
In another aspect, the invention also provides a method of treatment wherein a compound of the invention is administered with a variety of other therapeutic agents.
Inflammation, autoimmune diseases, hyperproliferative diseases and viral infections for the use of the pharmaceutical compositions of the invention and pharmaceutical compositions comprising the same are as described above for the uses and methods of the invention.
The compounds of the present invention may also be administered in combination with surgery, radiation therapy, transplantation (e.g., stem cell transplantation, bone marrow transplantation), immune tumor agents, and the like.
Other therapeutic agents used in combination with the present invention may be administered simultaneously, separately or sequentially with the compounds of the present invention via the same or different routes of administration. The additional therapeutic agent may be co-administered with the compound of the invention in a single pharmaceutical composition or separately administered in separate discrete units from the compound of the invention, e.g., a combination product, preferably in kit form, and may be administered simultaneously or sequentially when administered separately, which sequential administration may be near or distant in time. They may be prepared and/or formulated by the same or different manufacturers. Moreover, the compounds of the invention and other therapeutic agents may (i) be administered prior to delivery of the combination product to the physician (e.g., in the case of a kit comprising a compound of the invention and an additional drug); (ii) By the physician himself immediately prior to administration (or under the direction of the physician); (iii) The combination therapy is added by the patient himself, for example together during the sequential administration of the compounds of the invention and the other therapeutic agents.
Thus, in a further aspect, the invention also provides a kit comprising two or more separate pharmaceutical compositions, at least one of which comprises a cyclic dinucleotide compound of the invention, a stereoisomer, tautomer, stable isotopic variant, pharmaceutically acceptable salt, prodrug or solvate thereof, the remaining co-existing pharmaceutical composition comprising at least one other therapeutic agent, and means for separately containing said compositions, such as a container, a split-bottle or a discrete foil package, for example a blister package for packaging tablets, capsules or the like. The kit of the invention is particularly suitable for administration of different dosage forms, such as oral and parenteral dosage forms, or for administration of different compositions at different dosage intervals.
The additional therapeutic agent may be one or more additional compounds of the invention, or may be a second or additional (e.g., third) therapeutic agent that is compatible with, i.e., does not adversely affect each other, or has complementary or additional activity.
In particular embodiments, other therapeutic agents that may be used in combination with the compounds of the present invention include, but are not limited to, vaccines, adjuvants, immune checkpoint inhibitors, T cell receptor agonists, TLR agonists, therapeutic antibodies, lipids, liposomes, chemotherapeutic agents, immunoregulatory cell lines, and the like.
In a particular embodiment, adjuvants used in combination with the compounds of the present invention, by their nature, may be used to stimulate or otherwise utilize the immune system to react to cancer antigens present on tumor cells, including but not limited to lipids, liposomes, fire-fighting bacteria that induce innate immunity, compounds that mediate innate immune activation, and the like.
In a specific embodiment, the immune checkpoint inhibitor used in combination with a compound of the invention is for example selected from CTLA-4 pathway antagonist, PD-1 pathway antagonist, tim-3 pathway antagonist, vista pathway antagonist, BTLA pathway antagonist, LAG-3 pathway antagonist or TIGIT pathway antagonist.
In a specific embodiment, T cell receptor agonists used in combination with the compounds of the invention include, but are not limited to, CD28 agonists, OX40 agonists, GITR agonists, CD137 agonists, CD27 agonists or HVEM agonists.
In a specific embodiment, TLR agonists used in combination with the compounds of the invention include, but are not limited to, pam3Cys, CFA, MALP2, pam2Cys, FSL-1, hib-OMPC, poly-adenylate (poly AU), LPS, bacterial flagellin, monophosphoryl lipid a (MPL), imiquimod, lei Ximo t, loxoribine, and the like.
In a specific embodiment, chemotherapeutic agents used in combination with the compounds of the present invention include, but are not limited to, alkylating antineoplastic agents, platinum-based antineoplastic agents, antimetabolites, antimicrotubule agents, antimitotic agents, topoisomerase inhibitors, and antitumor antibiotics, among others.
In a specific embodiment, therapeutic antibodies for use in combination with the compounds of the invention include, but are not limited to, murominab-CD 3, infiniximabAnd adalimumabOmalizumab Daclizumab Rituximab Ibritumomab Tositumomab Cetuximab Trastuzumab Alemtuzumab Lym-1 Ipilimumab Vitaxin、Bevacizumab Abciximab Other therapeutic antibodies that may be used in combination include prolactin receptor inhibitors, HER3 inhibitors, EGFR2 and/or EGFR4 inhibitors, M-CSF inhibitors, anti-APRIL antibodies or anti-SIRPa or anti-CD 47 antibodies.
In other embodiments, the compounds of the invention may also be used in combination with PKC inhibitors, BCR-ABL inhibitors, HSP90 inhibitors, PI3K and/or inhibitors of mTOR, FGFR inhibitors, inhibitors of cytochrome P450 HDM2 inhibitors, aromatase inhibitors, P53 and/or P53/Mdm2 interaction inhibitors, or CSF-1R tyrosine kinase inhibitors.
Examples of the types of therapeutic agents described above, as well as other examples of therapeutic agents that may be used in combination with the compounds of the present invention, are described in WO2016/145102 and WO2018/060323, the respective contents of which are incorporated herein.
For the above-described compounds, pharmaceutical compositions, methods, uses, pharmaceutical combinations and kits of the present invention, the cyclic dinucleotide compounds of the present invention, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts, prodrugs or solvates of the various preferred embodiments described hereinabove or the pharmaceutical compositions thereof, more preferably the compounds defined in the specific embodiments, i.e. the compounds of examples 1 to 26, most preferably those compounds showing excellent activity in the active examples.
For the compounds, pharmaceutical compositions, methods, uses, pharmaceutical combinations and kits of the invention described above, it is preferred to use the free form or pharmaceutically acceptable salt or prodrug of the cyclic dinucleotide compound as defined herein, preferably the free form or pharmaceutically acceptable salt or prodrug of the substantially pure cyclic dinucleotide compound as defined herein.
For the therapeutic uses and methods of the invention described above, the subject preferably used or treated is a mammal, preferably a human.
When a dose of a drug or a pharmaceutically acceptable salt thereof is described herein, it is to be understood that the dose is based on the weight of the free base, excluding any hydrates or solvates thereof, unless otherwise indicated.
Process for the preparation of the compounds of the invention
General synthetic method
The compounds of the invention, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof can be prepared by a variety of methods well known in the art of organic synthesis, including the methods set forth below, the methods set forth in the examples, or similar methods as understood by those skilled in the art.
The following illustrates a general synthetic scheme for the synthesis of the compounds of the present invention. Suitable reaction conditions for the individual reaction steps are known to the person skilled in the art or can be determined routinely. In particular, the process steps for the synthesis of the compounds of the invention can be carried out under reaction conditions known per se, including those specifically mentioned, in the absence or generally in the presence of solvents or diluents, including, for example, solvents or diluents which are inert with respect to the reagents used and which dissolve them, in the absence or presence of catalysts, condensing agents or neutralizing agents, for example ion exchangers, such as cation exchangers, for example in the H+ form, at reduced, normal or elevated temperatures, for example from about-100℃to about 190℃including, for example, from about-78℃to about 150℃such as from about 0℃to about 125℃at room temperature, -20 to 40℃or reflux temperatures, under atmospheric pressure or in closed containers, when appropriate under pressure, and/or under an inert atmosphere, for example oxygen or nitrogen atmosphere, depending on the nature of the reaction and/or of the reactants.
The starting materials and reagents used in the preparation of these compounds are generally commercially available or may be prepared by the methods set forth below, methods analogous to those set forth below, or methods known in the art. If desired, the starting materials and intermediates in the synthetic reaction scheme may be isolated and purified using conventional techniques including, but not limited to, filtration, distillation, crystallization, chromatography, and the like. The materials may be characterized using conventional methods including physical constants and spectroscopic data.
Solvents suitable for use in any particular reaction include, unless otherwise indicated in the description of the process: those solvents specifically mentioned, or for example water; esters, such as lower fatty acid lower alkyl esters, e.g., ethyl acetate; ethers such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofuran or dioxane; liquid aromatic hydrocarbons such as benzene or toluene; alcohols, such as methanol, ethanol or 1-or 2-propanol, such as acetonitrile; halogenated hydrocarbons such as methylene chloride or chloroform; amides such as N, N-dimethylformamide or N, N-dimethylacetamide; bases, such as heterocyclic nitrogen bases, e.g., pyridine; carboxylic anhydrides, such as lower aliphatic carboxylic anhydrides, e.g. acetic anhydride; cyclic, linear or branched hydrocarbons, such as cyclohexane, hexane or isopentane; or a mixture of these solvents, such as an aqueous solution. Such solvent mixtures may also be used for working up, for example by chromatography or partitioning.
One skilled in the art will recognize the existence of stereocenters in the compounds of formula I. At all stages of the reaction, the resulting mixture of isomers may be separated into individual isomers, e.g. diastereomers or enantiomers, or into any desired mixture of isomers, e.g. racemates or mixtures of diastereomers, see e.g. E.L.Eliel, S.H.Wilen and l.n. Mander "Stereochemistry of Organic Compounds" (Wiley-Interscience, 1994).
Scheme 1 below illustrates one general synthetic route that may be used to prepare the compounds of formula II and their various embodiments as defined herein. The variables of the general formula in the schemes below have the same meaning as the compounds defined herein or in the various embodiments thereof, unless otherwise indicated.
Scheme 1
Wherein P is 1 And P 2 Is a suitable hydroxy protecting group, P 3 And P 4 Is a suitable hydroxy or amino protecting group including, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl). Deprotection as used in the synthetic schemes of the compounds of the present invention is carried out in the presence of acidic conditions (including but not limited to, e.g., acetic acid/water, trifluoroacetic acid/water, etc.), basic conditions (including but not limited to, e.g., aqueous ammonia, ammonia/methanol solutions, etc.), or fluorine-containing anion compounds (including but not limited to, e.g., tetrabutylammonium fluoride, triethylamine trihydrofluoride, etc.).
Scheme 2 illustrates the synthetic routes for compounds of formula a and formula C in scheme 1, as well as the synthesis of intermediates further used therein.
Scheme 2
Scheme 3 illustrates the synthetic route when the compound of formula H is a compound of formula H-1 (gemcitabine prodrug LY 2334737) as follows.
Scheme 3
Schemes 4-7 illustrate the synthetic routes for compounds of formula B in scheme 1, as well as the synthesis of intermediates further used therein.
Scheme 4
Scheme 5
Scheme 6
Scheme 7
Scheme 8 illustrates the synthetic route for the compound of formula D in scheme 1.
Scheme 8
Specifically, the present invention provides a process for producing the above-mentioned compound of the present invention, which comprises:
the compound of formula A
Wherein B is 1 、R 1 、R 1 ' having the meaning as defined hereinabove for the compounds of formula (II) of the invention or for the various embodiments thereof; p1 is a suitable hydroxy protecting group such as, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl);
with a compound of formula B in the presence of a base such as DBU,
wherein B is 2 、R 2 、R 2 ' having the meaning as defined hereinabove for the compounds of formula (II) of the invention or for the various embodiments thereof; p2 is a suitable hydroxy protecting group, and P3 and P4 are each a suitable hydroxy or amino protecting group, such as, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl);
or by reacting a compound of formula C
Wherein B is 1 、R 1 、R 1 ' and P1 are as defined above for compounds of formula A;
with a compound of formula D in the presence of a base such as DBU,
wherein B is 2 、R 2 、R 2 ' P2, P3 and P4 are as defined above for the compound of formula B;
to obtain a compound of the formula E,
selective deprotection of a compound of formula E above, e.g. under trifluoroacetic acid/water, tetrabutylammonium fluoride or triethylamine trihydrofluoride conditions, gives a compound of formula F
Wherein each group has the meaning defined above;
a) When R is 2 When the protecting group P3 is benzoyl, the compound of formula F is cyclized with a (-) -PSI reagent in the presence of a base, e.g., DBU, to give a cyclic dinucleoside compound of formula G
Then removing benzoyl protection in ammonia water or ammonia methanol solution to obtain a cyclic dinucleoside compound of formula II,
wherein R is 2 ' = -O (H), whichThe remaining groups have the meanings defined above for the compounds of the formula II or for the individual embodiments thereof;
or b) when R 2 When' = -F or-H, the compound of formula F is cyclized with a (-) -PSI reagent in the presence of a base, e.g., DBU, to give a cyclic dinucleoside compound of formula II
Wherein R is 2 ' the remaining groups have the meaning defined above for compounds of formula II or for the individual embodiments thereof.
The compounds of formula a and the compounds of formula C may be prepared as follows:
the compounds of formula H are selectively protected against primary alcohols in the presence of a base such as imidazole,
obtaining a compound of formula C
Reacting a compound of formula C above with a (+) -PSI reagent in the presence of a base, such as DBU, to provide a compound of formula A,
wherein B is 1 、R 1 、R 1 ' and P1 are as defined above for the compounds of formula A.
Wherein when the compound of formula H is a compound of formula H-1 (gemcitabine prodrug LY 2334737),
which is prepared by a compound of the formula H-1-1 and 2-propylvaleric acid,
the compound of formula B may be prepared as formula B-1 as follows:
the compound of formula H is protected at two hydroxy groups in the presence of a base such as imidazole,
to give a compound of the formula B-1-1
The compound of formula B-1-1 is subjected to selective removal of protecting groups on primary alcohols under, for example, trifluoroacetic acid/water conditions to give the compound of formula B-1
Wherein B is 2 、R 2 、R 2 ' P2 are as defined above for the compound of formula B.
The compounds of formula B may also be prepared as formula B-2 as follows:
the compounds of formula C are further protected against secondary alcohols in the presence of a base such as DBU/pyridine,
to give a compound of the formula B-2-1
Selectively removing the protecting group on the primary alcohol of the compound of the formula B-2-1 under the condition of acetic acid/water, for example, to obtain the compound of the formula B-2,
wherein B is 2 、R 2 、R 2 ' P1, P2 are as defined above for compounds of formula A or formula B.
The compounds of formula B may also be prepared as formula B-3 as follows:
the compounds of formula B-3-1 are prepared by selectively protecting a primary alcohol and a secondary alcohol in the presence of a base, such as imidazole,
to obtain a compound of the formula B-3-2,
reacting a compound of the above formula B-3-2 with a protecting group, such as benzoyl chloride, in the presence of a base, such as N-methylimidazole, with an unprotected secondary alcohol thereof and an amino group on the base to give a compound of the formula B-3-3
Selectively removing the protecting group on the primary alcohol of the compound of the formula B-3-3 under the condition of trifluoroacetic acid/water, for example, to obtain the compound of the formula B-3,
wherein B is 2 、R 2 、R 2 ' P1, P2 and P3 are as defined above for compounds of formula A or formula B.
The compounds of formula B may also be prepared as formula B-4 as follows:
reacting a compound of formula B-4-1 with a secondary alcohol unprotected therein using a protecting group, such as benzoyl chloride, in the presence of a base, such as N-methylimidazole,
to give a compound of the formula B-4-2,
selective removal of protecting groups from primary alcohols by compounds of formula B-4-2 under, for example, acetic acid/water conditions, gives compounds of formula B-4
Wherein B is 2 、R 2 、R 2 ' P1, P2 and P4 are as defined above for compounds of formula A or formula B.
The compounds of formula D may be prepared as follows:
reacting a compound of formula B with a (-) -PSI reagent in the presence of a base, such as DBU,
obtaining a compound of formula D
Wherein B is 2 、R 2 、R 2 ’、P 2 、P 3 P 4 As defined above for compounds of formula B.
Scheme 9 below illustrates one general synthetic route that may be used to prepare compounds of formula (I-b), (II-b), (III-b), (IV-b) and various embodiments thereof as defined herein. The variables of the general formula in the schemes below have the same meaning as the compounds defined herein or in the various embodiments thereof, unless otherwise indicated.
Scheme 9
Wherein each X is independently hydroxy or mercapto; r is R 1 、R 1 ’、B 1 And B 2 Each as defined above for formula (I-b), (II-b), (III-b), (IV-b) and the various sub-formulae and embodiments thereof; p (P) 1 And P 2 Is a suitable hydroxy protecting group, P 3 And P 4 Is a suitable hydroxy or amino protecting group including, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl). P (P) 5 Is a suitable hydroxy or sulfhydryl protecting group on phosphoric acid/phosphate esters, inclusionIncluding but not limited to nitrile ethyl.
Deprotection as used in the above-described synthetic schemes of the compounds of the present invention is carried out in the presence of acidic conditions (including but not limited to, e.g., acetic acid/water, trifluoroacetic acid/water, etc.), basic conditions (including but not limited to, e.g., aqueous ammonia, ammonia/methanol solution, methylamine/ethanol solution, lithium hydroxide, etc.), or fluorine-containing anion compounds (including but not limited to, e.g., tetrabutylammonium fluoride, triethylamine trihydrofluoride, ammonium fluoride, etc.). The oxidation conditions used in the synthetic schemes of the compounds of the present invention are carried out in the presence of, but not limited to, iodine and the sulfidation conditions are carried out in the presence of, but not limited to, 3H-1, 2-benzodisulfophen-3-one.
Scheme 10 illustrates the synthetic route for the compound of formula J in scheme 9, as well as the synthesis of intermediates further used therein.
Flow 10
Scheme 11 illustrates the synthetic route for the compounds of formula K, as well as the synthesis of intermediates used further therein.
Scheme 11
Specifically, the present invention provides a process for producing the above-mentioned compound of the present invention, which comprises:
compounds of formula J
Wherein B is 1 、R 1 、R 1 ' having the formula (I-b), (II-b), (III-b), (IV-b) or the individual embodiments of the compounds according to the invention described aboveDefined meanings; p3 is a suitable hydroxy protecting group such as, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl) is used,
with a compound of formula K in the presence of a base (e.g., DBU),
wherein B is 2 Having the meaning as defined above for the compounds of the formula (I-b), (II-b), (III-b), (IV-b) according to the invention or for the individual embodiments thereof; p1, P2 are suitable hydroxy protecting groups and P4 is a suitable hydroxy or amino protecting group such as, but not limited to, TBS (t-butyldimethylsilyl), DMTr (bis (4-methoxyphenyl) benzyl), bz (benzoyl), i- BuCO (isobutyryl) is used,
Or reacting a compound of formula J with a compound of formula L in the presence of tetrazole and oxidizing or sulfiding, wherein the oxidizing conditions include, but are not limited to, the use of iodine, t-butyl hydroperoxide, etc., and the sulfiding conditions include, but are not limited to, the use of N, N-dimethyl-N' - (3-thio-3H-1, 2, 4-dithiazol-5-yl) cimetidine (DDTT), 3H-1, 2-benzodisulfophen-3-one, etc.; the introduction and removal of the protecting groups is carried out according to standard methods well known to the person skilled in the art,
wherein B is 2 P1, P2 and P4 are as defined above for the compound of formula K;
to obtain the compound of the formula M,
wherein X is OH or SH, P 5 Is a suitable hydroxy or sulfhydryl protecting group on phosphoric acid/phosphate esters such as, but not limited to, nitrile ethyl;
selective deprotection of a compound of formula M above, e.g., under lithium hydroxide conditions, gives a compound of formula N
Wherein each group has the meaning defined above;
a) Reacting a compound of formula N with a (+) -PSI reagent or a (+) -PSI reagent in the presence of a base, such as DBU, to give a compound of formula O,
then deprotecting the P1 (e.g., DMTr) in acetic acid/water to obtain the compound of formula P,
closing the ring of the compound of formula P in the presence of a base, such as DBU, gives a cyclic dinucleoside compound of formula Q,
then deprotecting P4 (e.g., TBS) under conditions such as ammonium fluoride to give a cyclic dinucleoside compound of formula (I-b), (II-b), (III-b) or (IV-b), wherein R 2 Is OH and R 2 'is H, and the' is H,
wherein each group has the meaning defined above;
b) Or reacting a compound of formula N with diphenyl phosphite in the presence of a base, such as DBU, to give a compound of formula R,
then deprotecting the P1 (e.g., DMTr) in acetic acid/water to obtain the compound of formula S,
closing ring of compound of formula S in the presence of activating reagent such as pivaloyl chloride, and oxidizing or sulfurating to obtain compound of formula Q cyclic dinucleoside
Then deprotecting P4 (e.g., TBS) under conditions such as ammonium fluoride to give a cyclic dinucleoside compound of formula (I-b), (II-b), (III-b) or (IV-b), wherein R 2 Is OH and R 2 'is H, and the' is H,
wherein the individual radicals have the meanings defined above.
The compounds of formula J may be prepared as follows:
protecting a secondary alcohol by reacting a compound of formula C in the presence of a base such as N-methylimidazole,
to give a compound of the formula J-1,
selectively deprotecting the above compound of formula J-1 to give a compound of formula J,
wherein the individual radicals are as defined above.
The compounds of formula K can be prepared as follows:
reacting a compound of formula K-1 with a (+) -PSI reagent in the presence of a base, e.g., DBU,
obtaining a compound of formula K
Wherein the individual radicals are as defined above.
The experimental materials and reagents used in the above synthetic methods and schemes, unless otherwise indicated, are available from commercial sources, are prepared according to prior art methods or are prepared according to methods analogous to those disclosed herein. The synthesis conditions used in the above synthesis methods and schemes, unless otherwise specified, can be routinely determined by one of skill in the art.
The application also relates to a preparation method as follows: wherein the compound obtainable as an intermediate in any of the various preparation processes and schemes described herein is used as starting material and the remaining process steps are carried out, or wherein the starting material is formed in situ under the reaction conditions or is used in the form of a derivative, for example in protected form or in salt form, or the compound obtainable according to the process of the application is produced under the process conditions and is further treated in situ.
Examples
The application is further illustrated below with reference to examples. It should be noted that the following examples should not be construed as limiting the scope of the present application.
Unless a structural formula is clearly wrong, when the chemical name of any compound of the present application is not consistent with the given structural formula, the structural formula is subject.
The experimental procedures, under which the specific conditions are not noted in the examples below, are generally carried out according to the usual conditions for such reactions, or according to the conditions recommended by the manufacturer. The experimental materials and reagents used in the following examples were obtained from commercial sources, prepared according to the methods of the prior art or prepared according to methods similar to those disclosed in the present application unless otherwise specified.
Percentages and parts are weight percent and parts unless otherwise indicated; the ratio of the liquids is the volume ratio; all temperatures are given in degrees celsius unless otherwise indicated.
In the following examples of the embodiments described in the examples, 1 h NMR spectra 31 P NMR spectra are typically recorded using Bruker 400MHz NMR and 500MHz NMR, chemical shifts are expressed as delta (ppm); mass spectra were recorded using an Agilent1290 liquid chromatograph+6120b mass spectrometer LCMS liquid chromatography mass spectrometer. Silica gel column purification is carried out by Biotage SelektSEL-2SV or ISO-1 SV; preparative liquid chromatography purification was performed using Gilson 281 (column: waters Xridge 19X250mM,5 μm or WELCH C18,21.2X250mM,10 μm. Mobile phase: A: water (10 mM NH) 4 HCO 3 Or 0.05% formic acid), B: acetonitrile (or 0.05% formic acid). Flow rate: 20-30mL/min. Detection wavelength: 214nm/254 nm), or otherwise indicated.
The following abbreviations are used in the synthesis examples, and each abbreviation not listed has the meaning commonly understood by those skilled in the art.
List of abbreviations
CDCl 3 Deuterated chloroform
DMSO-d 6 Deuterated dimethyl sulfoxide
MHz megahertz
MS-ESI electrospray mass spectrometry
DBU 1, 8-diazabicyclo undec-7-ene
DMF N, N-dimethylformamide
CDI N, N' -carbonyldiimidazole
TMSCl trimethylchlorosilane
TFA trifluoroacetic acid
THF tetrahydrofuran
H 2 O water
HOAc acetic acid
BzCl benzoyl chloride
IFN interferon
FBS fetal bovine serum
PBS phosphate buffer solution
ELISA enzyme-linked immunosorbent assay
(+) -PSI reagent (2R, 3aR,6S,7 aR) -3 a-methyl-2- ((perfluorophenyl) thio) -6- (propyl-1-en-2-yl) hexahydrobenzo [ d ]][1,3,2]Oxathiolane-2-sulfide) (CAS: 2245335-71-9)
(-) -PSI reagent (2S, 3aS,6R,7 aS) -3 a-methyl-2- ((perfluorophenyl) thio) -6- (propyl-1-en-2-yl) hexahydrobenzo [ d][1,3,2]Oxathiolane-2-sulfide (CAS: 2245335-70-8)
Synthetic examples
Preparation of intermediate 1: 2-chloro-5 ' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine
To a solution of 2-chloro-2 '-deoxy-2' -fluoro-beta-adenosine (2.50 g,8.25 mmol) in DMF (15 mL) was added imidazole (1.12 g,16.5 mmol) and tert-butyldimethylchlorosilane (1.31 g,8.66 mmol) in sequence, and the resulting mixture was stirred at 20-25℃for 16 h. The reaction mixture was poured into water (150 mL) and extracted twice with ethyl acetate (100 mL each). The organic phases were combined, washed twice with saturated brine (100 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (2:3) to give a white solid (2.53 g).
1 H NMR(400MHz,DMSO-d 6 )δ8.18(s,1H),7.89(brs,2H),6.33(dd,J=12.0,4.8Hz,1H),6.00(d,J=4.8Hz,1H),5.28(dt,J=52,4.8Hz,1H),4.50-4.30(m,1H),3.98-3.73(m,3H),0.89(s,9H),0.07(s,6H)。
Preparation of intermediate 2: (2S, 3aR,6S,7 aR) -2- ((2R, 3R,4S, 5R) -5- (6-amino-2-chloro-9H-purin-9-yl) -2- ((tert-butyldimethylsilyl) oxy) methyl) -4-fluorotetrahydrofuran-3-yl) oxy) -3 a-methyl-6- (isopropyl-1-en-2-yl) hexahydrobenzo [ d ]][1,3,2]Oxathiophospholane 2-sulfides
To a solution of intermediate 1 (1.00 g,2.40 mmol) and (+) -PSI reagent (1.39 g,3.12 mmol) in tetrahydrofuran (10 mL) at 0deg.C was added 1, 8-diazabicycloundec-7-ene (474 mg,3.12 mmol) and the resulting mixture was stirred at 0-5deg.C for 1 hour. The reaction mixture was diluted with ethyl acetate (50 mL). The organic phase was washed with a 10% sodium dihydrogen phosphate aqueous solution (50 mL) and a saturated brine (50 mL) in this order, and then dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (3:7) to give a white solid (1.50 g).
1 H NMR(400MHz,CDCl 3 )δ8.09(s,1H),6.52-6.40(m,1H),6.23(brs,2H),5.55-5.40(m,1H),5.30-5.06(m,2H),4.92(s,1H),4.52-4.40(m,1H),4.20-4.12(m,1H),4.08-3.90(m,2H),2.62-2.52(m,1H),2.36-2.23(m,1H),2.20-2.08(m,1H),2.02-1.80(m,3H),1.80-1.62(m,7H),0.93(s,9H),0.12(s,6H)。 31 P NMR(162MHz,CDCl 3 )δ101.9。
Preparation of intermediate 3: 2-chloro-5 '-O-tert-butyldimethylsilyl-2' -deoxyadenosine
Intermediate 3 was obtained from 2' -deoxyadenosine by the route of intermediate 1.
1 H NMR(400MHz,DMSO-d 6 )δ8.29(s,1H),7.82(brs,2H),6.26(t,J=6.4Hz,1H),5.37(d,J=4.0Hz,1H),4.45-4.32(m,1H),3.90-3.76(m,1H),3.73-3.60(m,2H),2.75-2.61(m,1H),2.37-2.25(m,1H),0.84(s,9H),0.02(s,6H)。
Preparation of intermediate 4: (2S, 3aR,6S,7 aR) -2- ((2R, 3S, 5R) -5- (6-amino-2-chloro-9H-purin-9-yl) -2- ((tert-butyldimethylsilyl) oxy) methyl) tetrahydrofuran-3-yl) oxy) -3 a-methyl-6- (propyl-1-en-2-yl) hexahydrobenzo [ d ] ][1,3,2]Oxathiophospholane 2-sulfides
Intermediate 4 is obtained from intermediate 3 and (+) -PSI reagent by the route of intermediate 2.
1 H NMR(400MHz,CDCl 3 )δ8.65(s,1H),6.59-6.52(m,1H),5.49-5.40(m,1H),5.09(s,1H),4.92(s,1H),4.52-4.40(m,2H),4.25-4.12(m,2H),3.96(s,2H),2.90-2.81(m,1H),2.69-2.56(m,2H),2.20-2.08(m,2H),2.02-1.80(m,3H),1.80-1.62(m,7H),0.91(s,9H),0.14(s,6H)。 31 P NMR(162MHz,CDCl 3 )δ100.6。
Preparation of intermediate 5:5 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine
Intermediate 5 is obtained from 2' -deoxy-2 ',2' -difluorocytidine by the route of intermediate 1.
1 H NMR(400MHz,DMSO-d 6 )δ7.63(d,J=7.6Hz,1H),7.37(brs,2H),6.29(d,J=6.8Hz,1H),6.14(t,J=7.6Hz,1H),5.75(d,J=7.6Hz,1H),4.15-4.02(m,1H),3.98-3.73(m,3H),0.90(s,9H),0.09(s,6H)。MS-ESI[M+H] + :378.2。
Preparation of intermediate 6: 4-amino-1- ((2R, 4R, 5R) -5- ((tert-butyldimethylsilyl (oxy) methyl) -3, 3-difluoro-4- (((2S, 3aR,6S,7 aR) -3 a-methyl-6- (propyl-1-en-2-yl) -2-thiohexahydrobenzo [ d)][1,3,2]Oxathiolan-2-yl) oxy) tetrahydrofuran-2-yl) pyrimidin-2 (1H) -one
Intermediate 6 is obtained from intermediate 5 and (+) -PSI reagent by the route of intermediate 2.
1 H NMR(400MHz,DMSO-d 6 )δ7.73(d,J=8.0Hz,1H),6.34(t,J=8.0Hz,1H),6.21(d,J=8.0Hz,1H),5.36-5.24(m,1H),5.04(s,1H),4.87(s,1H),4.55-4.43(m,1H),4.17-4.06(m,1H),4.05-3.86(m,2H),2.66-2.52(m,1H),2.30-2.05(m,2H),2.02-1.80(m,3H),1.80-1.62(m,7H),0.92(s,9H),0.13(s,3H),0.12(s,3H)。
31 P NMR(162MHz,CDCl 3 )δ103.0。MS-ESI[M+H] + :624.2。
Preparation of intermediate 7: n4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine
To a pyridine solution (20 mL) of 2' -deoxy-2 ',2' -difluorocytidine hydrochloride (2.00 g,6.67 mmol) at 0℃was added dropwise trimethylchlorosilane (3.61 g,33.4 mmol), and the resulting mixture was stirred at 0-5℃for 2 hours. Simultaneously, carbonyldiimidazole (1.19 g,7.33 mmol) was added in portions to a solution of 2-propylpentanoic acid (1.06 g,7.33 mmol) in acetonitrile (20 mL), and the resulting mixture was stirred at 25℃for 2 hours. Then, the resulting acetonitrile mixture was added dropwise to the pyridine mixture at the previous 0℃and the resulting mixture was stirred at 45℃for 16 hours. Ethanol (20 mL) was added to the above mixture, and the mixture was stirred at 45℃for 0.5 hour. Water (20 mL) was then added to the above mixture, and the mixture was stirred at 45℃for 1 hour. The resulting reaction solution was dried by spin-drying and then diluted with water (50 mL). The pH of the above mixture was adjusted to 2-3 with 2N aqueous hydrochloric acid and extracted twice with ethyl acetate (50 mL each). The organic phases were combined, washed twice with water (50 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (2:3) to give a white solid (1.20 g).
1 H NMR(400MHz,DMSO-d 6 )δ11.06(brs,1H),8.25(d,J=8.0Hz,1H),7.33(d,J=8.0Hz,1H),6.32(d,J=8.0Hz,1H),6.17(t,J=8.0Hz,1H),5.32-5.25(m,1H),4.25-4.10(m,1H),3.97-3.75(m,2H),3.70-3.58(m,1H),2.70-2.55(m,1H),1.56-1.42(m,2H),1.38-1.10(m,6H),0.85(t,J=7.6Hz,6H)。MS-ESI[M+H] + :390.2。
Preparation of intermediate 8: n4- (2-propylpentanoyl) -5 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine
Intermediate 8 is obtained from intermediate 7 by the route of intermediate 1.
1 H NMR(400MHz,CDCl 3 )δ9.07(brs,1H),8.27(brs,1H),7.51(d,J=8.0Hz,1H),6.45-6.30(m,1H),4.46-4.33(m,1H),4.12-4.00(m,2H),3.91(dd,J=8.0,2.4Hz,1H),2.25-2.12(m,1H),1.71-1.60(m,2H),1.56-1.42(m,2H),1.40-1.25(m,4H),1.05-0.85(m,15H),0.13(s,6H)。
Preparation of intermediate 9: n- (1- ((2R, 4R, 5R) -5- ((tert-butyldimethylsilyl) oxy) methyl) -3, 3-difluoro-4- (((2S, 3aR,6S,7 aR) -3 a-methyl-6- (propyl-1-en-2-yl) -2-thiohexahydrobenzo [ d)][1,3,2]Oxathiolan-2-yl) oxy-tetrahydrofuran-2-yl) -2-oxo-1, 2-dihydropyrimidin-4-yl) -2-propylpentanoic acid amide
Intermediate 9 is obtained from intermediate 8 and (+) -PSI reagent by the route of intermediate 2.
1 H NMR(400MHz,CDCl 3 )δ9.63(brs,1H),8.29-8.15(m,1H),7.60-7.48(m,1H),6.48-6.39(m,1H),5.40-5.25(m,1H),5.04(s,1H),4.88(s,1H),4.55-4.45(m,1H),4.28-4.05(m,1H),4.00-3.88(m,2H),2.65-2.55(m,1H),2.46-2.35(m,1H),2.33-2.23(m,1H),2.20-1.80(m,5H),1.79-1.60(m,8H),1.58-1.45(m,2H),1.45-1.25(m,4H),0.98-0.85(m,15H),0.15(d,J=4.0Hz,6H)。
Preparation of intermediate 10: n6-bis (4-methoxyphenyl) benzyl-2-chloro-5 '-O-tert-butyldimethylsilyl 3' -bis (4-methoxyphenyl) benzyl-2 '-deoxy-2' -fluoro-beta-adenosine
To a pyridine solution (10 mL) of intermediate 1 (1.53 g,3.67 mmol) was added bis (4-methoxyphenyl) benzyl chloride (1.49 g,4.40 mmol), and the resulting mixture was stirred at 25℃for 2 hours. Bis (4-methoxyphenyl) benzyl chloride (497 mg,1.47 mmol) and 1, 8-diazabicyclo undec-7-ene (669 mg,4.40 mmol) were then added in sequence and the resulting mixture stirred at 20-25℃for 16 hours. Bis (4-methoxyphenyl) benzyl chloride (1.49 g,4.40 mmol) and 1, 8-diazabicyclo undec-7-ene (669 mg,4.40 mmol) were added in sequence and the resulting mixture stirred at 20-25℃for 24 hours. The reaction mixture was concentrated and diluted with ethyl acetate (100 mL). The organic phase was washed twice with saturated brine (100 mL each time) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether in ethyl acetate (5:1) to give a white solid (2.60 g).
1 H NMR(400MHz,DMSO-d 6 )δ8.00(s,1H),7.95(s,1H),7.50-7.10(m,18H),6.90-6.78(m,8H),6.33-6.20(m,1H),4.46-4.30(m,2H),4.25-4.15(m,1H),3.76-3.68(m,12H),3.55-3.49(m,2H),0.76(s,9H),-0.08(s,3H),-0.11(s,3H)。
Preparation of intermediate 11: n6-bis (4-methoxyphenyl) benzyl-2-chloro-3 ' -bis (4-methoxyphenyl) benzyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine
Tetrabutylammonium fluoride (1.33 g,5.10 mmol) was added to a solution of intermediate 10 (2.60 g,2.55 mmol) in tetrahydrofuran (15 mL) and the resulting mixture stirred at 25℃for 1 hour. The reaction mixture was concentrated and diluted with ethyl acetate (30 mL). The organic phase was washed with water (30 mL) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (2:3) to give a white solid (1.20 g).
1 H NMR(400MHz,DMSO-d 6 )δ8.17(d,J=2.4Hz,1H),7.97(s,1H),7.50-7.10(m,18H),6.90-6.78(m,8H),6.30-6.18(m,1H),4.86(t,J=6.0Hz,1H),4.30-4.10(m,3H),3.76-3.68(m,12H),3.49-3.30(m,2H)。
Preparation of intermediate 12: n6-bis (4-methoxyphenyl) benzyl-2-chloro-5 ' -O-tert-butyldimethylsilyl 3' -bis (4-methoxyphenyl) benzyl-2 ' -deoxyadenosine
To a pyridine solution (10 mL) of intermediate 3 (1.80 g,4.51 mmol) was added successively bis (4-methoxyphenyl) benzyl chloride (3.81 g,11.3 mmol) and 1, 8-diazabicycloundec-7-ene (743 mg,11.3 mmol), and the resulting mixture was stirred at 20-25℃for 16 hours. The reaction mixture was concentrated and poured into water (100 mL) and extracted twice with ethyl acetate (50 mL each). The organic phases were combined, washed twice with saturated brine (50 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether/ethyl acetate (3:7) to give a white solid (2.50 g).
1 H NMR(400MHz,DMSO-d 6 )δ8.19(s,1H),7.83(s,1H),7.50-7.10(m,18H),6.95-6.78(m,8H),6.28-6.18(m,1H),4.36-4.28(m,1H),4.00-3.88(m,1H),3.80-3.65(m,12H),3.50-3.41(m,1H),3.40-3.30(m,1H),2.33-2.25(m,1H),1.88-1.76(m,1H),0.71(s,9H),-0.14(s,6H)。
Preparation of intermediate 13: n6-bis (4-methoxyphenyl) benzyl-2-chloro-3 '-bis (4-methoxyphenyl) benzyl-2' -deoxyadenosine
Intermediate 13 is obtained from intermediate 12 by a route using intermediate 11.
1 H NMR(400MHz,DMSO-d 6 )δ8.32(s,1H),7.87(s,1H),7.50-7.10(m,18H),6.95-6.77(m,8H),6.24(t,J=6.8Hz,1H),4.83(brs,1H),4.39-4.28(m,1H),3.85-3.65(m,13H),3.22-3.12(m,1H),2.35-2.21(m,1H),1.88-1.70(m,1H)。
Preparation of intermediate 14:3' -5' -di-O-tert-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine
To a solution of 2' -deoxy-2 ',2' -difluorocytidine hydrochloride (3.00 g,10.0 mmol) in DMF (50 mL) was added imidazole (3.41 g,50.0 mmol) and tert-butyldimethylchlorosilane (4.53 g,30.0 mmol) in this order, and the resulting mixture was stirred at 20-25℃for 2 hours and then at 60℃for 16 hours. The reaction mixture was poured into a mixture of ethyl acetate (100 mL) and water (100 mL). The organic phase was separated, washed with water (100 mL) and then with saturated brine (100 mL), and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica eluting with methylene chloride: methanol (9:2) to give a white solid (2.70 g).
1 H NMR(400MHz,DMSO-d 6 )δ7.53(d,J=7.6Hz,1H),7.40(brs,2H),6.17(t,J=8.0Hz,1H),5.78(d,J=7.6Hz,1H),4.40-4.22(m,1H),3.98-3.83(m,2H),3.78-3.70(m,1H),1.00-0.75(m,18H),0.15-0.00(m,12H)。
Preparation of intermediate 15:3 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine
Intermediate 14 (1.00 g,2.04 mmol) was dissolved in a mixture of tetrahydrofuran solution (10 mL), trifluoroacetic acid (5 mL) and water (5 mL) at 0deg.C, and stirred at 0-5deg.C for 2 hours. The pH of the reaction mixture was adjusted to about 9 with saturated aqueous sodium bicarbonate solution, and then poured into a mixture of ethyl acetate (50 mL) and water (50 mL). The organic phase was separated, washed with saturated brine (50 mL) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with ethyl acetate: methanol (17:3) to give a white solid (620 mg).
1 H NMR(400MHz,DMSO-d 6 )δ7.67(d,J=7.6Hz,1H),7.50-7.35(m,2H),6.14(t,J=8.0Hz,1H),5.79(d,J=7.6Hz,1H),5.26(d,J=7.6Hz,1H),4.40-4.22(m,1H),3.86-3.72(m,2H),3.68-3.50(m,1H),0.88(s,9H),0.11(s,3H),0.10(s,3H)。MS-ESI[M+H] + :378.2。
Preparation of intermediate 16: n4- (2-propyl-pentanoyl) -3' -5' -di-O-tert-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine
To a solution of 2-propylpentanoic acid (322 mg,2.24 mmol) in acetonitrile (5 mL) at 25℃was added carbonyldiimidazole (803 mg,2.24 mmol), and the resulting mixture was stirred at 25℃for 2 hours. The resulting mixture was added dropwise to a solution of intermediate 14 (1.0 g,2.03 mmol) in pyridine (10 ml) at 0deg.C. The resulting mixture was stirred at 45℃for 16 hours, and the resulting reaction solution was dried by spinning, diluted with water (20 mL), and extracted twice with ethyl acetate (30 mL each time). The organic phases were combined, washed twice with water (50 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column on silica gel with petroleum ether: ethyl acetate (1:1) to give a white solid (430 mg).
1 H NMR(400MHz,CDCl 3 )δ8.09(d,J=7.6Hz,1H),7.49(d,J=7.6Hz,1H),6.33(d,J=10.0Hz,1H),4.39-4.29(m,1H),4.02(d,J=12.0Hz,1H),3.96(d,J=7.6Hz,1H),3.81(d,J=11.2Hz,1H),2.50-2.30(m,1H),1.49-1.44(m,2H),1.33(q,J=7.6Hz,6H),0.99-0.80(m,24H),0.13(s,9H),0.10(s,3H)。MS-ESI[M+H] + :619.4。
Preparation of intermediate 17: n4- (2-propyl-pentanoyl) -3 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine
Intermediate 17 is obtained from intermediate 16 by a route employing intermediate 15.
1 H NMR(400MHz,CDCl 3 )δ8.38(s,1H),8.00(d,J=7.6Hz,1H),7.50(d,J=7.6Hz,1H),6.25(t,J=7.6Hz,1H),4.49-4.40(m,1H),4.08(d,J=12.4Hz,1H),4.02-3.90(m,1H),3.88-3.75(m,1H),2.51(s,1H),2.40-2.30(m,1H),1.75-1.55(m,2H), 1.52-1.41(m,2H),1.38-1.28(m,4H),0.95-0.85(m,15H),0.14(d,J=2.8Hz,6H)。MS-ESI[M+H] + :504.2。
Intermediate 18:5 '-O-tert-butyldimethylsilyl-2' -tert-butyldimethylsilyl-adenosine, and
intermediate 19:5 '-O-tert-butyldimethylsilyl-3' -tert-butyldimethylsilyl-adenosine
To a solution of adenosine (3.35 g,12.5 mmol) in tetrahydrofuran (40 mL) was added triethylenediamine (7.0 g,62.5 mmol) and silver nitrate (5.08 g,28.8 mmol) in this order at room temperature (10 ℃ C.). The mixture was stirred at room temperature (10 ℃) for 30min, then t-butyldimethylchlorosilane (4.71 g,30 mmol) was added and the resulting mixture was stirred at 10℃for 16 h. The reaction mixture was filtered through celite, the filtrate was quenched with water (50 mL) and then extracted twice with ethyl acetate (50 mL each). The organic phases were combined, washed twice with saturated brine (100 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether in ethyl acetate (1:1) to give intermediate 18 (1.0 g, white solid). Elution with ethyl acetate afforded intermediate 19 (4.4 g, white solid).
Intermediate 18:
1 H NMR(400MHz,CDCl 3 )δ8.35(s,1H),8.24(s,1H),6.11(d,J=4.8Hz,1H),5.72(brs,2H),4.64(t,J=4.8Hz,1H),4.31-4.26(m,1H),4.24-4.20(m,1H),4.02(dd,J=11.6,2.8Hz,1H),3.87(dd,J=11.6,2.8Hz,1H),2.76(d,J=3.6Hz,1H),0.96(s,9H),0.84(s,9H),0.18-0.11(m,6H),-0.05(s,3H),-0.12(s,3H)。
intermediate 19:
1 H NMR(400MHz,CDCl 3 )δ8.35(s,1H),8.10(s,1H),6.03(d,J=3.6Hz,1H),5.75(brs,2H),4.56(d,J=1.6Hz,1H),4.12(d,J=2.8Hz,1H),3.92(dd,J=11.6,3.6Hz,1H),3.77(dd,J=11.2,2.8Hz,1H),3.31(brs,1H),0.95(s,9H),0.90(s,9H),0.17(s,6H),0.06(s,3H),-0.01(s,3H)。
preparation of intermediate 20: n4-benzoyl-2 ' -5' -di-O-t-butyldimethylsilyl-3 ' -benzoyl-adenosine
To a solution of intermediate 18 (500 mg,1.01 mmol) and N-methylimidazole (248 mg,3.03 mmol) in dichloromethane (10 mL) was added benzoyl chloride (354.4 mg,2.52 mmol) dropwise at room temperature (25 ℃). The resulting mixture was reacted at room temperature for 16 hours. The reaction was quenched with water (20 mL), the organic phase separated and the aqueous phase extracted twice with dichloromethane (20 mL each). The organic phases were combined and washed once with saturated brine (20 mL) and then dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether in ethyl acetate (1:1) to give a white solid (400 mg).
1 H NMR(400MHz,CDCl 3 )δ9.36(s,1H),8.85(s,1H),8.48(s,1H),8.15-8.10(m,2H),8.08-8.05(m,2H),7.70-7.55(m,2H),7.55-7.47(m,4H),6.33(d,J=6.0Hz,1H),5.63-5.50(m,1H),4.89(d,J=1.2Hz,1H),4.48(d,J=2.4Hz,1H),4.05(d,J=2.4Hz,1H),4.00(d,J=2.4Hz,1H),0.99(s,9H),0.65(s,9H),0.19(d,J=4.4Hz,6H),-0.10(s,3H),-0.26(s,3H)。MS-ESI[M+H] + :704.3。
Preparation of intermediate 21: n4-benzoyl-2 '-O-tert-butyldimethylsilyl-3' -benzoyl-adenosine
Intermediate 21 is obtained from intermediate 20 by a route employing intermediate 15.
1 H NMR(500MHz,CDCl 3 )δ8.86(s,1H),8.17-8.09(m,3H),8.08-8.03(m,2H),7.66-7.60(m,2H),7.55(t,J=7.6Hz,2H),7.50(t,J=7.6Hz,2H),5.97(d,J=7.6Hz,1H),5.75(d,J=5.2Hz,1H),5.27(dd,J=7.6,5.2Hz,1H),4.51(s,1H),4.08-4.03(m,1H),3.93(d,J=12.8Hz,1H),0.62(s,9H),-0.10(s,3H),-0.43(s,3H)。MS-ESI[M+H] + :591.2。
Preparation of intermediate 22: n4-benzoyl-5 ' -O-tert-butyldimethylsilyl-3 ' -tert-butyldimethylsilyl-2 ' -benzoyl-adenosine
Intermediate 22 is obtained from intermediate 19 and benzoyl chloride by a route employing intermediate 20.
1 H NMR(400MHz,CDCl 3 )δ9.02(brs,1H),8.83(s,1H),8.41(s,1H),8.08-7.99(m,4H),7.64-7.49(m,4H),7.47-7.39(m,2H),6.54(d,J=6.0Hz,1H),5.81(t,J=5.6Hz,1H),4.84(dd,J=4.8,3.6Hz,1H),4.26(dd,J=6.0,3.2Hz,1H),4.02(dd,J=11.6,3.2Hz,1H),3.85(dd,J=11.6,2.8Hz,1H),0.95(s,9H),0.84(s,9H),0.18-0.11(m,6H),0.03--0.05(m,6H)。
Preparation of intermediate 23: n4-benzoyl-3 '-tert-butyldimethylsilyl-2' -benzoyl-adenosine
Intermediate 23 is obtained from intermediate 22 by a route employing intermediate 15.
1 H NMR(400MHz,CDCl 3 )δ9.08(brs,1H),8.84(s,1H),8.18(s,1H),8.08-7.98(m,4H),7.67-7.41(m,6H),6.32(d,J=7.6Hz,1H),6.01(dd,J=7.6,5.2Hz,1H),4.94(d,J=5.2Hz,1H),4.35(d,J=0.8Hz,1H),4.07(dd,J=13.2,1.6Hz,1H),3.84(d,J=12.4Hz,1H),0.89(s,9H),0.11(s,3H),0.00(s,3H)。
Preparation of intermediate 24: n2-isobutyryl-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -O-benzoyl-2 ' -O-t-butyldimethylsilyl-guanosine
Intermediate 24 was obtained from N2-isobutyryl-5 '-O-bis (4-methoxyphenyl) benzyl-2' -O-tert-butyldimethylsilyl-guanosine using the route of intermediate 20. MS-ESI [ M+H ]] + :874.3。
Preparation of intermediate 25: n2-isobutyryl-3' -O-benzoyl-2' -O-tert-butyldimethylsilyl-guanosine
Intermediate 24 (5.50 g,6.30 mmol) was dissolved in a mixture of glacial acetic acid (60 mL) and water (10 mL) and stirred at 25℃for 2 h. The reaction mixture was dried by spinning and then co-evaporated twice (25 mL each) with ethyl acetate. The crude product was purified by column on silica gel eluting with ethyl acetate to give a white solid (2.2 g).
1 H NMR(400MHz,CDCl 3 )δ12.15(brs,1H),8.35(s,1H),8.15-8.02(m,2H),7.74(s,1H),7.68-7.56(m,1H),7.55-7.42(m,2H),5.80(d,J=7.6Hz,1H),5.66(dd,J=5.6,1.2Hz,1H),5.52(d,J=9.2Hz,1H),4.98(dd,J=7.2,5.6Hz,1H),4.45(d,J=1.2Hz,1H),4.06-3.82(m,2H),2.78-2.62(m,1H),1.35-1.25(m,6H),0.66(s,9H),-0.09(s,3H),-0.33(s,3H)。MS-ESI[M+H] + :572.3。
Preparation of intermediate 26: n2-isobutyryl-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -tert-butyldimethylsilyl-2 ' -benzoyl-guanosine
Intermediate 26 was obtained from N2-isobutyryl-5 '-O-bis (4-methoxyphenyl) benzyl-3' -O-tert-butyldimethylsilyl-guanosine using the route of intermediate 20. MS-ESI [ M+H ]] + :874.3。
Preparation of intermediate 27: n2-isobutyryl-3 '-tert-butyldimethylsilyl-2' -benzoyl-guanosine
Intermediate 27 is obtained from intermediate 26 by a route using intermediate 25.
1 H NMR(400MHz,CDCl 3 )δ12.06(brs,1H),8.24(brs,1H),8.01-7.96(m,2H),7.83(s,1H),7.62-7.55(m,1H),7.43(d,J=8.0Hz,2H),6.13(d,J=7.2Hz,1H),5.81(dd,J=7.2,5.6Hz,1H),4.80(dd,J=4.8,1.6Hz,1H),4.30-4.25(m,1H),4.02(dd,J=12.8,2.0Hz,1H),3.78(d,J=12.4Hz,1H),2.71-2.60(m,1H),1.30-1.28(m,6H),0.83(s,9H),0.07(s,3H),0.00(s,3H)。
Preparation of intermediate 28:2'-5' -di-O-tert-butyldimethylsilyl-guanosine
Intermediate 28 is obtained from guanosine by the route of intermediate 18. MS-ESI [ M+H ]] + :513.3。
Preparation of intermediate 29:2' -5' -di-O-tert-butyldimethylsilyl-3 ' -benzoyl-guanosine
Intermediate 29 is obtained from intermediate 28 by a route that employs intermediate 20.
1 H NMR(400MHz,CDCl 3 )δ12.04(brs,1H),8.14-8.09(m,2H),7.94(s,1H),7.61(t,J=7.6Hz,1H),7.49(t,J=7.6Hz,2H),6.09(s,2H),6.00(d,J=6.0Hz,1H),5.53(s,1H),4.76(t,J=5.6Hz,1H),4.41(d,J=2.8Hz,1H),4.01(d,J=11.6Hz,1H),3.93(dd,J=11.6,2.4Hz,1H),0.97(s,9H),0.70(s,9H),0.16(d,J=3.2Hz,6H),-0.09(s,3H),-0.16(s,3H)。MS-ESI[M-H] - :615.3。
Preparation of intermediate 30:2 '-O-tert-Butyldimethylsilyl-3' -benzoyl-Guanosine (guanosine)
Intermediate 30 is obtained from intermediate 29 by a route using intermediate 15.
1 H NMR(400MHz,DMSO-d 6 )δ10.71(brs,1H),8.05(d,J=7.2Hz,3H),7.75-7.68(m,1H),7.59(t,J=7.6Hz,2H),6.51(s,2H),5.84(d,J=7.6Hz,1H),5.58(dd,J=5.6,1.6Hz,1H),5.42(t,J=5.6Hz,1H),4.96(dd,J=7.6,5.2Hz,1H),4.39-4.25(m,1H),4.20-4.00(m,1H),0.57(s,9H),-0.12(s,3H),-0.29(s,3H)。MS-ESI[M-H] - :501.3。
Preparation of intermediate 31:3'-5' -di-O-tert-butyldimethylsilyl-2 '-deoxy-2' -fluoroadenosine
Imidazole (1.81 g,26.6 mmol) and 4-dimethylaminopyridine (50 mg,0.41 mmol) were added to a solution of 2 '-deoxy-2' -fluoroadenosine (1.10 g,4.09 mmol) in DMF (10 mL), and tert-butyldimethylchlorosilane (2.16 g,14.3 mmol) was then added and the resulting mixture stirred at 15-20℃for 16 hours. The reaction mixture was poured into water (100 mL) and extracted twice with ethyl acetate (100 mL each). The organic phases were combined, washed twice with saturated brine (100 mL) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with petroleum ether: ethyl acetate (3:2) to give a white solid (1.90 g).
1 H NMR(400MHz,CDCl 3 )δ8.35(s,1H),8.12(s,1H),6.25(dd,J=16.0,2.4Hz,1H),5.66(brs,2H),5.48-5.20(m,1H),4.79-4.62(m,1H),4.18-3.95(m,2H),3.85-3.72(m,1H),1.00-0.80(m,18H),-0.05-0.18(m,12H).MS-ESI[M+H] + :498.4。
Preparation of intermediate 32:3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoroadenosine
Intermediate 31 is obtained from intermediate 31 by a route using intermediate 15.
1 H NMR(400MHz,DMSO-d 6 )δ8.37(s,1H),8.14(s,1H),7.38(brs,2H),6.22(dd,J=16.4,3.6Hz,1H),5.70-5.40(m,1H),5.29(t,J=5.6Hz,1H),4.80-4.62(m,1H),4.02-3.90(m,1H),3.79-3.68(m,1H),3.63-3.48(m,1H),0.90(s,9H),0.14(s,3H),0.13(s,3H)。
Preparation of intermediate 33: (2R, 3aS,6R,7 aS) -2- ((2R, 3R,4R, 5R) -5- (6-amino-9H-purin-9-yl) -3- ((tert-butyldimethylsilyl) oxy) -4-fluorotetrahydrofuran-2-yl) methoxy) -3 a-methyl-6- (propyl-1-en-2-yl) hexahydrobenzo [ d][1,3,2]Oxathiophospholane 2-sulfides
Intermediate 33 is obtained from intermediate 32 and the (-) -PSI reagent by the route of intermediate 2.
1 H NMR(400MHz,CDCl 3 )δ8.36(s,1H),8.05(s,1H),6.30-6.18(m,1H),5.58(brs,2H),5.50-5.22(m,1H),4.80-4.62(m,3H),4.50-4.25(m,4H),2.59-2.48(m,1H),2.33-2.20(m,1H),2.15-2.02(m,1H),1.99-1.76(m,3H),1.75-1.55(m,7H),0.93(s,9H),0.19(s,3H),0.15(s,3H)。
Preparation of intermediate 34:3'-5' -di-O-tert-butyldimethylsilyl-2 '-deoxy-2' -fluoro-guanosine
Intermediate 34 is obtained from 2 '-deoxy-2' -fluoroguanosine by the route of intermediate 31.
1H NMR(400MHz,DMSO-d 6 )δ10.69(brs,1H),7.84(s,1H),6.52(brs,2H),6.00 (d,J=16.8Hz,1H),5.36(d,J=52.0Hz,1H),4.62-4.45(m,1H),3.98-3.86(m,2H),3.74(d,J=11.6Hz,1H),0.93-0.85(m,18H),0.20-0.00(m,12H)。MS-ESI[M+H] + :515.3。
Preparation of intermediate 35:3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoro-guanosine
Intermediate 35 is obtained from intermediate 34 by a route using intermediate 15.
1 H NMR(400MHz,DMSO-d 6 )δ10.71(brs,1H),7.97(s,1H),6.56(s,2H),5.36(dd,J=52.5,5.0Hz,1H),5.20(s,1H),4.52(dd,J=12.6,6.4Hz,1H),3.96-3.89(m,1H),3.70(d,J=12.0Hz,1H),0.89(s,9H),0.12(s,6H)。MS-ESI[M+H] + :401.2。
Preparation of intermediate 36: 2-amino-9- ((2R, 3R,4R, 5R) -4- ((tert-butyldimethylsilyl) oxy) -3-fluoro-5- ((((2R, 3aS,6R,7 aS) -3 a-methyl-6- (propyl-1-en-2-yl) -2-thiohexahydrobenzo [ d)][1,3,2]Oxathiolan-2-yl) oxy) methyl) tetrahydrofuran-2-yl) -1, 9-dihydro-6H-purin-6-one
Intermediate 36 was obtained from intermediate 35 and (-) -PSI reagent in DMF using the route of intermediate 2. MS-ESI [ M+H ]] + :646.4。
Preparation of intermediate 37: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine]- (3 ',5 ') - [ N6-bis (4-methoxyphenyl) benzyl-2-chloro-3 ' -bis (4-methoxyphenyl) benzyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
To a solution of intermediate 2 (280 mg,0.422 mmol) and intermediate 11 (383 mg,0.422 mmol) in acetonitrile (3 mL) was added 1, 8-diazabicyclo undec-7-ene (192 mg,1.27 mmol) and the resulting mixture was stirred at 25℃for 1 hour. The reaction mixture was diluted with ethyl acetate (25 mL). The organic phase was washed with water (25 mL) and saturated brine (25 mL) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with ethyl acetate: methanol (7:3) to give a white solid (260 mg).
1 H NMR(400MHz,DMSO-d 6 )δ8.26(s,1H),8.08(s,1H),7.91(brs,3H),7.45-7.10(m,18H),6.95-6.76(m,8H),6.32-6.15(m,2H),5.40-5.18(m,2H),5.00-4.80(m,2H),4.40-4.02(m,3H),3.85-3.62(m,15H),0.84(s,9H),0.01(s,6H)。
Preparation of intermediate 38: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxyadenosine]- (3 ', 5') - [ N6-bis (4-methoxyphenyl) benzyl-2-chloro-3 '-bis (4-methoxyphenyl) benzyl-2' -deoxyadenosine]
Intermediate 38 was obtained by route using intermediate 37 from intermediate 4 and intermediate 13 in a mixed solution of tetrahydrofuran and acetonitrile.
1 H NMR(400MHz,DMSO-d 6 )δ8.68(s,1H),8.22(s,1H),7.82(brs,2H),7.75(s,1H),7.42(d,J=8.0Hz,2H),7.39-7.10(m,16H),6.95-6.78(m,8H),6.32-6.10(m,2H),4.90-4.80(m,1H),4.43-4.36(m,1H),4.15-4.08(m,1H),3.85-3.40(m,17H),2.50-2.20(m,4H),0.81(s,9H),0.00(s,3H),-0.02(s,3H)。
Preparation of intermediate 39: [3' -O-phosphorothioate diester-5 ' -O-t-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine]- (3 ', 5') - [3 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine]
Intermediate 39 is obtained by route using intermediate 37 from intermediate 6 and intermediate 15 in a mixed solution of tetrahydrofuran and acetonitrile.
1 H NMR(400MHz,DMSO-d 6 )δ9.58(brs,1H),7.79-7.60(m,2H),7.48-7.25(m,4H),6.28-6.05(m,2H),5.76(dd,J=7.6,1.6Hz,2H),5.00-4.85(m,1H),4.38-4.25 (m,1H),4.15-3.80(m,6H),0.95-0.80(m,18H),0.18-0.00(m,12H)。
Preparation of intermediate 40: [3' -O-phosphorothioate diester-N4- (2-propylpentanoyl) -5' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine]- (3 ', 5') - [ N4- (2-propylpentanoyl) -3 '-O-tert-butyldimethylsilyl-2' -deoxy-2 ',2' -difluorocytidine]
Intermediate 40 is obtained from intermediate 17 and intermediate 9 by a route employing intermediate 37.
1 H NMR(500MHz,DMSO-d 6 )δ11.07(brs,1H),11.02(brs,1H),8.32(d,J=7.6Hz,1H),8.24(d,J=7.6Hz,1H),7.36-7.30(m,2H),6.28-6.15(m,2H),4.97(t,J=10.4Hz,1H),4.40-4.30(m,1H),4.17(s,1H),4.08(q,J=9.1,8.5Hz,2H),4.04*3.99(m,2H),3.93(t,J=7.8Hz,1H),2.62(d,J=3.2Hz,2H),1.60-1.45(m,4H),1.42-1.27(m,4H),1.25-1.20(m,9H),1.00-0.75(m,30H),0.11(d,J=3.0Hz,6H),0.05(s,3H),0.02(s,3H)。
Preparation of intermediate 41: [3 '-O-phosphorothioate diester-2-chloro-5' -O-t-butyldimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosin ]- (3 ', 5') - [ N6-benzoyl-2 '-O-tert-butyldimethylsilyl-3' -O-benzoyl-adenosine]
Intermediate 41 is obtained from intermediate 21 and intermediate 2 by a route employing intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ11.19(brs,1H),9.10(brs,1H),8.76(s,1H),8.12(d,J=2.4Hz,1H),8.05(dd,J=7.6,2.0Hz,4H),7.89(s,2H),7.75-7.61(m,2H),7.60-7.45(m,4H),6.33(dd,J=18.0,4.0Hz,1H),6.23(d,J=7.2Hz,1H),5.77(d,J=5.2Hz,1H),5.30(dd,J=7.6,5.2Hz,1H),5.04(s,1H),4.53(s,1H),4.21(t,J=10.0Hz,1H),4.18-4.08(m,3H),4.00-3.92(m,1H),3.92-3.85(m,1H),0.85(s,9H),0.49(s,9H),0.05(s,6H),-0.12(s,3H),-0.42(s,3H)。MS-ESI[(M+2H)/2] + :543.7。
Preparation of intermediate 42: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxyadenosine]- (3 ', 5') - [ N6-benzoyl-2 '-O-tert-butyldimethylsilyl-3' -O-benzoyl-adenosine]
Intermediate 42 is obtained from intermediate 21 and intermediate 4 by a route employing intermediate 37. MS-ESI [ (M+H)] + :1067.2。
Preparation of intermediate 43: [3 '-O-phosphorothioate diester-2-chloro-5' -O-tert-butyl ]Dimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosines]- (3 ', 5') - [ N6-benzoyl-3 '-O-tert-butyldimethylsilyl-2' -O-benzoyl-adenosine]
Intermediate 43 is obtained from intermediate 23 and intermediate 2 in solution in N, N' -dimethylformamide, by route employing intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ11.23(brs,1H),8.96(s,1H),8.76(s,1H),8.11(d,J=2.4Hz,1H),8.02(d,J=7.6Hz,2H),7.88(d,J=7.6Hz,4H),7.64(dd,J=6.8,6.0Hz,2H),7.65-7.45(m,4H),6.51(d,J=5.6Hz,1H),6.32(dd,J=18.8,3.6Hz,1H),5.92(t,J=5.6Hz,1H),5.55-5.30(m,1H),5.11-4.98(m,1H),4.83(t,J=4.0Hz,1H),4.29(s,1H),4.28-4.15(m,1H),4.15-4.05(m,1H),4.00-3.89(m,2H),3.87-.80(m,1H),0.85(s,9H),0.76(s,9H),0.08(s,3H),0.04(s,6H),-0.09(s,3H)。MS-ESI[M+H] + :1085.3。
Preparation of intermediate 44: [3 '-O-phosphorothioate diester-2-chloro-5' -O-t-butyldimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosin]- (3 ', 5') - [ N2-isobutyryl-3 '-O-benzoyl-2' -O-tert-butyldimethylsilyl-guanosine]
Intermediate 44 was obtained from intermediate 2 and intermediate 25 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ12.59(brs,1H),12.11(brs,1H),8.37(s,1H),8.14(d,J=2.8Hz,1H),8.08-7.85(m,4H),7.80-7.50(m,3H),6.35(dd,J=18.0,4.0Hz,1H),5.92(d,J=8.0Hz,1H),5.68(d,J=5.2Hz,1H),5.55-5.38(m,2H),5.15-4.98(m,1H),4.52-4.40(m,1H),4.20-3.95(m,5H),2.90-2.80(m,1H),1.25-1.05(m,6H),0.87(s,9H),0.53(s,9H),0.07(s,6H),-0.13(s,3H),-0.38(s,3H)。MS-ESI[(M-2H)/2] - :532.2。
Preparation of intermediate 45: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxyadenosine]- (3 ', 5') - [ N2-isobutyryl-3 '-O-benzoyl-2' -O-tert-butyldimethylsilyl-guanosine]
Intermediate 45 was obtained from intermediate 4 and intermediate 25 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ12.76(brs,1H),12.12(brs,1H),8.37(s,1H),8.27(s,1H),8.03(d,J=7.6Hz,2H),7.81(brs,2H),7.70-7.50(m,3H),6.29(t,J=6.8Hz,1H),5.92(d,J=7.6Hz,1H),5.76-5.65(m,1H),5.55-5.45(m,1H),5.10-4.96(m,1H),4.47(s,1H),4.30-4.20(m,1H),4.10-3.75(m,3H),2.98-2.83(m,1H),2.81-2.55(m,2H),1.20-1.05(m,6H),0.84(s,9H),0.54(s,9H),0.03(s,6H),-0.11(s,3H),-0.36(s,3H)。MS-ESI[(M+2H)/2] + :525.3。
Preparation of intermediate 46: [3' -O-phosphorothioate diester-5 ' -O-t-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine]- (3 ', 5') - [ N2-isobutyryl-3 '-O-benzoyl-2' -O-tert-butyldimethylsilyl-guanosine]
Intermediate 46 was obtained from intermediate 6 and intermediate 25 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ12.49(brs,1H),12.09(brs,1H),8.39(brs,1H),8.05(d,J=7.2Hz,2H),7.78-7.50(m,4H),7.40(brs,2H),6.19(t,J=8.4Hz,1H),5.91(d,J=8.0Hz,1H),5.78(d,J=7.6Hz,1H),5.66(d,J=4.8Hz,1H),5.49-5.35(m,1H),5.10-4.90(m,1H),4.45(s,1H),4.20-3.80(m,5H),2.95-2.77(m,1H),1.12(d,J=6.8Hz,6H),0.89(s,9H),0.54(s,9H),0.15-0.05(m,6H),-0.10(s,3H),-0.37(s,3H)。MS-ESI[(M+2H)/2] + :514.3。
Preparation of intermediate 47: [3' -O-phosphorothioate diester-N4- (2-propylpentanoyl) -5' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ',2' -difluorocytidine]- (3 ', 5') - [2 '-O-tert-butyldimethylsilyl-3' -O-benzoyl-guanosine]
Intermediate 47 is obtained from intermediate 30 and intermediate 9 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ11.10(brs,1H),10.60(brs,1H),8.22(d,J=7.6Hz,1H),8.16(s,1H),8.02(d,J=7.6Hz,2H),7.68(t,J=7.6Hz,1H),7.56(t,J=7.6Hz,2H),7.35(d,J=7.6Hz,1H),6.51(brs,2H),6.23(t,J=7.6Hz,1H),5.82(d,J=7.6Hz,1H),5.67(d,J=5.2Hz,1H),5.09(dd,J=7.6,5.2Hz,1H),5.02-4.95(m,1H),4.41(d,J=3.6Hz,1H),4.18-3.92(m,4H),2.70-2.55(m,1H),1.65-1.50(m,2H),1.45-1.30(m,2H),1.28-1.15(m,4H),0.96-0.88(m,12H),0.87-0.80(m,9H),0.53(s,9H),-0.11(s,3H),-0.31(s,3H)。MS-ESI[(M-2H)/2] - :540.3。
Preparation of intermediate 48: [3 '-O-phosphorothioate diester-2-chloro-5' -O-t-butyldimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosin ]- (3 ', 5') - [ N2-isobutyryl-3 '-tert-butyldimethylsilyl-2' -benzoyl-guanosine]
Intermediate 48 was obtained from intermediate 27 and intermediate 2 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ12.53(brs,1H),12.13(brs,1H),8.36(s,1H),8.14(d,J=2.8Hz,1H),7.98-7.80(m,4H),7.64(t,J=7.6Hz,1H),7.48(t,J=8.0Hz,2H),6.36(dd,J=18.0,4.0Hz,1H),6.22(d,J=7.2Hz,1H),5.88(dd,J=7.2,5.2Hz,1H),5.60--5.43(m,1H),5.19-5.05(m,1H),4.72(d,J=4.8Hz,1H),4.29-4.22(m,1H),4.22-4.09(m,2H),4.08-3.91(m,3H),2.96-2.86(m,1H),1.18-1.09(m,6H),0.87(s,9H),0.72(s,9H),0.07(s,6H),0.00(s,3H),-0.02(s,3H)。
Preparation of intermediate 49: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxy-adenosines]- (3 ', 5') - [ N2-isobutyryl-3 '-tert-butyldimethylsilyl-2' -benzoyl-guanosine]
Intermediate 49 was obtained from intermediate 27 and intermediate 4 in-DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ12.55(brs,1H),12.14(brs,1H),8.37(s,1H),8.28(s,1H),7.94-7.76(m,4H),7.65(t,J=7.6Hz,1H),7.49(t,J=8.0Hz,2H),6.33-6.21(m,2H),5.95-5.85(m,1H),5.10-5.02(m,1H),4.75(d,J=4.8Hz,1H),4.31-4.23(m,2H),4.22-4.15(m,1H),4.14-4.09(m,1H),3.91-3.78(m,2H),2.93-2.86(m,1H),2.76-2.71(m,1H),2.68-2.60(m,1H),1.20-1.10(m,6H),0.84(s,9H),0.74(s,9H),0.07-0.00(m,9H),-0.10(s,3H)。
Preparation of intermediate 50: [3 '-O-phosphorothioate diester-2-chloro-5' -O-t-butyldimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosin]- (3 ',5 ') - [3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoroadenosine]
Intermediate 50 is obtained from intermediate 32 and intermediate 2 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ8.49(s,1H),8.18-8.08(m,2H),7.92(brs,2H),7.34(brs,2H),6.32-6.20(m,2H),5.50(dt,J=52.0,4.4Hz,1H),5.30(d,J=52.0Hz,1H),5.05-4.94(m,1H),4.78-4.65(m,1H),4.14-4.06(m,3H),3.95-3.70(m,3H),0.90-0.75(m,18H),0.10(d,J=8.8Hz,6H),0.04(s,6H)。
Preparation of intermediate 51: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxyadenosine]- (3 ',5 ') - [3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoroadenosine]
Intermediate 51 is obtained from intermediate 3 and intermediate 33 by a route employing intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ8.51(s,1H),8.25(s,1H),8.13(s,1H),7.82(brs,2H),7.31(brs,2H),6.30-6.15(m,2H),5.69(dt,J=52.4,4.0Hz,1H),5.00-4.86(m,1H),4.80-4.65(m,1H),4.19-4.00(m,3H),3.99-3.85(m,1H),3.78-3.62(m,2H),2.75-2.62(m,1H),2.60-2.50(m,1H),0.88(s,9H),0.81(s,9H),0.14(s,3H),0.11(s,3H),0.00(s,3H),-0.01(s,3H)。
Preparation of intermediate 52: [3 '-O-phosphorothioate diester-2-chloro-5' -O-t-butyldimethylsilyl-2 '-deoxy-2' -fluoro-beta-adenosin]- (3 ',5 ') - [3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoroguanosine]
Intermediate 52 was obtained from intermediate 1 and intermediate 36 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ10.64(brs,1H),8.12(s,1H),8.05(s,1H),7.93-7.82(m,3H),6.54(brs,2H),6.36-6.25(m,1H),6.00(dd,J=14.8,4.8Hz,1H),5.58-5.28(m,2H),5.15-4.98(m,1H),4.60-4.50(m,1H),4.15-4.00(m,3H),3.98-3.88(m,3H),0.86(s,18H),0.10-0.04(m,12H)。
Preparation of intermediate 53: [3' -O-phosphorothioate diester-2-chloro-5 ' -O-t-butyldimethylsilyl-2 ' -deoxyadenosine]- (3 ',5 ') - [3' -O-tert-butyldimethylsilyl-2 ' -deoxy-2 ' -fluoroguanosine]
Intermediate 53 was obtained from intermediate 3 and intermediate 36 in DMF solution, using the route of intermediate 37.
1 H NMR(400MHz,DMSO-d 6 )δ10.65(brs,1H),8.25(s,1H),8.05(s,1H),7.80(brs,2H),6.55(brs,2H),6.25(t,J=7.2Hz,1H),6.03-5.95(m,1H),5.47(d,J=52.0Hz,1H),4.97(s,1H),4.65-4.50(m,1H),4.19(s,1H),4.15-4.00(m,3H),3.99-3.85(m,1H),3.82(d,J=11.2Hz,1H),3.76-3.69(m,1H),2.75-2.60(m,1H),0.88(s,9H),0.80(s,9H),0.11(s,3H),0.10(s,3H),0.02(s,6H)。MS-ESI[(M-H)] - :875.4。
Preparation of intermediate 54: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ', 5') - [ 2-chloro-2 '-deoxy-2' -fluoro-beta-adenosine]
Intermediate 37 (260 mg,0.188 mmol) was dissolved in a mixture of tetrahydrofuran (2 mL), trifluoroacetic acid (2 mL) and water (1 mL) and stirred at 25℃for 2 hours. The reaction mixture was dried by spinning, diluted with methanol (5 mL), and the pH of the mixture was adjusted to about 8-9 with saturated aqueous sodium bicarbonate. After spin-drying, the mixture was diluted again with methanol (5 mL) and filtered. The filtrate was concentrated, and the crude product obtained was purified by a silica gel column eluting with ethyl acetate: methanol (7:3) to give a white solid (120 mg).
1 H NMR(400MHz,DMSO-d 6 )δ8.30(s,1H),8.23(s,1H),7.88(brs,4H),6.40-6.18(m,2H),6.06(d,J=5.2Hz,1H),5.45-5.12(m,2H),5.04(t,J=6.0Hz,1H),5.00- 4.85(m,1H),4.50-4.40(m,1H),4.18-3.90(m,4H),3.75-3.58(m,2H)。MS-ESI[M-H] - :682.8。
Preparation of intermediate 55: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]- (3 ',5 ') - [ 2-chloro-2 ' -deoxyadenosine]
Tetrabutylammonium fluoride (34 mg,0.132 mmol) was added to a tetrahydrofuran solution (5 mL) of intermediate 38 (150 mg,0.110 mmol), and the resulting mixture was stirred at 25℃for 3 hours. The reaction mixture was dried by spin-drying and then dissolved in a mixed solution of glacial acetic acid (2 mL) and water (0.5 mL), and the mixture was stirred at 25℃for 0.5 hours. The reaction mixture was dried by spinning, and the obtained crude product was purified by silica gel column eluting with ethyl acetate: methanol (3:1) to obtain a white solid (45 mg). MS-ESI [ M-H ]] - :647.0。
Preparation of intermediate 56: [3 '-O-phosphorothioate diester-2' -deoxy-2 ',2' -difluorocytidine]- (3 ',5 ') - [2' -deoxy-2 ',2' -difluorocytidine]
Into a solution of intermediate 39 (350 mg, 0.426 mmol) in tetrahydrofuran (5 mL)Tetrabutylammonium fluoride (329 mg,1.26 mmol) was added and the resulting mixture was stirred at 25℃for 2 hours. The reaction mixture was dried by spinning, and the crude product obtained was purified by silica gel column eluting with ethyl acetate: methanol (3:1) to give a white solid (130 mg). 1 H NMR(400MHz,DMSO-d 6 )δ7.77(d,J=7.2Hz,1H),7.65(d,J=7.6Hz,1H),7.45-7.25(m,4H),6.30(d,J=6.4Hz,1H),6.20-6.05(m,2H),5.89-5.70(m,2H),5.13(t,J=6.0Hz,1H),4.90-4.75(m,1H),4.20-4.00(m,2H),4.00-3.60(m,5H)。
Preparation of intermediate 57: [3 '-O-phosphorothioate diester-N4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine]- (3 ',5 ') - [ N4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine ]
Intermediate 57 is obtained from intermediate 40 by a route employing intermediate 56. MS-ESI [ M+H ]] + :858.2,[(M+2H)/2] + :429.1。
Preparation of intermediate 58: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ',5 ') - [ N6-benzoyl-3 ' -O-benzoyl-adenosine]
Intermediate 58 is obtained from intermediate 41 by a route employing intermediate 56. MS-ESI [ M+H ]] + :857.0。
Preparation of intermediate 59: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]- (3 ',5 ') - [ N6-benzoyl-3 ' -O-benzoyl-adenosine]
Intermediate 59 is obtained from intermediate 42 by a route employing intermediate 56. MS-ESI [ M+H ]] + :839.0。
Preparation of intermediate 60: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ',5 ') - [ N6-benzoyl-2 ' -O-benzoyl-adenosine]
To intermediate 43 (250 mg,0.23 mmol) in tetrahydrofuran (5 mL) was added triethylamine trihydrofluoride (223 mg,1.38 mmol) and the mixture was stirred at 30℃for 16 hours. The reaction mixture was neutralized to pH 7-8 with triethylamine. Spin-drying the obtained mixed solution, purifying the obtained crude product by using a silica gel column, and using ethyl acetate: methanol (8:2) gave a white solid (160 mg). MS-ESI [ M+H ]] + :857.1。
Preparation of intermediate 61: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines ]- (3 ',5 ') - [ N2-isobutyryl-3 ' -O-benzoyl-guanosine]
Intermediate 61 is obtained from intermediate 44 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ12.56(brs,1H),12.11(brs,1H),8.35-8.20(m,2H),8.10-8.00(m,2H),7.91(brs,2H),7.75-7.45(m,3H),6.38-6.18(m,1H),6.00-5.75(m,2H),5.70-5.21(m,2H),5.16-4.90(m,2H),4.50-4.36(m,1H),4.28-4.00(m,2H),3.90-3.58(m,2H),3.00-2.82(m,1H),1.36-1.05(m,6H)。MS-ESI[M-H] - :837.0。
Preparation of intermediate 62: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]- (3 ',5 ') - [ N2-isobutyryl-3 ' -O-benzoyl-guanosine]
Intermediate 62 is obtained from intermediate 45 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ12.72(brs,1H),12.13(brs,1H),8.34(s,1H),8.32(s,1H),8.03(d,J=7.6Hz,1H),7.98-7.81(m,3H),7.71-7.62(m,1H),7.60-7.43(m,1H),6.38-6.20(m,1H),5.99-5.70(m,2H),5.70-5.57(m,1H),5.40-5.28(m,1H),5.18-4.93(m,2H),4.42(s,1H),4.32-4.00(m,3H),3.75-3.52(m,2H),2.93-2.81(m,1H), 2.80-2.62(m,1H),1.20-1.05(m,6H)。MS-ESI[M+H] + :821.2。
Preparation of intermediate 63: [3 '-O-phosphorothioate diester-2' -deoxy-2 ',2' -difluorocytidine]- (3 ',5 ') - [ N2-isobutyryl-3 ' -O-benzoyl-guanosine]
Intermediate 63 is obtained from intermediate 46 by a route that employs intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ12.42(brs,1H),12.12(brs,1H),8.35(s,1H),8.08(d,J=7.6Hz,2H),7.80-7.53(m,4H),7.48-7.34(m,2H),6.24-6.12(m,1H),5.97-5.87(m,1H),5.86-5.76(m,1H),5.57(d,J=5.2Hz,1H),5.30-5.18(m,1H),5.17-5.05(m,1H),4.95-4.75(m,1H),4.41(s,1H),4.25-3.70(m,6H),2.90-2.77(m,1H),1.20-1.05(m,6H)。
Preparation of intermediate 64: [3 '-O-phosphorothioate diester-N4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine]- (3 ',5 ') - [3' -O-benzoyl-guanosine]
Intermediate 64 is obtained from intermediate 47 by a route using intermediate 56. MS-ESI [ (M+2H)/2] + :428.0。
Preparation of intermediate 65: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ',5 ') - [ N2-isobutyryl-2 ' -benzoyl-guanosine]
Intermediate 65 is obtained from intermediate 48 by a route using intermediate 56. MS-ESI [ M-H ]] - :837.0。
Preparation of intermediate 66: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines ]- (3 ',5 ') - [ N2-isobutyryl-2 ' -benzoyl-guanosine]
Intermediate 66 is obtained from intermediate 49 by route using intermediate 56. MS+ESI [ M+H ]] + :821.1。
Preparation of intermediate 67: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ', 5') - [2 '-deoxy-2' -fluoroadenosine]
Intermediate 67 is obtained from intermediate 50 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ8.47(s,1H),8.24(s,1H),8.15(s,1H),7.91(brs,2H),7.35(brs,2H),6.32-6.20(m,2H),5.88(brs,1H),5.55-5.30(m,2H),5.18-4.87(m,2H),4.53(d,J=16.4Hz,1H),4.18-4.08(m,2H),4.10-4.02(m,1H),4.01-3.92(m,1H),3.75-3.59(m,2H)。MS-ESI[M+H] + :651.0。
Preparation of intermediate 68: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]- (3 ', 5') - [2 '-deoxy-2' -fluoroadenosine]
Intermediate 68 is obtained from intermediate 51 by route using intermediate 56.
1 H NMR(400MHz,DMSO-d 6 )δ8.50(s,1H),8.34(s,1H),8.15(s,1H),7.84(brs,2H),7.31(brs,2H),6.30-6.18(m,2H),5.90(d,J=5.6Hz,1H),5.45(dt,J=52.0,3.6Hz,1H),5.12-4.90(m,2H),4.60-4.45(m,1H),4.18-4.00(m,3H),3.99-3.85(m,1H),3.65-3.50(m,2H),2.70-2.55(m,1H),2.50-2.40(m,1H)。
Preparation of intermediate 69: [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]- (3 ', 5') - [2 '-deoxy-2' -fluoroguanosine]
Intermediate 69 is obtained from intermediate 52 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ10.64(s,1H),8.24(s,1H),8.00(s,1H),7.88(brs,2H),6.54(brs,2H),6.28(dd,J=18.0,3.6Hz,1H),6.02(dd,J=16.0,3.6Hz,1H),5.80(d,J=5.4Hz,1H),5.44(d,J=4.4Hz,1H),5.30(d,J=7.6Hz,1H),5.04(d,J=6.0Hz,1H),4.95(t,J=14.8Hz,1H),4.42(dt,J=10.4,5.6Hz,1H),4.11-3.99(m,3H),3.98-3.89(m,1H),3.77-3.60(m,2H)。MS-ESI[M+H] + :667.2。
Preparation of intermediate 70: [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]- (3 ', 5') - [2 '-deoxy-2' -fluoroguanosine]
Intermediate 69 is obtained from intermediate 53 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ10.67(brs,1H),8.37(s,1H),8.04(s,1H),7.85(brs,2H),6.58(brs,2H),6.26(t,J=7.2Hz,1H),6.03(d,J=15.6Hz,1H),5.85(d,J=5.6Hz,1H),5.38(d,J=52.8Hz,1H),5.10(s,1H),4.97(s,1H),4.46(d,J=14.8Hz,1H),4.20-4.00(m,4H),4.00-3.90(m,1H),3.70-3.50(m,2H),2.78-2.60(m,1H)。MS-ESI[M-H] - :647.2。
Preparation of intermediate 71: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-N6-benzoyl-3' -benzoyl-adenosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines ]
To a solution of intermediate 58 (95 mg,0.11 mmol) and (-) -PSI reagent (150 mg,0.33 mmol) in DMF (10 mL) was added 1, 8-diazabicyclo undec-7-ene (255 mg,1.66 mmol) and the resulting mixture stirred at 25℃for 1 hour. The reaction mixture was dried by spinning and reprecipitated with ethyl acetate (20 mL), and the obtained solid was slurried with ethyl acetate (20 mL), filtered and dried to obtain a crude product (200 mg), which was directly used as the next reaction. MS-ESI [ M+2+H ]] + :937.0。
Preparation of intermediate 72: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-N6-benzoyl-3' -O-benzeneFormyl-adenosines]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]
To a solution of intermediate 59 (210 mg,0.25 mmol) and (-) -PSI reagent (335 mg,0.75 mmol) in DMF (10 mL) was added 1, 8-diazabicyclo undec-7-ene (570 mg,3.75 mmol) and the resulting mixture was stirred at 25℃for 1 hour. The reaction mixture was dried by spinning and reprecipitated with ethyl acetate (20 mL), and the obtained solid was slurried with ethyl acetate (20 mL), filtered and dried to obtain a crude product. The crude product was purified by preparative liquid chromatography (10 mM ammonium bicarbonate as additive) and lyophilized to give a white solid (15.8 mg) which was used directly in the next reaction. MS-ESI [ M+H ]] + :917.0。
Preparation of intermediate 73: (3 ',3 ') -cyclo-bis- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosin-e ]- [3 '-O-phosphorothioate diester-N6-benzoyl-2' -O-benzoyl-adenosine]
Intermediate 73 is obtained from intermediate 60 and the (-) -PSI reagent by a route employing intermediate 71. MS-ESI [ M-H ]] - :932.8。
Intermediate 74: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-3' -O-benzoyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Isomer 1 and intermediate 75: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-3' -O-benzoyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Preparation of isomer 2
Intermediate 74 and intermediate 75 are obtained from intermediate 61 and the (-) -PSI reagent using the route of intermediate 72.
Intermediate 74: MS-ESI [ M-H ]] - 844.8, retention time: 4.96min;
intermediate 75: MS-ESI [ M-H ]] - 844.9, retention time: 5.20min. Analysis LCMS: agilent1100+G1946D LCMS,4.6x150mm Waters XBridge C18 3.5 μm analytical column with mobile phase A of 10mM NH 4 HCO 3 Aqueous solution, B is acetonitrile, flow rate 1.0ml/min, dual wavelength ultraviolet absorption monitoring of 214 and 254nm, gradient elution: 0-0.1min,5% B;0.1-8min,5-95% B;8-15min,95% B.
Intermediate 76: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-N2-isobutyryl-3' -O-benzoyl-guanosine ]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]Isomer 1, and
intermediate 77: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-N2-isobutyryl-3' -O-benzoyl-guanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]Preparation of isomer 2
Intermediate 76 and intermediate 77 are obtained from intermediate 62 and the (-) -PSI reagent using the route of intermediate 72.
Intermediate 76: MS-ESI [ M-H ]] - 897.2, retention time: 1.69min;
intermediate 77: MS-ESI [ M-H ]] - 897.2, retention time: 1.74min. Analysis LCMS: agilent 1100+G1946D LCMS,4.6x50mm Waters XBridge C18 3.5 μm analytical column with mobile phase A of 10mM NH 4 HCO 3 Aqueous solution, B is acetonitrile, flow rate 1.8min/min, dual-wavelength ultraviolet absorption monitoring of 214 and 254nm, gradient elution: 0-0.1min,5%B;0.1-2.5min,5-95%B;2.5-5min,95%B。
Intermediate 78Is prepared from the following steps: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-N2-isobutyryl-3' -O-benzoyl-guanosine]- [3 '-O-phosphorothioate diester-2' -deoxy-2 ',2' -difluorocytidine]
Intermediate 78 is obtained from intermediate 63 and the (-) -PSI reagent, following the route of intermediate 72. Intermediate 78: MS-ESI [ (M-2H)/2] - :437.0。
Intermediate 79Is prepared from the following steps: (2 ', 3') -cyclo- [2 '-O-phosphorothioate diester-3' -O-benzoyl-guanosine ]- [3 '-O-phosphorothioate diester-N4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine]
Intermediate 79 is obtained from intermediate 64 and the (-) -PSI reagent by route employing intermediate 72. MS-ESI [ M-H ]] - :931.0。
Intermediate 80: (3 ', 3') -cyclo- [3 '-O-phosphorothioate diester-N2-isobutyryl-2' -benzoyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Isomer 1, and
intermediate 81: (3 ', 3') -cyclo- [3 '-O-phosphorothioate diester-N2-isobutyryl-2' -benzoyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Isomer 2
Intermediates 80 and 81 are routes taken from intermediate 65 and (-) -PSI reagent using intermediate 72. Intermediate 80: MS-ESI [ M-H ]] - 915.0, retention time: 1.50min;
intermediate 81: MS-ESI [ M-H ]] - 915.0, retention time: 1.53min. Analysis LCMS: agilent1100+G1946D LCMS,4.6x50mm Waters XBridge C18 3.5 μm analytical column with mobile phase A of 10mM NH 4 HCO 3 Aqueous solution, B is acetonitrile, flow rate 1.8ml/min, dual wavelength ultraviolet absorption monitoring of 214 and 254nm, gradient elution: 0-0.2min,5% B;0.2-1.5min,5-95% B;1.5-3min,95% B.
Preparation of intermediate 82 : (3 ', 3') -cyclo- [3 '-O-phosphorothioate diester-N2-isobutyryl-2' -benzoyl-guanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]
Intermediate 82 follows the route of intermediate 72 and is derived from intermediate 66 and the (-) -PSI reagent. MS-ESI [ M-H ]] - :896.8。
Preparation of intermediate 83: 2-chloro-5 '-O-tert-butyldimethylsilyl-3' -O-benzoyl-2 '-deoxy-2' -fluoro-beta-adenosine
Intermediate 83 is obtained from intermediate 1 by route using intermediate 20.
1 H NMR(400MHz,DMSO-d 6 )δ8.21(d,J=2.4Hz,1H),8.08(d,J=7.2Hz,2H),7.95(brs,2H),7.72(t,J=7.6Hz,1H),7.58(t,J=7.6Hz,2H),6.50(dd,J=16.6,4.0Hz,1H),5.80-5.77(m,1H),5.75-5.66(m,1H),4.32(q,J=4.8Hz,1H),3.97(d,J=4.4Hz,2H),0.86(s,9H),0.06(s,6H)。
Preparation of intermediate 84: 2-chloro-3 ' -O-benzoyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine
Intermediate 84 is obtained from intermediate 83 by a route using intermediate 60.
1 H NMR(400MHz,DMSO-d 6 )δ8.40(d,J=2.4Hz,1H),8.12(d,J=7.6Hz,2H),7.99(brs,2H),7.74(t,J=7.2Hz,1H),7.60(t,J=7.6Hz,2H),6.55(dd,J=18.2,3.6 Hz,1H),5.81-5.78(m,1H),5.77-5.66(m,1H),4.35(q,J=4.4Hz,1H),3.89-3.80(m,2H)。
Preparation of intermediate 85: n- (9- ((2R, 3R,4R, 5R) -5- ((bis (4-methoxyphenyl) (phenyl) methoxy) methyl) -4- ((tert-butyldimethylsilyl) oxy) -3- ((2S, 3aR,6S,7 aR) -3 a-methyl-6- (prop-1-en-2-yl) -2-thiohexahydrobenzo [ d)][1,3,2]Oxathiolan-2-yl) oxy) tetrahydrofuran-2-yl) -6-oxy-6, 9-dihydro-1H-purin-2-yl) isobutyramide
Intermediate 85 is obtained from N2-isobutyryl-5 '-O-bis (4-methoxyphenyl) benzyl-3' -O-tert-butyldimethylsilyl-guanosine and the (+) -PSI reagent using the route of intermediate 2.
1 H NMR(400MHz,DMSO-d 6 )δ12.11(s,1H),11.62(s,1H),8.14(s,1H),7.38(d,J=8.0Hz,2H),7.31-7.22(m,7H),6.86(d,J=8.4Hz,4H),6.00(d,J=6.8Hz,1H),5.67-5.59(m,1H),4.89(s,1H),4.75(s,1H),4.42(d,J=4.0Hz,1H),4.22(d,J=12.0Hz,1H),4.06-4.02(m,1H),3.73(s,6H),3.41-3.35(m,2H),3.24(dd,J=12.0Hz,1H),2.82-2.73(m,1H),2.07(d,J=12.0Hz,1H),1.93-1.83(m,2H),1.76-1.65(mf,6H),1.54(s,3H),1.12(d,J=6.8Hz,6H),0.83(s,9H),0.08(s,3H),0.04(s,3H)。
Preparation of intermediate 86: [2' -O-Rp-phosphorothioate diester-N2-isobutyryl-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -O-t-butyldimethylsilyl-guanosine]- (2 ',5 ') - [ 2-chloro-3 ' -O-benzoyl-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
Intermediate 86 is obtained from intermediate 84 and intermediate 85 in DMF solution, using the route of intermediate 37.
MS-ESI[M+H] + :1255.0。
Preparation of intermediate 87: [2' -O-Rp-phosphorothioate diester-N2-isobutyryl-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -O-t-butyldimethylsilyl-guanosine]- (2 ', 5') - [ 2-chloro-2 '-deoxy-2' -fluoro-beta-adenosine]
Intermediate 86 (1.0 g,0.8 mmol) was added to a mixed solution of tetrahydrofuran/water/methanol (10 mL) at 8:4:1, cooled to 0deg.C, lithium hydroxide monohydrate (54 mg,1.6 mmol) was added and the resulting mixture stirred at 0deg.C for 20min. The reaction mixture was poured into water (50 mL) and extracted twice with ethyl acetate (50 mL each). The organic phases were combined, washed twice with saturated brine (100 mL each) and dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by column chromatography on silica gel eluting with ethyl acetate: methanol (10:1) to give a white solid (733 mg).
1 H NMR(400MHz,DMSO-d 6 )δ12.02(s,1H),11.67(s,1H),8.21(t,J=5.2Hz,1H),8.14(s,1H),7.84(s,2H),7.37(d,J=7.6Hz,2H),7.29-7.14(m,7H),6.87-6.77(m,4H),6.23(dd,J=13.9,4.5Hz,1H),6.08-5.98(m,2H),5.49-5.38(m,1H),5.23-5.04(m,1H),4.66-4.59(m,1H),4.35-4.24(m,1H),3.99-3.95(m,1H),3.77-3.71(m,2H),3.71-3.63(m,7H),3.22-3.00(m,2H),2.79-2.68(m,1H),1.12-1.07(m,6H),0.82(s,9H),0.13(d,J=13.4Hz,6H)。
Preparation of intermediate 88: o- ((2R, 3R,4S, 5R) -5- (6-amino-2-chloro-9H-purin-9-yl) -4-fluoro-3- ((2R, 3aS,6R,7 aS) -3 a-methyl-6- (prop-1-en-2-yl) -2-thiohexahydrobenzo [ d ] ][1,3,2]Oxathiolan-2-yl) methyl) -O- ((2R, 3R,4R, 5R) -5- ((bis (4-methoxyphenyl) (phenyl) methoxy) methyl) -4- ((tert-butyldimethylsilyl) oxy) -2- (2-isobutylamino-6-oxy-1, 6-dihydro-9H-purin-9-yl) tetrahydrofuran-3-yl) - (R) -phosphorothioate diester
Intermediate 88 is a crude product prepared from intermediate 87 and (-) -PSI reagent in tetrahydrofuran, using the route of intermediate 2, directly used in the next reaction. MS-ESI [ M+H-DMTr] + :1095.0。
Preparation of intermediate 89: o- ((2R, 3R,4S, 5R) -5- (6-amino-2-chloro-9H-purin-9-yl) -4-fluoro-3- ((2R, 3aS,6R,7 aS) -3 a-methyl-6- (prop-1-en-2-yl) -2-thiohexahydrobenzo [ d ]][1,3,2]Oxathiolan-2-yl) methyl) -O- ((2R, 3R,4R, 5R) -4- ((tert-butyldimethylsilyl) oxy) -2- (2-isobutylamino-6-oxy-1, 6-dihydro-9H-purin-9-yl) tetrahydrofuran-3-yl) - (R) -thiophosphoric acid diester
Intermediate 89 is obtained from intermediate 88 by a route employing intermediate 25.
MS-ESI[M+H] + :1095.0。
Preparation of intermediate 90: (2 ', 3') -cyclo- (Rp, sp) - [2 '-O-phosphorothioate diester-N2-isobutyryl-3' -O-t-butyldimethylsilyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]
Intermediate 89 (90 mg,0.082 mmol) was dissolved in anhydrous DMF (7 mL), 4A molecular sieve (90 mg) was added and dried under stirring at room temperature (28 ℃ C.) under nitrogen for 0.5 h. DBU (75.0 mg,0.492 mmol) was then added via syringe and stirred at room temperature (28 ℃ C.) for 0.5 hours, then filtered through celite, and the filtrate was spin-dried to give the crude intermediate 90 (100 mg, crude) which was used directly in the next reaction.
MS-ESI[M+H] + :926.8。
Preparation of intermediate 91: (2R, 3R,4S, 5R) -5- (6-amino-2-chloro-9H-purin-9-yl) -2- (((((((2R, 3R,4R, 5R) -5- ((bis (4-methoxyphenyl) (phenyl) methoxy) methyl) oxy) 4- ((tert-butyldimethylsilyl) oxy) -2- (2-isobutylamino-6-oxy-1, 6-dihydro-9H-purin-9-yl) tetrah-ran-3-yl) oxy) (2-cyanoethoxy) phosphoryl) oxy) methyl) -4-fluorotetrahydrofuran-3-yl benzoate
Intermediate 84 (500 mg,1.23 mmol) and (2R, 3R,4R, 5R) -5- ((bis (4-methoxyphenyl) (phenyl) methoxy) methyl) -4- ((tert-butyldimethylsilyl) oxy) -2- (2-isobutylamino-6-oxo-1, 6-dihydro-9H-purin-9-yl) tetrahydrofuran-3-yl (2-cyanoethyl) diisopropylphosphoramide (1.78 g,1.84 mmol) were dissolved in a mixed solution of acetonitrile/tetrahydrofuran (8 mL/8 mL), and 4A molecular sieve (500 mg) was added and stirred at room temperature (28 ℃ C.) under nitrogen for 0.5 hours. Then a syringe was used to add tetrazole in acetonitrile (8.17 ml,0.45 m); after stirring at room temperature (28 ℃) for 2 hours, 70% t-butanol peroxide (520.9 mg,4.05mmol,0.544 mL) was added, and after stirring at room temperature (28 ℃) for 20min, it was quenched with 50% aqueous sodium thiosulfate pentahydrate (5 mL); the mixture was filtered through celite, the filtrate was diluted with ethyl acetate (30 mL) and washed with water (20 mL, twice) and the organic phase was dried over anhydrous sodium sulfate. The crude product obtained by concentration was purified by silica gel column eluting with petroleum ether: ethyl acetate (10:1-1:10) to give a mixture (1.9 g) which was used directly in the next step.
MS-ESI[M+H] + :1296.0。
Preparation of intermediate 92: [2' -O-phosphodiester-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -O-t-butyldimethylsilyl-guanosine]- (2 ', 5') - [ 2-chloro-2 '-deoxy-2' -fluoro-beta-adenosine]
To intermediate 91 (1 g,0.775 mmol) was added 33% methanolic methylamine solution (8 mL) and stirred at room temperature for 3 hours. The reaction mixture was spin-dried and purified on a C18 reverse phase column, eluting with water (0.1% ammonium bicarbonate) acetonitrile (3:2), and freeze-dried to give a pink solid (550 mg).
1 H NMR(400MHz,DMSO-d 6 )δ10.65(s,1H),8.18(d,J=2.0Hz,1H),7.93-7.77(m,2H),7.38-7.15(m,10H),7.13-6.92(m,2H),6.90-6.77(m,4H),6.49-6.20(m,4H),5.92(d,J=5.6Hz,1H),5.26-5.09(m,2H),4.61-4.24(m,3H),3.98-3.82(m,3H),3.79-3.67(m,7H),0.79(s,9H),0.14-0.02(m,6H)。MS-ESI[M+H] + :1064.8。
Preparation of intermediate 93: [2' -O-phosphodiester-5 ' -O-bis (4-methoxyphenyl) benzyl-3 ' -O-t-butyldimethylsilyl-guanosine]- (2 ',5 ') - [ 2-chloro-3 ' -O-phosphite monoester-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
Diphenyl phosphite (137 mg,0.47 mmol) was added intermediate 92 (250 mg,0.235 mmol), DBU (143 mg,0.94 mmol) and a solution of 4A molecular sieve in pyridine (8 mL) under nitrogen at 0 ℃ and the resulting mixture stirred at room temperature for 2 hours. The reaction solution was filtered and dried by spin-drying, and was used as it is for the next reaction. MS-ESI [ M+H ]] + :1129.8。
Preparation of intermediate 94: [2 '-O-phosphodiester-3' -O-t-butyldimethylsilyl-guanosine]- (2 ',5 ') - [ 2-chloro-3 ' -O-phosphite monoester-2 ' -deoxy-2 ' -fluoro-beta-adenosine ]
Intermediate 94 is obtained from intermediate 93 by a route employing intermediate 25.
1 H NMR(400MHz,DMSO-d 6 )δ8.26(s,1H),7.99(s,1H),7.88(s,2H),6.55(s,2H),6.27(dd,J=17.2,4.0Hz,1H),6.00-5.70(m,3H),5.44-5.21(m,1H),5.09-4.98(m,1H),4.87-4.71(m,1H),4.47(d,J=4.0Hz,1H),4.07-3.97(m,1H),3.91-3.73(m,3H),3.56-3.49(m,5H),0.89(s,9H),0.14(s,6H)。 31 P NMR(162MHz,DMSO-d 6 )δ1.72,-1.58。MS-ESI[M+H] + :826.8。
Preparation of intermediate 95: (2 ', 3') -cyclo- [2 '-O-phosphodiester-3' -O-t-butyldimethylsilyl-guanosine]- [3' -O-phosphodiester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
To intermediate 94 (55 mg,0.067 mmol) in pyridine (15 mL) was added pivaloyl chloride (48 mg,0.40 mmol) at 0deg.C, and the resulting reaction was stirred at room temperature for 3 hours. Then acetonitrile (0) of elemental iodine (26 mg,0.10 mmol) was added thereto.5 mL) and water (0.1 mL), and the resulting reaction solution was stirred at room temperature for 16 hours. The reaction mixture was quenched with 10% aqueous sodium thiosulfate (5 mL) and then dried by spin-drying, followed by purification on a C18 reverse phase column, eluting with water (0.1% ammonium bicarbonate in acetonitrile (4:1)), and freeze-drying to give a white solid (20 mg). MS-ESI [ M+H ]] + :825.0。
Preparation of intermediate 96: (2 ', 3') -cyclo- [2 '-O-Rp-phosphorothioate diester-N2-isobutyryl-3' -O-t-butyldimethylsilyl-guanosine]- [3' -O-phosphodiester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
Intermediate 96 was prepared from [2' -O-phosphodiester-N2-isobutyryl-3 ' -O-tert-butyldimethylsilyl-guanosine ] - (2 ',5 ') - [ N6-benzoyl-2-chloro-3 ' -O-phosphomonoester-2 ' -deoxy-2 ' -fluoro-beta-adenosine ] using the route of intermediate 95.
1 H NMR(400MHz,DMSO-d 6 )δ12.92(s,1H),12.12(s,1H),11.54(s,1H),8.55(d,J=2.0Hz,1H),8.23(s,1H),8.06(d,J=7.2Hz,2H),7.71-7.61(m,1H),7.59-7.51(m,3H),6.47(dd,J=21.6,2.4Hz,1H),5.97-5.74(m,2H),5.60-5.40(m,1H),5.12-5.00(m,1H),4.41(d,J=3.2Hz,1H),4.27-4.15(m,1H),4.13-3.94(m,5H),2.97-2.84(m,1H),1.65-1.54(m,1H),1.09(d,J=7.2Hz,3H),0.98(d,J=6.8Hz,3H),0.93(s,9H),0.20(d,J=8.6Hz,6H)。
Preparation of intermediate 97: (2 ', 3') -cyclo- [2 '-O-phosphodiester-3' -O-t-butyldimethylsilyl-guanosine]- [3' -O-Rp-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]
Intermediate 97 is prepared by the reaction of intermediate 92 with (+) -PSI reagent, deprotection and cyclization reaction, and purification by preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C18 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,20% B, 2-14min,20-40% B, 14-14.2min,40-95% B, 14.2-18min,95% B. The compound retention time 12.5 min).
1 H NMR(400MHz,DMSO-d 6 )δ10.59(s,1H),8.18(d,J=2.4Hz,1H),8.11(s,1H),7.45–6.98(m,5H),6.33–6.23(m,1H),5.82(d,J=8.6Hz,1H),5.42(s,1H),5.32–5.23(m,2H),4.33(d,J=4.0Hz,1H),4.19–3.70(m,9H),0.91(s,9H),0.15(s,6H)。 19 F NMR(376MHz,DMSO)δ-196.51(s)。 31 P NMR(162MHz,DMSO)δ53.76(s),-0.49(s)。MS-ESI[M+H] + :841.0。
Preparation of intermediate 98: (2 ', 3') -cyclo- (Rp, rp) - [2 '-O-phosphorothioate diester-N2-isobutyryl-3' -O-t-butyldimethylsilyl-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]
Intermediate 98 is obtained by deprotection and cyclization reactions using the synthetic route of intermediate 90 from intermediate 87 and the (+) -PSI reagent.
1 H NMR(400MHz,DMSO-d6)δ12.41(brs,1H),11.90(s,1H),11.31(s,1H),8.35(d,J=2.0Hz,1H),8.03(s,1H),7.85(d,J=7.2Hz,2H),7.45(t,J=7.2Hz,1H),7.35(t,J=7.6Hz,2H),6.88(t,J=48Hz,6H),6.25(dd,J=20.0,4.0Hz,1H),5.65(d,J=8.4Hz,1H),5.59-5.46(m,1H),5.32(d,J=50.0Hz,1H),5.02(t,J=11.2Hz,1H),4.15(d,J=4.0Hz,1H),4.07-4.01(m,1H),3.97-3.64(m,5H),2.74-2.62(m,1H),0.86(d,J=6.4Hz,3H),0.81-0.66(m,12H),-0.01(d,J=9.6Hz,6H)。MS+ESI--[M+H]:1030.8。
Example 1: (3 ',3 ') -cyclo- (Rp, rp) -di- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosine ]1.5-ammonium-0.5-1, 8-diazabicyclo undec-7-ene-salts
To a solution of intermediate 54 (120 mg,0.150 mmol) and (-) -PSI reagent (201 mg,0.450 mmol) in DMF (8 mL) was added 1, 8-diazabicyclo undec-7-ene (348 mg,2.25 mmol) and the resulting mixture stirred at 25℃for 1 hour. The reaction mixture was dried by spinning and reprecipitated with ethyl acetate (20 mL). The solid was collected and the crude product obtained was purified by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-3min,0-3% B, 3-14min,3-30% B, 14-14.3min,30-95% B, 14.3-20min,95% B. The compound retention time 13.5 min) to give a white solid (20 mg) after lyophilization.
1 H NMR(400MHz,DMSO-d 6 )δ8.37(s,1H),8.22(s,1H),7.90(brs,4H),7.12(brs,6H),6.33-6.20(m,2H),5.56-5.30(m,2H),5.20-5.00(m,2H),4.22-3.85(m,6H),3.55- 3.40(m,2H),3.35-3.15(m,1H),2.70-2.55(m,1H),1.96-1.85(m,1H),1.73-1.52(m,3H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.47。MS-ESI[M-H] - :760.9。
Example 2: (3 ', 3') -cyclo- (Rp, rp) -di- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]
Example 2 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column with mobile phase A of 0.05% aqueous formic acid, B of 0.05% formic acid in acetonitrile, flow rates 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution of 0-3min,0-5% B, 3-14min,5-50% B, 14-14.3min,50-95% B, 14.3-20min,95% B. This compound retention time is 12 min) following the route of example 1, reaction from intermediate 55 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ8.53(s,1H),8.38(s,1H),7.84(brs,4H),6.33-6.18(m,2H),5.20-4.75(m,2H),4.30-3.75(m,6H),3.00-2.70(m,2H),2.70-2.50(m,2H). 31 P NMR(162MHz,DMSO-d 6 )δ53.68,52.69。MS-ESI[M-H] - :724.9。
Example 3: (3 ', 3') -cyclo- (Rp, rp) -bis- [3 '-O-phosphorothioate diester-2' -deoxy-2 ',2' -difluorocytidine]
Example 3 was prepared by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column with mobile phase A of 0.05% aqueous formic acid, B of 0.05% formic acid in acetonitrile, flow rates 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution of 0-3min,0-5% B, 3-14min,5-30% B, 14-14.3min,30-95% B, 14.3-20min,95% B. This compound retention time 18 min) following the route of example 1, reaction with intermediate 56 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ9.20-8.80(m,2H),8.31(d,J=6.4Hz,1H),8.28-8.00(m,2H),7.90(d,J=6.0Hz,1H),6.20-6.05(m,3H),6.00(d,J=6.4Hz,1H),4.95-4.68(m,2H),4.30-4.05(m,4H),3.90-3.70(m,2H)。 31 P NMR(162MHz,DMSO- d 6 )δ53.27,52.67。MS-ESI[M+H] + :683.0。
Example 4: (3 ',3 ') -cyclo- (Rp, rp) -bis- [3' -O-phosphorothioate diester-N4- (2-propane)Acyl) -2' -deoxy-2 ',2' -difluorocytidine]Diammonium salt
Example 4 was obtained by purification using preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column with mobile phase A of 10mM ammonium bicarbonate aqueous solution, B of acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-3min,0-35% B;3-14min,35-70% B;14-14.3min,70-95% B;14.3-20min,95% B. The compound retention time is 14 min.) following the procedure of example 1, after reaction with intermediate 57 and (-) -PSI reagent.
1H NMR(400MHz,DMSO-d 6 )δ11.05(s,1H),11.00(s,1H),8.39(d,J=7.6Hz,1H),8.16(d,J=7.6Hz,1H),7.40(d,J=7.6Hz,1H),7.34(d,J=7.6Hz,1H),7.12(t,J=52.0Hz,8H),6.30-6.18(m,2H),5.02-4.75(m,2H),4.35-4.10(m,4H),3.83(t,J=11.2Hz,2H),2.70-2.52(m,2H),1.62-1.45(m,4H),1.40-1.17(m,12H),0.85(t,J=7.2Hz,12H)。31P NMR(162MHz,DMSO)δ54.54,53.29。MS-ESI[M+H] + :935.1。
Example 5: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-adenosines]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
To intermediate 71 (200 mg,0.214 mmol) was added 7M methanolic ammonia (2 mL) and the resulting mixture was stirred at 25℃for 6 hours. And spin-drying the reaction mixture to obtain a crude product. The crude product obtained was purified by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A0.05% aqueous formic acid, B0.05% aqueous formic acid in acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3min,0-5% B, 3-14min,5-30% B, 14-14.3 min,30-95% B, 14.3-20min,95% B. This compound retention time 14 min) to give a white solid (4.0 mg) after lyophilization.
1 H NMR(500MHz,DMSO-d 6 )δ8.63(s,1H),8.23(s,1H),8.17(s,1H),7.92(brs,2H),7.29(brs,2H),6.33-6.28(m,1H),6.12(d,J=8.5Hz,1H),5.40-5.15(m,2H),5.05-4.95(m,1H),4.55(t,J=3.5Hz,1H),4.36-4.24(m,2H),4.19(s,1H),4.10-3.95(m,2H),3.90-3.78(m,1H),3.72(d,J=12.0Hz,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.08,48.81。MS-ESI[M-H] - :724.8。
Example 6: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-adenosines]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]Diammonium salt
Example 6 was obtained by purification by preparative liquid chromatography (10 mM ammonium bicarbonate as additive) following the route of example 5, after reaction of intermediate 72 with 28% ammonia. Analysis LCMS: agilent1100+G1946D LCMS,4.6x50mm Waters XBridge C18 3.5 μm analytical column, mobile phase A is 10mM ammonium bicarbonate aqueous solution, mobile phase B is acetonitrile, flow rate is 1.8ml/min,214 and 254nm dual wavelength ultraviolet absorption monitoring, gradient elution: 0-1.5min,5-95% B;1.5-3min,95% B. The retention time of this compound was 0.35min.
1 H NMR(400MHz,DMSO-d 6 )δ8.61(s,1H),8.40(s,1H),8.17(s,1H),7.86(brs,2H),7.41-7.06(m,10H),6.30-6.18(m,1H),6.09(d,J=8.4Hz,1H),5.42-5.30(m,2H),5.30-5.20(m,1H),4.27(d,J=4.4Hz,1H),4.20-4.05(m,2H),4.04-3.90(m,2H),3.72-3.62(m,2H),2.85-2.55(m,2H)。31P NMR(162MHz,DMSO-d 6 )δ56.62,53.65。MS-ESI[M-H] - :707.0。
Example 7: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-thiophosphorusAcid diester-adenosines]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Example 7 was obtained by purification by preparative liquid chromatography (10 mM ammonium bicarbonate as additive) following the route of example 5, by reaction of intermediate 73 with 28% aqueous ammonia. Analysis LCMS: agilent1100+G1946D LCMS,4.6x150mm Waters XBridge C18 3.5 μm analytical column, mobile phase A is 10mM ammonium bicarbonate aqueous solution, mobile phase B is acetonitrile, flow rate is 1ml/min,214 and 254nm dual wavelength ultraviolet absorption monitoring, gradient elution: 0-8min,5-95% B;8-15min,95% B. The retention time of this compound was 4.1min.
1 H NMR(400MHz,DMSO-d 6 )δ8.62(s,1H),8.23(s,1H),8.19(s,1H),7.95(brs,2H),7.65(brs,2H),7.14(t,J=52.0Hz,8H),6.30-6.22(m,1H),6.12(d,J=8.4Hz,1H),5.50-5.32(m,2H),5.30-5.15(m,1H),5.08-4.92(m,1H),4.30-4.00(m,5H),3.95-3.86(m,1H),3.68(d,J=12.0Hz,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ57.14,54.13。MS-ESI[(M-2H)/2] - :362.0。
Example 8: (2 ',3 ') -cyclo- (Sp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt, and
example 9: (2 ',3 ') -cyclo- (Sp, sp) - [2' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Examples 8 and 9 were obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3min,0-2% B, 3-14min,2-37% B, 14-14.3min,37-95% B, 14.3-20min,95% B. Compound 8 retention time 13min, compound 9 retention time 15 min) following the route of example 5, reaction from intermediate 74 and 28% aqueous ammonia.
Example 8:
1 H NMR(400MHz,DMSO-d 6 )δ10.62(brs,1H),8.19(s,1H),8.11(s,1H),7.94(brs,2H),7.10(t,J=50.8Hz,8H),6.63(brs,2H),6.25(dd,J=24.0,2.8Hz,1H),5.85(d,J=8.8Hz,1H),5.50-5.15(m,4H),4.30-4.13(m,2H),4.12-3.92(m,4H),3.72(d,J=12.0Hz,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ56.84,54.05。MS-ESI[M-H] - :741.0。
example 9:
1 H NMR(400MHz,DMSO-d 6 )δ10.67(brs,1H),8.23(s,1H),8.08(s,1H),7.93(brs,2H),7.10(t,J=51.2Hz,8H),6.67(brs,2H),6.30(dd,J=18.8,3.6Hz,1H),5.77(d,J=8.0Hz,1H),5.58-5.38(m,1H),5.02-4.92(m,1H),4.86-4.78(m,1H),4.69-4.60(m,1H),4.33-3.75(m,7H)。 31 P NMR(162MHz,DMSO-d 6 )δ59.03,54.74。MS-ESI[M-H] - :740.9。
example 10: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Example 10 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3% B, 3-14min,3-38% B, 14-14.3min,38-95% B, 14.3-20min,95% B. This compound retention time 13 min) following the route of example 5, reaction from intermediate 75 and 28% aqueous ammonia. Example 10 was also obtained by purification by preparative liquid chromatography following the route of example 25, deprotected via intermediate 98. Two different synthetic routes give exactly the same product.
1 H NMR(400MHz,DMSO-d 6 )δ10.63(brs,1H),8.22(s,1H),8.18(s,1H),7.93(brs,2H),7.12(t,J=51.2Hz,8H),6.69(brs,2H),6.28(dd,J=24.0,2.4Hz,1H),5.88(d,J=8.4Hz,1H),5.42-4.80(m,4H),4.45(d,J=4.0Hz,1H),4.39-4.10(m,3H),4.08-3.94(m,1H),3.93-3.70(m,2H)。 31 P NMR(162MHz,DMSO-d 6 )δ54.07,50.13。MS-ESI[M-H] - :741.0。
Example 11: (2 ',3 ') -cyclo- (Sp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]Diammonium salt
Example 11 was obtained by purification by preparative liquid chromatography (10 mM ammonium bicarbonate as additive) following the route of example 5, by reaction of intermediate 76 with 7M methanolic ammonia. Analysis of LCNS: agilent1100+G1946D LCMS,4.6x150mm Waters XBridge C18 3.5 μm analytical column, mobile phase A is 10mM ammonium bicarbonate aqueous solution, mobile phase B is acetonitrile, flow rate is 1ml/min,214 and 254nm dual wavelength ultraviolet absorption monitoring, gradient elution: 0-8min,5-95% B;8-15min,95% B. The retention time of this compound was 3.3min.
1 H NMR(400MHz,DMSO-d 6 )δ10.63(brs,1H),8.39(s,1H),8.21(s,1H),7.83(brs,2H),7.10(t,J=50.8Hz,8H),6.67(brs,2H),6.30-6.18(m,1H),5.87(d,J=8.4Hz,1H),5.35-5.20(m,2H),4.28(d,J=4.0Hz,1H),4.21-3.92(m,4H),3.80-3.60(m,2H),2.92-2.70(m,1H),2.69-2.55(m,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ56.96,54.34。MS-ESI[M-H] - :722.9。
Example 12: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]Diammonium salt
Example 12 was obtained by purification by preparative liquid chromatography (10 mM ammonium bicarbonate as additive) following the route of example 5, by reaction of intermediate 77 with 7M methanolic ammonia. Analysis LCMS: agilent1100+G1946D LCMS,4.6x150mm Waters XBridge C18 3.5 μm analytical column, mobile phase A is 10mM ammonium bicarbonate aqueous solution, mobile phase B is acetonitrile, flow rate is 1ml/min,214 and 254nm dual wavelength ultraviolet absorption monitoring, gradient elution: 0-8min,5-95% B;8-15min,95% B. The retention time of this compound was 3.3min.
1 H NMR(400MHz,DMSO-d 6 )δ10.61(brs,1H),8.39(s,1H),8.20(s,1H),7.82(brs,2H),7.11(t,J=51.2Hz,8H),6.70(brs,2H),6.30-6.18(m,1H),5.87(d,J=8.4Hz,1H),5.25-5.13(m,2H),4.96(s,1H),4.50(d,J=4.4Hz,1H),4.31-3.71(m,6H),3.03- 2.82(m,1H),2.65-2.50(m,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ54.26,49.51。MS-ESI[M-H] - :722.8。
Example 13: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3 '-O-phosphorothioate diester-2' -deoxy-2 ',2' -difluorocytidine]Diammonium salt
Example 13 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3% B, 3-14min,3-25% B, 14-14.3min,25-95% B, 14.3-20min,95% B. This compound retention time is 2 min) following the route of example 5, reaction from intermediate 78 and 28% aqueous ammonia.
1 H NMR(400MHz,DMSO-d 6 )δ8.16(s,1H),7.60-6.70(m,11H),6.58(brs,2H),6.35-6.20(m,1H),5.90-5.70(m,2H),5.22-5.10(m,1H),5.00-4.80(m,2H),4.52(s,1H),4.32-4.15(m,2H),4.11(s,1H),4.00-3.82(m,2H),3.78-3.65(m,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.24,48.05。MS-ESI[(M-2H)/2] - :350.0。
Example 14: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3 '-O-phosphorothioate diester-N4- (2-propylpentanoyl) -2' -deoxy-2 ',2' -difluorocytidine]Diammonium salt
Example 14 and was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3min,0-10% B, 3-14min,10-55% B, 14-14.3min,55-95% B, 14.3-20min,95% B. This compound retention time 11 min) following the route of example 5, reaction from intermediate 79 and 7M methanolic ammonia solution.
1 H NMR(400MHz,DMSO-d 6 )δ11.07(brs,1H),10.62(s,1H),8.68-7.97(m,2H),7.40-6.97(m,9H),6.59(brs,2H),6.34-6.20(m,1H),5.90-5.70(m,1H),5.47-5.09(m,2H),5.05-4.74(m,1H),4.29(d,J=4.4Hz,1H),4.20(d,J=9.6Hz,1H),4.13-3.99(m,2H),3.95-3.80(m,1H),3.85-3.65(m,1H),2.70-2.55(m,1H),1.60-1.46(m,2H),1.45-1.10(m,6H),0.86(t,J=7.2Hz,6H)。 31 P NMR(162MHz,DMSO-d 6 )δ56.16,53.39。MS-ESI[M-H] - :827.0。
Example 15: (3 ',3 ') -cyclo- (Sp, rp) - [3' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Example 15 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3% B, 3-14min,3-33% B, 14-14.3min,33-95% B, 14.3-20min,95% B. This compound retention time is 11 min) following the route of example 5, reaction from intermediate 80 and 7M methanolic ammonia solution.
1 H NMR(400MHz,DMSO-d 6 )δ8.20(d,J=2.8Hz,1H),8.04(s,1H),8.03-6.87(m,10H),6.60(brs,2H),6.22(dd,J=24.0,2.0Hz,1H),5.85(d,J=8.8Hz,1H),5.51-5.15(m,3H),4.35-3.89(m,6H),3.72(d,J=12.4Hz,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ56.65,54.03。MS-ESI[M-H] - :741.0。
Example 16: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Example 16 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3% B, 3-14min,3-33% B, 14-14.3min,33-95% B, 14.3-20min,95% B. This compound retention time is 13 min) following the route of example 5, reaction from intermediate 81 and 7M methanolic ammonia solution.
1 H NMR(400MHz,DMSO-d 6 )δ10.63(brs,1H),8.22(s,1H),8.09(s,1H),7.94(brs,2H),7.38-6.94(m,8H),6.65(brs,2H),6.28(dd,J=24.0,2.4Hz,1H),5.87(d,J=8.8Hz,1H),5.42-5.11(m,3H),5.00(s,1H),4.56-4.42(m,1H),4.39-4.21(m,2H),4.13(s,1H),4.05-3.94(m,1H),3.89-3.71(m,2H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.97,49.08。MS-ESI[M-H] - :740.9。
Example 17: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-guanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxy-adenosines]Diammonium salt
Example 17 was obtained by purification using preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-3% B, 3-14min,3-33% B, 14-14.3min,33-95% B, 14.3-20min,95% B. This compound retention time 11 min) following the procedure of example 5 after reaction from intermediate 82 and 7M methanolic ammonia solution.
1 H NMR(400MHz,DMSO-d 6 )δ10.59(brs,1H),8.41(s,1H),8.11(s,1H),7.85(brs,2H),7.15(brs,8H),6.66(brs,2H),6.25(dd,J=12.8,6.0Hz,1H),5.85(d,J=8.8Hz,1H),5.35-5.11(m,2H),4.91(s,1H),4.59-4.50(m,1H),4.31-3.71(m,6H),3.03-2.87(m,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.46,47.84。MS-ESI[M-H] - :740.9。
Example 18: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-2 ' -deoxy-2 ' -fluoroadenosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]-0.5-ammonium-1.5-1, 8-diazabicyclo undec-7-ene-salt
Example 18 was obtained by purification using preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column with mobile phase A of 10mM ammonium bicarbonate aqueous solution, B of acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-3min,0-5% B;3-14min,5-33% B;14-14.3min,33-95% B;14.3-20min,95% B. The compound retention time is 11 min) following the procedure of example 1, after reaction with intermediate 67 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ9.62(brs,2H),8.39(s,1H),8.26(d,J=2.0Hz,1H),8.18(s,1H),7.36(brs,2H),7.15(t,J=52.0Hz,2H),6.31-6.20(m,2H),5.46-5.25(m,2H),5.15-5.00(m,1H),4.95-4.83(m,1H),4.42-4.25(m,1H),4.10-3.98(m,2H),3.80-3.60(m,2H),3.60-3.40(m,6H),3.30-3.20(m,3H),2.70-2.55(m,3H),1.96-1.85(m,3H),1.73-1.50(m,9H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.37,52.10。MS-ESI[M+H] + :729.0。
Example 19: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-2 ' -deoxy-2 ' -fluoroadenosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]0.5-1, 8-diazabicyclo undec-7-ene-salts
Example 19 was obtained by purification by preparative liquid chromatography (Gilson 281 preparative HPLC,19x250mm Welch 10 μm preparative column with mobile phase A of 0.05% aqueous formic acid, B of 0.05% formic acid in acetonitrile, flow rates 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution of 0-3min,0-10% B, 3-14min,10-50% B, 14-14.3min,50-95% B, 14.3-20min,95% B. This compound retention time is 10 min) following the procedure of example 1, after reaction with intermediate 68 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ9.56(s,1H),8.60(s,1H),8.50-8.35(m,1H),8.28(s,1H),7.85(brs,2H),6.40-6.20(m,2H),5.70-5.40(m,1H),5.18-4.80(m,2H),4.60-3.95(m,6H),3.65-3.45(m,2H),3.30-3.20(m,1H),2.85-2.71(m,1H),2.70-2.55(m,2H),1.96-1.85(m,1H),1.73-1.50(m,3H)。 31 P NMR(162MHz,DMSO-d 6 )δ54.78,54.00。MS-ESI[M+H] + :710.8。
Example 20: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-2 ' -deoxy-2 ' -fluoroguanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Example 20 was obtained by purification by preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C18 10 μm preparative column with mobile phase A of 10mM ammonium bicarbonate aqueous solution, B of acetonitrile, flow rates 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,5% B, 2-9min,5-15% B, 9-19min,15-25% B, 19-19.5min,25-95% B, 19.5-22.5min,95% B. This compound retention time 9 min) following the procedure of example 1 after reaction with intermediate 69 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ10.69(brs,1H),8.26(s,1H),7.98(s,1H),7.92(brs,2H),7.15(t,J=52.0Hz,6H),6.64(brs,2H),6.26(dd,J=17.2,3.6Hz,1H),6.08(d,J=16.8Hz,1H),5.43-5.35(m,1H),5.31-5.22(m,1H),5.18-5.02(m,1H),4.99-4.85(m,1H),4.42-4.30(m,1H),4.26(d,J=8.8Hz,1H),4.15-4.00(m,2H),3.90-3.70(m,2H)。 31 P NMR(162MHz,DMSO-d 6 )δ54.20,53.14。MS-ESI[M-H] - :743.2。
Example 21: (3 ',3 ') -cyclo- (Rp, rp) - [3' -O-phosphorothioate diester-2 ' -deoxy-2 ' -fluoroguanosine]- [3 '-O-phosphorothioate diester-2-chloro-2' -deoxyadenosine]Diammonium salt
Example 21 was obtained by purification by preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C18 10 μm preparative column with mobile phase A of 10mM ammonium bicarbonate aqueous solution, B of acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,5% B, 2-10min,5-20% B, 10-10.6min,20-95% B, 10.6-12.6min,95% B. This compound retention time 7.3 min) following the procedure of example 1 after reaction with intermediate 70 and (-) -PSI reagent.
1 H NMR(400MHz,DMSO-d 6 )δ10.67(brs,1H),8.49(s,1H),7.92(s,1H),7.82(brs,2H),7.14(t,J=52.0Hz,6H),6.62(s,2H),6.25(dd,J=6.4,4.8Hz,1H),6.05(d,J=16.8Hz,1H),5.44(dd,J=52.0,4.0Hz,1H),5.05-4.85(m,2H),4.33(d,J=12.4Hz, 1H),4.23(d,J=9.2Hz,1H),4.10-3.92(m,2H),3.98-3.50(m,2H),2.78-2.54(m,2H)。 31 P NMR(162MHz,DMSO-d 6 )δ52.56,52.51。MS-ESI[M-H] - :725.2。
Example 22: (2 ',3 ') -cyclo- [2' -O-phosphodiester-guanosine]- [3' -O-phosphodiester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosine]
Example 22 was obtained by purification using the route of example 9, after reaction of intermediate 95 and ammonium fluoride in methanol solution, using preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C1810 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,5% B, 2-15.6min,5-20% B, 15.6-15.8min,20-95% B, 15.8-18min,95% B. This compound retention time 6.6 min).
1 H NMR(400MHz,DMSO-d 6 )δ10.77(s,1H),8.20(d,J=2.8Hz,1H),7.99(s,1H),7.93(brs,2H),6.71(brs,2H),6.33(dd,J=24.0,2.4Hz,1H),5.90(d,J=8.0Hz,1H),5.40(d,J=49.6Hz,1H),5.20–5.00(m,2H),4.42-4.30(m,2H),4.20-3.80(m,6H)。 31 P NMR(162MHz,DMSO-d 6 )δ1.61,-0.07。MS-ESI[M+H] + :710.9。
Example 23: (2 ',3 ') -cyclo- [2' -O-Rp-phosphorothioate diester-guanosine]- [3' -O-phosphodiester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosine]Diammonium salt
To intermediate 96 (75 mg,0.074 mmol) was added 33% methanolic methylamine (3 mL) and stirred at room temperature for 3 hours. After the reaction mixture was dried by spin, ammonium fluoride (82 mg,2.2 mmol) and methanol (3 mL) were added thereto and stirred at 60℃for 16 hours. Purification by preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C1810 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rates 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,5% B, 2-18.9min,5-15% B, 18.9-19.4min,15-95% B, 19.4-22.4min,95% B. Retention time of the compound 8.5 min) gave a white solid (30.3 mg) after lyophilization.
1 H NMR(400MHz,DMSO-d 6 )δ10.60(s,1H),8.21(d,J=2.8Hz,1H),8.01(s,1H),7.91(s,2H),7.35-6.94(m,6H),6.63(s,2H),6.29(dd,J=24.4,1.6Hz,1H),5.86(d,J=8.0Hz,1H),5.42-5.17(m,2H),5.09-4.90(m,2H),4.48(d,J=4.0Hz,1H),4.36(dd,J=10.8,5.2Hz,1H),4.28-4.17(m,1H),4.09(s,1H),3.98-3.73(m,3H)。 31 P NMR(162MHz,DMSO-d 6 )δ48.92,-2.42。MS-ESI[M+H] + :726.8。
Example 24: (2 ',3 ') -cyclo- [2' -O-phosphodiester-guanosine]- [3' -O-Rp-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]
Example 24 was obtained by purification using the route of example 9, after reaction of intermediate 97 and ammonium fluoride in methanol solution, using preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C18 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,20% B, 2-14min,20-40% B, 14-14.2min,40-95% B, 14.2-18min,95% B. This compound retention time 2 min).
1 H NMR(400MHz,DMSO-d 6 )δ10.58(s,1H),8.19(d,J=2.8Hz,1H),8.04–7.82(m,2H),7.37–7.00(m,4H),6.73–6.53(m,1H),6.27(dd,J=24.0,2.4Hz,1H),5.83(d,J=8.3Hz,1H),5.44–5.19(m,2H),5.18–5.06(m,1H),5.04–4.95(m,1H),4.36–4.27(m,2H),4.12(s,1H),4.09–3.96(m,2H),3.79–3.65(m,2H). 31 P NMR(162MHz,DMSO-d 6 )δ53.89(s),-1.20(s)。 19 F NMR(376MHz,DMSO-d 6 )δ-195.45(s)。MS-ESI[M+H] + :726。
Example 25: (2 ',3 ') -cyclo- (Rp, sp) - [2' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-chloro-2 ' -deoxy-2 ' -fluoro-beta-adenosines]Diammonium salt
Intermediate 90 (100 mg, crude) was dissolved in 7M methanolic ammonia (3 mL), stirred at room temperature (28 ℃ C.) for 4 hours and then dried by spinning. The residue was dissolved in methanol (1 mL), ammonium fluoride (60.5 mg,1.63 mmol) was added, heated (60 ℃ C.) and stirred for 20 hours, after completion of the reaction by LCMS and HPLC monitoring, the reaction mixture was purified by preparative liquid chromatography (Gilson 281 preparation of a HPLC,19x250mm Waters XBridge C18 10 μm preparative column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25mL/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution: 0-2min,5% B, 2-18.9min,5-15% B, 18.9-19.4min,15-95% B, 19.4-22.4min,95% B. Retention time 10 min) to give a white solid (21 mg) after lyophilization.
1 H NMR(400MHz,DMSO-d 6 )δ10.71(s,1H),8.44(s,1H),8.23(s,1H),7.92(s,2H),7.11(t,J=52Hz,6H),6.56(s,2H),6.29(d,J=28Hz,1H),5.95(d,J=8.4Hz,1H),5.35(d,J=50.4Hz,1H),5.20(t,J=11.8Hz,1H),5.11-5.05(m,1H),4.48(d,J=3.6Hz,1H),4.39-4.28(m,2H),4.14(s,1H),4.02-3.96(m,1H),3.88-3.83(m,1H),3.81-3.76(m,1H)。 31 P NMR(162MHz,DMSO-d 6 )δ53.30,50.07。 19 F NMR(376MHz,DMSO-d 6 )δ-196.87。MS-ESI[M+H] + :742.8。
Example 26: (2 ',3 ') -cyclo- (Rp, rp) - [2' -O-phosphorothioate diester-guanosine]- [3' -O-phosphorothioate diester-2-methylamino-2 ' -deoxy-2 ' -fluoro-beta-adenosin]Diammonium salt
Example 26 was prepared by deprotecting the by-product from the reaction of intermediate 98 with an ethanol solution of methylamine, using the route of example 25, using preparative liquid chromatography (Gilson 281 preparation HPLC,19x250mm Waters XBridge C18 10 μm preparation column, mobile phase A10 mM ammonium bicarbonate aqueous solution, B acetonitrile, flow rate 25ml/min, dual wavelength UV absorption monitoring at 214 and 254nm, gradient elution 0-2min,5% B, 2-15.6min,5-15% B, 15.6-15.8min,15-95% B, 15.8-18min,95% B. This compound retention time 14.1 min), and freeze drying to give a white solid.
1 H NMR(400MHz,DMSO-d6)δ10.69(brs,1H),8.27(s,1H),8.02(s,1H),7.34-6.97(m,3H),6.31(brs,1H),6.28(d,J=2.4Hz,1H),5.90(d,J=0.8Hz,1H),5.44-5.15(m,3H),4.44(d,J=0.4Hz,1H),4.35-4.27(m,1H),4.24-4.12(m,2H),4.03-3.94(m,1H),3.93-3.79(m,2H),2.85(s,3H)。 31 P NMR(162MHz,DMSO-d 6 )δ54.02,51.06。MS+ESI--[M+H]:738.0。
Active examples
Example 1: activation of THP-1 cells to secrete IFN- β by Compounds of the invention
THP-1 cells are a human monocytic leukemia cell line with a STING phenotype of the HAQ type, R71H-G230A-R293Q. This experiment uses the Human IFN-beta DuoSet ELISA kit from R & D (R & D, cat# DY 814-05) and DuoSet ELISA Ancillary Reagent Kit 2 (R & D, cat# DY 008) to evaluate the activation of THP-1 cells secreting IFN- β by representative compounds of the invention.
When recovering THP-1 cells (ATCC # TIB-202), the cell cryopreservation tube was rapidly shaken in a 37℃water bath to allow them to thaw within 1 min. The thawed cell suspension was mixed with RPMI1640 medium (Hyclone, cat# SH 30027.01) containing 10% FBS (Lifetechnology, cat# 10099-141) and centrifuged at 1000 rpm for 5min, and the supernatant was discarded. Cell pellet was resuspended in 5mL of complete medium (RPMI 1640 medium containing 10% FBS) and placed in a bottom area of 25cm 2 Placing the cell culture flask of (C) in 37 ℃ and 5% CO 2 Culturing in a cell culture box with 95% humidity. And when the cell confluency rate reaches about 80%, carrying out cell passage. When the cells are passaged, all the cells in the culture flask are transferred into a 15mL centrifuge tube,centrifuging at 1000 rpm for 5min, and discarding supernatant. 5mL of fresh complete medium was added to resuspend the cell pellet, 1mL was placed at bottom area 25cm 2 In the cell culture flask of (2), 4mL of fresh complete medium was fed for further culture. The cells were plated when the confluency rate reached again about 80%. With reference to the cell passage method at plating, 1/5 of the cell suspension was kept for continued culture, and the remaining 4/5 of the cell suspension was placed in a 15mL centrifuge tube. The old medium was discarded by centrifugation, the cells were washed once with RPMI1640 medium (without serum addition), and the supernatant was removed by centrifugation. Cells were resuspended in RPMI1640 medium (without serum addition). Cell viability was measured by trypan blue exclusion and plated at a cell viability of 95% or higher. The density was 1.1X10 using RPMI1640 medium (without serum addition) 6 180. Mu.L of the cell suspension was added to a 96-well cell culture plate well (NUNC Co., cat# 167008) to give a cell density of 2X 10 in the culture plate 5 Living cells/well.
First, a DMSO stock of 10mM compound was serially diluted with DMSO (Sigma, cat#d2650) at a dilution factor of 3.16 to the 5 th concentration, and a DMSO control containing no compound at the 6 th concentration was set. Then, the DMSO solutions containing the compounds at different concentrations were diluted 10-fold with PBS to give DMSO contents of 10% in each concentration of the compound solutions. Finally, 20 mu L of the solution is added into a corresponding cell culture plate, the initial concentration of the compound is 100 mu M, the dilution multiple of the adjacent concentration is 3.16 times, and the DMSO content in the cell culture plate is 1%. The cell plates were placed in a cell incubator for continued culture for 24 hours.
ELISA detection was performed with reference to the R & D System, cat#DY814-05 instructions.
ELISA plate coating: the capture antibody (mouse anti-human IFN-. Beta.capture antibody ParT# 844508) was diluted with PBS (R & D System Cat#DY006) to a working concentration, 100. Mu.L of capture antibody working solution was added to a 96-well ELISA plate, sealed plates and left to incubate overnight at room temperature. The capture antibody working solution was discarded and the cell plates were washed 3 times with wash buffer (0.05% Tween-20 in PBS pH 7.2-7.4,R&D System Cat#WA126), 400. Mu.L of wash buffer was used per well, wash buffer was removed well for each wash, and the plate was inverted on clean paper for the last wash to remove wash buffer completely. mu.L of blocking buffer (pH 7.2-7.4,R&D System Cat#DY995 in 1% BSA in PBS) was added to each well, and the wells were incubated at room temperature for 1-2 hours. The above washing steps were repeated, preparing each plate for sample addition.
Sample detection: mu.L of sample or standard (recombinant human IFN-. Beta.standard, PART # 844510) was added to each well, the plates were sealed and left to incubate at room temperature for 2 hours, and the wash steps of the plate coating method above were repeated. Then, 100. Mu.L of detection antibody (biotinylated murine anti-human IFN-. Beta.detection antibody, PART # 844509) was added to each well, the plates were sealed and left to incubate at room temperature for 2 hours, and the wash steps of the above plate coating method were repeated. Subsequently, 100. Mu.L of strepavidin-HRP (PART# 893975) working solution was added to each well, the plates were closed and left to incubate at room temperature for 20min, during which time the washing steps of the above plate coating method were repeated, protected from light. Then, 100. Mu.L of a substrate solution (Colar Reagent A (H) 2 O 2 ) And Color Reagent B (tetramethylbenzidine), R&Dsystem cat#dy999), plates were closed and left to incubate at room temperature for 20min, this process being protected from light. Finally, 50. Mu.L of stop solution (2N H) was added to each well 2 SO 4 ,R&Dsystem cat#dy 994), the cell plate was tapped to ensure adequate mixing.
OD450 per well was determined using a multifunctional microplate reader (Molecular Devices company, spectramax M3). If the wavelength calibration function is available, it is set to 540nm or 570nm. If the wavelength calibration function is not available, OD450 minus OD540 or OD570 is used. This process was completed within 30min after the addition of the stop solution.
2'3' -cGAMP (Invivogen, cat# tlrl-nacga 23) was used as a positive control compound, and ADU-S100 (MCE, cat# HY12885 a) was used as a control compound:
the data were analyzed using GraphPad Prism 7.0 software and standard curves for ELISA IFN- β content were determined using double log mapping. Substituting the OD value obtained by detection of each sample hole into a standard curve equation to obtain the corresponding IFN-beta concentration. The EC50 value is calculated by fitting the data to a non-linear S-curve regression to obtain a dose-response curve of IFN- β concentration versus compound concentration.
TABLE 1 activation of THP-1 cell IFN- β secretion by representative example Compounds (EC 50, μM)
Examples THP-1 Activity Examples THP-1 Activity
5 B 16 A
8 B 17 A
9 B 18 B
10 A 19 B
11 B 21 B
12 A 2',3'-cGAMP B
13 B ADU-S100 A
15 B 26 A
Wherein: a: EC50 is 1-10 mu M; b: EC50 of 10.1-100 mu M
Experimental results indicate that the compounds of the examples have an activating effect on secretion of IFN- β by THP-1 cells. As can be seen from table 1 and fig. 1, the compounds of examples 10, 12, 16, 17 and 26 have particularly significant STING agonist activity: their EC50 is stronger than that of 2',3' -cGAMP, and is equivalent to ADU-S100; wherein the compounds of examples 10, 16 and 17 all stimulated THP-1 cells to secrete IFN- β at a higher maximum concentration than ADU-S100.
Example 2: inhibitory Activity of the Compounds of the invention on CT26 cell proliferation
CT26 cells are a mouse colorectal cancer cell line. The inhibitory activity of the compounds on CT26 cell proliferation was evaluated in this experiment using the CellTiter-Glo Luminescence Cell Viability Assay kit from Promega.
When CT26 cells (ATCC #CRL-2638) were recovered, the cell cryopreservation tube was rapidly shaken in a water bath at 37℃to be thawed within 1 min. The thawed cell suspension was collected and purified by using a solution containing 10% FBS (GIBCO, cat#10099-141) (GE company, cat# SH 30243.01), centrifuging at 1000 rpm for 5min, and discarding the supernatant. Cell pellet was resuspended in 5mL of complete medium (DMEM medium containing 10% FBS) and placed at a bottom area of 25cm 2 Placing the cell culture flask of (C) in 37 ℃ and 5% CO 2 Culturing in a cell culture box with 95% humidity. And (5) carrying out cell passage when the cell confluence rate reaches 70% -80%. At the time of cell passage, all cells in the culture flask are transferred to a 15mL centrifuge tube, centrifuged at 1000 rpm for 5min, and the supernatant is discarded. 5mL of fresh complete medium was added to resuspend the cell pellet, 1mL was placed at bottom area 25cm 2 In the cell culture flask of (2), 4mL of fresh complete medium was fed for further culture. Plating is performed when the cell confluency rate reaches 70% -80% again. With reference to the cell passage method at plating, 1/5 of the cell suspension was kept for continued culture, and the remaining 4/5 of the cell suspension was placed in a 15mL centrifuge tube. The old medium was discarded by centrifugation, the cells were washed once with DMEM medium (without serum addition), and the supernatant was removed by centrifugation. Cells were resuspended in DMEM medium (without serum addition). Cell viability was measured by trypan blue exclusion and plated at a cell viability of 95% or higher. The density was 1.1X10 using DMEM medium (without serum addition) 4 Each viable cell/mL of cell suspension was added to a 96-well clear flat bottom black wall cell culture plate well (Corning, cat# 3603) to give a cell density of 1000 viable cells/well in the culture plate. A control group containing no cells, no compound, only complete medium (i.e., a broth control) was set, and a control group containing no compound, no cells (i.e., a cell control) was set. The cell plates were placed in a cell incubator for overnight incubation.
First, a 10mM stock solution of compound in DMSO (Sigma, cat#D2650) was serially diluted at a dilution factor of 3.16 to the 9 th concentration, and a 10 th concentration of DMSO control without compound was set. Then, the DMSO solutions containing the compounds at different concentrations were diluted 10-fold with PBS (Solarbio, cat#P1020) to give 10% DMSO content in each concentration. Finally, 10 mu L of the solution is added into a corresponding cell culture plate, the initial concentration of the compound is 100 mu M, the dilution multiple of the adjacent concentration is 3.16 times, and the DMSO content in the cell culture plate is 1%. The cell plates were placed in a cell incubator for continued culture for 120 hours.
After 120 hours, the CellTiter-Glo reagent (Promega, cat# G7572) was thawed and the plates were moved to room temperature for 30min to equilibrate, 100uL of CellTiter-Glo was added to each well of the plates, the cells were allowed to lyse well by shaking on an orbital shaker for 5min, the plates were left at room temperature for 20min to stabilize the luminescence signal, and the luminescence values of each well were scanned using a multifunctional microplate reader (Molecular Devices Co., specramax M3) full wavelength.
The following compounds and ADU-S100 (MCE, cat# HY 12885A) were used as controls: clofarabine (Clofarabine; ruhu Hua Ren technology, cat# HR-00701002), cladribine (Cladribine; CSNpharm, cat# CSN 10004), gemcitabine hydrochloride (Gemcitabine Hydrochloride; shao; cat# SY 014538), gemcitabine prodrug LY2334737 (self-made, intermediate 7).
Cell viability under the action of each concentration of compound was calculated using the following formula:
cell viability (%) = (Lum) Drug to be tested -Lum Culture broth control )/(Lum Cell control -Lum Culture broth control )×100%。
The data were analyzed using GraphPad Prism 7.0 software, and non-linear S-curve regression was used to fit the data to yield the dose-response curve, and IC50 values were calculated therefrom.
Table 2 inhibition of CT26 cell proliferation (EC 50, μM) in the examples
Examples CT26,IC50(μM) Examples CT26,IC50(μM)
1 B 13 A
2 B 16 B
3 A 17 B
5 B Clofarabine A
7 B Cladribine A
10 A Gemcitabine A
12 B LY2334737 B
ADU-S100 NA
Wherein: a: IC50<1 μm; b: IC50 is 1-10 mu M; NA: IC50> 10. Mu.M
Experimental results show that the representative example compounds are capable of inhibiting the in vitro growth of CT26 tumor cells; of these, the inhibition ability of the compounds of examples 3, 10 and 13 was particularly pronounced, whereas ADU-S100 had no significant tumor cell inhibition ability. The results of example 1, combined with activity, demonstrate that the compounds of the examples possess multifunctional antitumor properties, namely tumor immune activity of STING agonists and cytotoxic effects of antimetabolite anticancer agents.
Example 3: antitumor Activity of the Compounds of the invention in a bilateral transplantation tumor model of CT26 isogenic mice
BALB/c mice (purchased from Shanghai Ling Biotechnology Co., ltd.) were inoculated subcutaneously 5X 10 on the left and right rear backs, respectively 5 CT26 cells (available from Taicangzexin Biotech Co., ltd., ATCC #CRL-2638) were inoculated in a volume of 0.1 mL/side. When the tumor grows to an average volume of 100mm 3 At this time, the animals were randomly grouped according to tumor size and mouse weight and dosing was started. The day of the first administration was day 0. Two times, on days 0, 4, 50ug compound/mouse (i.e., 2.5 mg/kg) was given by intratumoral injection on the right side, with the same volume of PBS (Hyclone Co., ltd.; cat#SH 30258.01) as a control. Left tumor did not undergo any treatment. Control mice were sacrificed 13 days after dosing and treatment mice were sacrificed 21 days after dosing. During the experiment, tumor volumes and body weights were measured 3 times a week, the calculation formula of the tumor volume is v=d×d×d 2 (wherein D)Represents the tumor major diameter, and d represents the tumor minor diameter).
The data on day 13 post-dose demonstrate that the tested compounds of the present invention are capable of significantly inhibiting bilateral tumor growth. The effect was more pronounced in the right-hand group and most of the tumors faded (fig. 2-a). Of these, the ADU-S100 treated group had a tumor growth inhibition of 99.1%, example 17 of 99.5%, and the remaining groups had 100% inhibition of tumors, with no tumor palpation. At the same time, untreated left tumor growth slowed (FIG. 2-B). On day 13 post-dose, the growth inhibition rates for the left tumor groups were: ADU-S100, 64.2%; example 10, 91.2%; example 12, 75.3%; example 16, 71.7%; example 17, 67.4%. In addition, individual mice were observed to lose weight within 15% after dosing, and body weight was restored after dosing was stopped in this experiment (fig. 2-C). The above results indicate that the compounds of the present invention exhibit tumor suppression capacity comparable to or better than that of ADU-S100 in both CT26 isogenic bilateral tumor models.
Example 4: antitumor activity of the compound in CT26 nude mice transplantation tumor model
BALB/c nude mice (purchased from Shanghai Ling Biotechnology Co., ltd.) were inoculated subcutaneously 5X 10 on the back of the right-hand side, respectively 5 CT26 cells (supplied by Taicangzexin Biotech Co., ltd., ATCC #CRL-2638) were inoculated in a volume of 0.1mL. When the tumor grows to an average volume of 100mm 3 At this time, the administration was randomly grouped according to tumor size and mouse weight. The day of the first administration was day 0. Two times, on day 0 and day 4, 50ug compound/mouse (i.e., 2.5 mg/kg) was injected intratumorally on the right side, with the same volume of PBS (Hyclone Corp., cat#SH 30258.01) as a control. Control mice were sacrificed 9 days after dosing, ADU-S100 and compound of example 10 treated mice were sacrificed 14 days, 16 days later. Tumor volume and body weight were measured 3 times per week during the experiment, the calculation formula of the tumor volume is V =d×d×d-2 (supra).
The data on day 9 post-dose demonstrate that the tested compound of example 10 of the present invention is capable of significantly inhibiting tumor growth in T cell immunodeficient nude mice with a 94.4% inhibition rate. In contrast, ADU-S100 only exhibited partial tumor inhibition, with an inhibition rate of 61.4%, possibly due to its residual immune activity or other unknown reasons. Whether example 10 or ADU-S100, they inhibited tumor growth better than nude mice (fig. 2D). The experiment verifies that ADU-S100 takes activation of T cell mediated immunity as a main action mechanism; the compound of example 10, however, demonstrates in this case superior tumor-inhibiting ability to the STING agonist ADU-S100 due to its multi-functional mechanism tumor-inhibiting activity.
Example 5: immune memory function of the compound in CT26 isogenic mouse or nude mouse transplantation tumor model
BALB/c immunized healthy mice or nude mice (purchased from Shanghai Ling Biotechnology Co., ltd.) were subcutaneously inoculated 5X 10 to the right back side 5 CT26 cells (supplied by Taicangzexin Biotech Co., ltd., ATCC #CRL-2638) were inoculated in a volume of 0.1mL. When the tumor grows to an average volume of 100mm 3 At this time, the animals were randomly grouped according to tumor size and mouse weight and dosing was started. The day of the first administration was day 0. Two doses were administered and 50ug of compound per mouse (i.e. 2.5 mg/kg) was injected intratumorally on days 0, 4. The same volume of PBS (Hyclone, cat# SH 30258.01) was used as a control. Mice in the control group in nude mice or BALB/c mice experiments were sacrificed 9 days or 13 days after dosing, respectively. After 21 days, mice were inoculated subcutaneously 5X 10 again on the back left side 5 The inoculation volume of CT26 cells was 0.1mL. Blank BALB/c or nude mice without any treatment were also vaccinated as controls. During the experiment, tumor volumes and body weights were measured 3 times a week, the calculation formula of the tumor volume is V =d×d×d-2 (supra).
The results show that the compound of example 10 of the present invention tested significantly inhibited CT26 tumor growth, both in immunocompetent and immunodeficient mice. The efficacy of the immune healthy mice was more prominent (FIG. 3-A). On day 5 post-dose, the tumors of the treatment group of example 10 all disappeared. CT26 cells were re-seeded on day 21 in the compound treated group of example 10, respectively. After 7 days (day 30), the average tumor volume of the non-immunized blank BALB/c mice was 75mm 3 WhileThe average tumor volume of mice previously treated in example 10 was 29mm 3 . By the end of the 33 th experiment, the average tumor volumes of the mice in the control group and the immunized group of example 10 were 648mm, respectively 3 And 101mm 3 . This experiment demonstrates that immune memory is developed in immunocompetent mice following treatment with the compound of example 10, and strong immune rejection is developed against the vaccinated allogeneic cells, effectively preventing tumor recurrence.
On the other hand, in immunodeficient nude mice, individual mice developed a tumor growth phenomenon after discontinuation of dosing (fig. 3-B). Finally 3 mice with smaller tumor volumes were selected and re-vaccinated with CT26 cells on day 21. The results showed that the re-inoculated CT26 tumor cells grew at a similar rate as the blank murine tumor cells. The results demonstrate that no immune memory was generated in the nude mice experiment, and further demonstrate that the compound of example 10 also has the tumor-inhibiting ability of cytotoxic action without participation of the immune system in the model.
Example 6: hepatocyte metabolic stability test of the Compounds of the invention
The test of the metabolic stability of the compounds of the invention in hepatocytes of 5 species (mouse, rat, dog, monkey, human) was similarly performed according to standard methods of in vitro metabolic stability studies conventional in the art, for example, as described in (Kerns, edward H.and Di Li (2008): drug-like Properties: peptides, structure Design and Methods: from ADME to Toxicity optimizations. San Diego: academic Press; di, li et al, optimization of a Higher Throughput Microsomal Stability Screening Assay for Profi1ing Drug Discovery Candidates, J biomol. Screen.2003,8 (4), 453).
The hepatocytes used in the experiments were: human hepatocytes (SHQY, lot#hep 190006); dog hepatocytes (IVT, lot#zmb); monkey hepatocytes (Xenotech, lot # 2010022); rat hepatocytes (SHQY, lot#hep 134045); mouse hepatocytes (BioIVT, cat.#m005052, lot.#meo).
The cryopreserved tube hepatocytes were removed from the liquid nitrogen tank and immediately placed in an oscillating water bath at 37±1 ℃ for 2 minutes±15 seconds. Transfer of hepatocytes to 50mL of hepatocyte thawing medium (composition: Williams E medium, 35mL, invitrogen corporation, cat# A1217601; isotonic Percoll solution, 13.5mL, general company, cat# 17-0891-01; du's phosphate buffer, 1.5mL, invitrogen company, cat# 14200-075; glutamax,500 μL, invitrogen corporation, cat# 35050061; HEPES,750 μl, invitrogen corporation, cat# 15630106; fetal bovine serum, 2.5ml, invitrogen company, cat# 10091130; recombinant human insulin, 50 μl, invitrogen company, cat# 12585014; dexamethasone (in 10mM DMSO solution), 5. Mu.L, sigma, cat# D1756) was gently mixed and the heart was isolated at 500 rpm for 3min. After centrifugation, the supernatant was carefully aspirated (without disturbing the cell pellet), 10 Xvolumes of pre-warmed KHB buffer (Krebs-Henseleit buffer, sigma, cat#K3753-10X 1L) and 5.6g/L HEPES were added, the cell pellet was resuspended, gently mixed, and the heart was isolated at 500 revolutions/3 min. The supernatant was aspirated and cell pellet was not touched and cell viability and number were determined. Hepatocytes were counted and then the cell suspension KHB buffer was diluted to an appropriate density (viable cell density = 2 x 10 6 Individual cells/mL). The hepatocyte solution was placed on ice until use.
2 Xdosing solution was prepared in preheated KHB (1% dimethyl sulfoxide) and centrifuged at 5594g for 15min (Thermo Multifure. Times.3R), wherein 200. Mu.M of dosing solution: to 980. Mu.L of dimethyl sulfoxide was added 20. Mu.L of a stock solution of the compound (10 mM, DMSO solution); 2 x dosing solution: to 990. Mu.L KHB was added 10. Mu.L of 200. Mu.M stock solution (2. Mu.M after dilution).
To wells at the indicated different time points 50 μl of pre-heated 2 x dosing solution was added. mu.L of the pre-warmed hepatocyte solution (2X 10) 6 Individual cells/mL) was added to the designated wells for 15min, 30min, 60min and 120min detection, and then timing was started and the reaction plates were placed in an incubator at 37 ℃.
100. Mu.L of IS (salidrool or imipramine) containing acetonitrile (Merck, cat. # CN 34854-4L) was added to wells designed for 0min, gently mixed, and then 50. Mu.L of a pre-heated hepatocyte solution (2X 10) 6 Individual cells/mL), closed well plates. 100. Mu.L of IS-containing acetonitrile was added to the wells at 15min, 30min, 60min and 120min, respectively, and then blocked. After quenching, shakePlates were shaken on an actuator (IKA, MTS 2/4) for 10min (600 rpm). The plates were sonicated for 2min and then centrifuged at 5594g for 15min (Thermo multiplex. Times.3R). mu.L of supernatant from each well was transferred to a 96-well sample plate containing 50. Mu.L of ultrapure water (milipore, ZMQS50F 01) for LC/MS analysis.
The concentration of the compound to be tested (C0) at the time point T0 is taken as 100%, the concentrations at other incubation time points are converted into percentage residual amounts, the natural logarithm of the percentage residual amounts at each time point is subjected to linear regression on the incubation time to obtain a slope K, and then the hepatocyte clearance rate (Cl) is calculated according to the following formula int ) And in vitro half-life (T1/2):
T1/2=1n2/K=0.693/K
Cl int = (0.693/T1/2) × (1/hepatocyte density) ×scale factor
The hepatocyte density is the final concentration of hepatocytes in the incubation system of the experiment: 1X 10 6 And each mL. Scale factor = hepatocyte count x liver weight (mice 11812.5 x 10 for 5 species of hepatocytes, respectively) 6 One/kg, rat 4680X 10 6 Dog 6880X 10 per kg 6 Every kg, monkey 3900X 10 6 Per kg, human 2544.3X 10 6 And (3) per kg).
As can be seen from fig. 4A, the compound of the present invention, such as the compound of example 10, showed better metabolic stability in hepatocytes of 5 species; it has a long metabolic half-life and a low clearance rate.
Example 7: discovery and identification of metabolites of the Compounds of the invention
The primary metabolites of the compounds of the present invention in 5 species (mouse, rat, dog, monkey, human) of hepatocytes were similarly identified according to standard methods of in vitro metabolic stability studies conventional in the art, for example, those described in (Kerns, edward H.and Di Li (2008): drug-like Properties: peptides, structure Design and Methods: from ADME to Toxicity optimization. San Diego: academic Press; di, li et al, optimization of a Higher Throughput Microsomal Stability Screening Assay for Profi1ing Drug Discovery Candidates, J biomol. Screen.2003,8 (4), 453).
The hepatocytes used in the experiments were: human hepatocytes (SHQY, cat. #BQHPCH10, lot. #HEP190006-TA 05); dog liver cells (BioIVT, cat.#m00205, lot.#zmb); monkey hepatocytes (XENOTECH, cat.# ppch2000, lot.# 2010022); rat hepatocytes (SHQY, cat.# BQR 1000.h15, lot.# HEP 134049); mouse hepatocytes (BioIVT, cat.#m005052, lot.#meo).
HI hepatocyte maintenance medium (BIOVT, cat. # Z99009; lot. # C02060C) was warmed to 37 ℃. The cryopreserved tube hepatocytes were removed from the liquid nitrogen tank and immediately placed in an oscillating water bath at 37+ -1deg.C for 2 min+ -15 seconds. The hepatocytes were transferred to 50ml of HI medium, gently mixed, and the heart was isolated at 500 rpm for 3min. After centrifugation, the supernatant was carefully aspirated away (without disturbing the cell pellet). 10 Xvolumes of pre-warmed HI medium was added and the pellet resuspended, gently mixed, and the heart was isolated at 500 rpm for 3min. The supernatant was aspirated and no cell pellet was touched. Hepatocytes were counted and then the cell suspension was diluted to an appropriate density with HI medium (viable cell density=2×10 6 Individual cells/mL), the hepatocyte solution was placed on ice until use.
2 Xdosing solutions were prepared in pre-warmed HI medium. A2 Xdosing solution (20. Mu.M) was prepared as follows: to 3992. Mu.L HI medium was added 8. Mu.L of 10mM compound stock (20. Mu.M, 0.2% dimethyl sulfoxide after dilution).
Preheating the hepatocyte solution and 2 x dosing solution to the prescribed T 240 And T 240-w/o 200. Mu.L of the pre-heated 2 Xdosing solution was added to the wells of (C). For T0, 1200. Mu.L of acetonitrile (Merck, cat. # CN 34854-4L) and 200. Mu.L of hepatocyte solution (2X 10) were added to the wells 6 Individual cells/mL), then 200 μl of pre-warmed 2 x dosing solution was added and the well plate was blocked. 200. Mu.L of the preheated hepatocyte solution (2X 10) 6 Individual cells/mL) to be added to the specified for T 240 Is in the hole of (2); to be designated for T 240-w/o 200. Mu.L of pre-warmed HI medium was added to the wells of (C) and the timing was started; the reaction plate was placed at 37℃in CO 2 In an incubator.
At 240min, 1200 μl acetonitrile was added to each of the designated wells, and the well plates were then blocked. After quenching, the plates were sonicated for 2min and then centrifuged at 1400 rpm for 5min. 1200 μl of supernatant was evaporated under nitrogen flow until dryness. The dried extract was then redissolved with 200. Mu.L acetonitrile/water (1:3 v/v), vortexed for 2min and centrifuged at 14000 rpm for 5min. 2/5. Mu.L of the supernatant was injected into LC-UV-MS for analysis.
As can be seen from FIG. 4-B, the compound of the present invention such as the compound of example 10 can detect its decomposition to form the cytotoxic small molecule Clofarabine (Clofarabine) in hepatocytes of all 5 species, and this is the major metabolite of the compound of example 10. In combination with the results of activity example 6, it is highly likely that CDN molecules with STING agonistic activity and small cytotoxic molecules are present simultaneously over a period of time into the body, thereby achieving a combination at the molecular level providing an enhanced, even synergistic, anti-tumor effect. The results also mechanistically explain why they are well-behaved in mouse tumor suppression experiments.
Example 8: drug efficacy comparison of the anti-tumor Activity of the Compounds of the invention in combination with pure CDN STING agonist and cytotoxic drug in CT26 isogenic mice bilateral transplantation tumor model
BALB/c mice (purchased from Shanghai Ling Biotechnology Co., ltd.) were inoculated subcutaneously 5X 10 on the left and right rear backs, respectively 5 CT26 cells (available from Taicangzexin Biotech Co., ltd., ATCC #CRL-2638) were inoculated in a volume of 0.1 mL/side. When the tumor grows to an average volume of 100mm 3 At this time, the animals were randomly grouped according to tumor size and body weight of mice (compound group of the present invention; simple CDN STING agonist single drug group; cytotoxic drug single drug group; simple CDN STING agonist and cytotoxic drug combination group) and started to be administered. The day of the first administration was day 0. Three times, on days 0, 4, and 7, the right intratumoral injection was administered, with the same volume of PBS (Hyclone Co., ltd. # SH 30258.01) as a control. Left tumor did not undergo any treatment. Control mice were sacrificed 13 days after dosing and treatment mice were sacrificed 21 days after dosing. During the experiment, tumor volumes and body weights were measured 3 times a week, the calculation formula of the tumor volume is V =d×d×d-2 (supra).
The experimental result shows that the tumor growth inhibition rate of the compound group is obviously higher than that of the CDN STING single drug group and the cytotoxic drug single drug group, the activity of the compound group is equivalent to the sum of the activities of the CDN STING single drug group and the cytotoxic drug single drug group, and even higher, and the compound group has improved and even synergistic tumor inhibition activity.
The general or preferred definitions of the features specified in the various enumerated embodiments of the present invention may be combined with the general or preferred definitions of other specified features to yield additional embodiments of the present invention. As if such combinations were set forth specifically and individually herein, unless the context clearly shows otherwise.
In this specification, a number of prior publications are referenced. These publications are not considered relevant to the patentability of the invention, but are incorporated herein by reference in their entirety. The reference in this specification to any prior publication (or information derived from it) is, and should not be taken as, an acknowledgement or admission or any form of suggestion that the corresponding prior publication (or information derived from it) forms part of the common general knowledge in the field of technology as referred to in this specification.

Claims (31)

  1. A cyclic dinucleotide compound of formula (II),
    Wherein the method comprises the steps of
    B 1 Is adenine substituted by XWherein X is selected from Cl, F or NHC 1-6 An alkyl group; or optionally by R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group;
    R 1 and R is 1 ' each independently selected from H, F or-OH;
    B 2 selected from adenine optionally substituted with XWherein X is selected from H, F or Cl; optionally by R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 An alkyl group; or guanineWherein OH is optionally C 1-6 Alkyl substitution;
    represents that the phosphate bond can be attached to the pentose at the 2 'or 3' position, and the site of the pentose, which is not cyclic with the phosphoric acid, is R 2 And R is 2 ' substitution; and
    R 2 and R is 2 ' each independently selected from H, -OH or F;
    provided that when B 1 Or B is a 2 One of which is optionally R a In the case of substituted cytosine, the carbon atom adjacent to the pentose ring to which it is attached is substituted with two F;
    stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts, prodrugs or solvates thereof.
  2. A compound of formula (II), a stereoisomer, a tautomer, a stable isotope variant, a pharmaceutically acceptable salt, a prodrug or a solvate thereof according to claim 1 whereinRepresents a phosphate bond attached to the 3' position of pentose, formula (II) having
  3. A compound according to claim 2 wherein B 1 Is thatR 1 And R is 1 ' are H, or R 1 And R is 1 ' one is H and the other is F.
  4. A compound according to claim 2 wherein B 1 Is adenine substituted by XWherein X is selected from NHC 1-6 Alkyl, preferably NHCH 3 ,R 1 And R is 1 ' are H, or R 1 And R is 1 ' one is H and the other is F.
  5. A compound according to claim 2 wherein B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 Alkyl, R 1 And R is 1 ' are both F.
  6. The compound according to any one of claims 2-5, wherein B 2 Is guanineOr adenineR 2 And R is 2 ' one is H and the other is selected from-OH or F.
  7. The compound according to any one of claims 2-5, wherein B 2 Is adenine substituted by XWherein X is Cl, R 2 And R is 2 ' one is H and the other is F, or R 2 And R is 2 ' are all H.
  8. The compound according to any one of claims 2-5, wherein B 2 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 Alkyl, R 2 And R is 2 ' are both F.
  9. A compound of formula (II) according to claim 1, stereoisomers, tautomers thereofAn isomer, stable isotopic variant, pharmaceutically acceptable salt, prodrug or solvate, whereinRepresents a phosphate bond attached to the 2' -position of pentose, formula (II) having
  10. The compound according to claim 9, wherein B 1 Is thatR 1 And R is 1 ' are H, or R 1 And R is 1 ' one is H and the other is F.
  11. The compound according to claim 9, wherein B 1 Is adenine substituted by XWherein X is NHC 1-6 Alkyl, preferably NHCH 3 ,R 1 And R is 1 ' are H, or R 1 And R is 1 ' one is H and the other is F.
  12. The compound according to claim 9, wherein B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O))-C 1-14 Alkyl, R 1 And R is 1 ' are both F.
  13. A compound according to any one of claims 9 to 12 wherein B 2 Is guanineOr adenineR 2 And R is 2 ' one is H and the other is-OH.
  14. The compound according to claim 1, 2 or 9, which is
    Or a pharmaceutically acceptable salt or solvate thereof.
  15. The compound according to claim 14, wherein B 1 Is thatR 1 And R is 1 ' are H, or R 1 Is F and R 1 ' is H.
  16. The compound according to claim 14, wherein B 1 Is adenine substituted by XWherein X is NHC 1-6 Alkyl, preferably NHCH 3 ,R 1 And R is 1 ' are H, or R 1 Is F and R 1 ' is H.
  17. The compound according to claim 14, wherein B 1 Is optionally R a Substituted cytosinesWherein R is a Selected from H or-C (O) -C 1-14 Alkyl, R 1 And R is 1 ' are both F.
  18. A compound according to any one of claims 14 to 17, wherein in formula (II-a'), B 2 Is guanine Or adenineR 2 ' is H, R 2 Selected from-OH or F.
  19. A compound according to any one of claims 14 to 17, wherein in formula (II-a'), B 2 Is adenine substituted by XWherein X is Cl, R 2 Is H, another R 2 ' is F, or R 2 And R is 2 ' are all H.
  20. A compound according to any one of claims 14 to 17, wherein in formula (II-B'), B 2 Is bird typePurine(s)Or adenineR 2 ' is H, R 2 is-OH.
  21. A cyclic dinucleotide compound selected from
    Or a pharmaceutically acceptable salt or solvate thereof.
  22. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1-21 and a pharmaceutically acceptable excipient.
  23. The pharmaceutical composition according to claim 22, which is in the form of a topical administration.
  24. A method for the prevention or treatment of a disease associated with or mediated by STING, in particular a viral infection or tumour, in a mammal, in particular a human, which method comprises administering an effective amount of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22 to 23.
  25. Use of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22-23 as a STING agonist for the treatment or prophylaxis of diseases associated with or mediated by STING, in particular viral infections or tumours.
  26. Use of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22-23 as a cytotoxic agent for the treatment or prophylaxis of viral infections or tumors.
  27. Use of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22-23 as a multifunctional active agent for immunotherapy and cytotoxic therapy.
  28. Use according to claim 27, wherein the multifunctional agent is used to activate STING signalling pathways to activate the immune system to function against tumour and antiviral replication, to cause tumour cell death or to prevent viral replication by release of cytotoxic agents, to subsequently activate STING to kill tumour cells by release of tumour DNA, and to generate an antibody-antigen response by release of tumour neoantigens to provide the ability to "immune memory" or persistent immunity to tumours.
  29. Use of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22-23 in the manufacture of a medicament for the treatment or prophylaxis of a disease associated with or mediated by STING, particularly viral infection or tumour.
  30. Use of a compound according to any one of claims 1 to 21 or a pharmaceutical composition according to any one of claims 22 to 23 in the manufacture of a cytotoxic agent for the treatment or prophylaxis of a viral infection or tumour.
  31. The method or use according to any one of claims 24-26 and 28-30, wherein the tumor is selected from brain cancer, head and neck cancer, skin cancer, melanoma, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, blood cancer, lung cancer, small cell lung cancer, non-small cell lung cancer, bone cancer, colorectal cancer, liver cancer, renal cell cancer, pancreatic cancer, hodgkin's lymphoma or leukemia.
CN202180071565.7A 2020-10-20 2021-10-19 Multifunctional cyclic dinucleotide and application thereof Pending CN117015383A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2020111240678 2020-10-20
CN202011124067 2020-10-20
PCT/CN2021/124704 WO2022083584A1 (en) 2020-10-20 2021-10-19 Multifunctional cyclic dinucleotide and use thereof

Publications (1)

Publication Number Publication Date
CN117015383A true CN117015383A (en) 2023-11-07

Family

ID=83475602

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202111201201.4A Pending CN115160392A (en) 2020-10-20 2021-10-15 Multifunctional cyclic dinucleotide and application thereof
CN202180071565.7A Pending CN117015383A (en) 2020-10-20 2021-10-19 Multifunctional cyclic dinucleotide and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202111201201.4A Pending CN115160392A (en) 2020-10-20 2021-10-15 Multifunctional cyclic dinucleotide and application thereof

Country Status (1)

Country Link
CN (2) CN115160392A (en)

Also Published As

Publication number Publication date
CN115160392A (en) 2022-10-11

Similar Documents

Publication Publication Date Title
CA3038860C (en) Cyclic dinucleotide compounds
CN110382515B (en) Modified cyclic dinucleotide compounds
CA2906157C (en) Highly active anti-neoplastic and anti-proliferative agents
EP3707151B1 (en) Sting modulator compounds, and methods of making and using
ES2390921T3 (en) E1 activating enzyme inhibitors
JP7317014B2 (en) Cyclic dinucleotides as STING agonists
WO2019125974A1 (en) Cyclic di-nucleotide compounds as sting agonists
EP4242212A2 (en) Sting modulator compounds with sulfamate linkages, and methods of making and using
CN111918871A (en) Cyclopentane-based STING (interferon gene stimulator) modulators
CA3112326A1 (en) Antiviral pyridopyrazinedione compounds
CN111971291A (en) Cyclic dinucleotide compounds containing 2-aza-hypoxanthine or 6H-pyrazolo [1,5-D ] [1,2,4] triazin-7-one as STING agonists
WO2016014527A2 (en) Stabilized nucleotides for medical treatment
TW201825509A (en) Cyclic dinucleotide
WO2022083584A1 (en) Multifunctional cyclic dinucleotide and use thereof
CN117015383A (en) Multifunctional cyclic dinucleotide and application thereof
RU2809547C2 (en) New cyclic dinucleotide derivative and its antibody-drug conjugate
BR112018001695B1 (en) PYRROLO[2,3-d]PYRIMIDINE COMPOUND, JAK3 INHIBITOR, PHARMACEUTICAL COMPOSITION, AGENT, USES OF SAID COMPOUND AND METHOD FOR PRODUCING THE SAME

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20231107