WO2019144885A1 - 取代的吡唑并[1,5-a]嘧啶类的大环化合物 - Google Patents

取代的吡唑并[1,5-a]嘧啶类的大环化合物 Download PDF

Info

Publication number
WO2019144885A1
WO2019144885A1 PCT/CN2019/072833 CN2019072833W WO2019144885A1 WO 2019144885 A1 WO2019144885 A1 WO 2019144885A1 CN 2019072833 W CN2019072833 W CN 2019072833W WO 2019144885 A1 WO2019144885 A1 WO 2019144885A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
hydrogen
mmol
hydrazine
Prior art date
Application number
PCT/CN2019/072833
Other languages
English (en)
French (fr)
Inventor
王义汉
赵九洋
Original Assignee
深圳市塔吉瑞生物医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Priority to EP19743399.8A priority Critical patent/EP3744723B1/en
Priority to JP2020540580A priority patent/JP7162915B2/ja
Priority to US16/964,069 priority patent/US11345703B2/en
Publication of WO2019144885A1 publication Critical patent/WO2019144885A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention belongs to the technical field of medicine, and in particular relates to a substituted macrocyclic compound of pyrazolo[1,5-a]pyrimidine and a composition comprising the same and use thereof. More particularly, the present invention relates to certain deuterium-substituted methyl 9-difluoro-15-aza -2,11,16,20,21,24- six dioxolane [16.5.2.0 2,6, 0 7, 12, 0 21, 25] pentacosa-1 (24), 7,9,11,18 (25), 19,22- seven-en-17-one compounds and stereoisomers, such as deuterium substituted
  • the compounds exhibit Trk family protein tyrosine kinase inhibition and are useful in the treatment of pain, inflammation, cancer and certain infectious diseases, and these oxime substituted compounds have superior pharmacokinetic properties.
  • Trk is a high-affinity receptor tyrosine kinase that is activated by a group of soluble growth factors that are neurotrophic factors (NT).
  • the Trk receptor family has three members, namely TrkA, TrkB and TrkC.
  • the neurotrophic factors are (1) nerve growth factor (NGF) that activates TrkA, (2) brain-derived neurotrophic factor (BDNF) and NT-4/5, which activate TrkB, and (3) TrkC that activates TrkC.
  • NGF nerve growth factor
  • BDNF brain-derived neurotrophic factor
  • TrkC that activates TrkC.
  • Trk is widely expressed in neuronal tissues and is involved in the maintenance, signaling and survival of neuronal cells.
  • Trk overexpression, activation, amplification and/or mutations are associated with many cancers including neuroblastoma, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer, multiple myeloma, astrocytoma and Neuroblastoma, glioma, melanoma, thyroid cancer, pancreatic cancer, large cell neuroendocrine tumor and colorectal cancer.
  • inhibitors of the Trk/neurotrophin pathway have been shown to be effective in a variety of preclinical animal models for the treatment of pain and inflammatory diseases.
  • the neurotrophin/Trk pathway has also been implicated in the pathogenesis of neurodegenerative diseases, including multiple sclerosis, Parkinson's disease, and Alzheimer's disease.
  • the modulating neurotrophic factor/Trk pathway can be used to treat these and related diseases.
  • TrkA receptor is critical for the disease process in the parasitic infection of Trypanosoma cruzi (Chagas disease) in human hosts. Therefore, TrkA inhibitors can be used to treat Chagas disease and related protozoal infections.
  • Trk inhibitors can also be used to treat diseases associated with imbalances in bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastasis.
  • Bone metastases are a common complication of cancer, up to 70% in patients with advanced breast or prostate cancer and about 15 in patients with lung, colon, stomach, bladder, uterine, rectal, thyroid or kidney cancer Up to 30%. Bone metastasis can cause severe pain, pathological fractures, life-threatening hypercalcemia, spinal cord compression and other neuro-compression syndromes. For these reasons, bone metastases are costly and serious cancer complications. Therefore, an agent that can induce apoptosis of proliferating bone cells is very advantageous.
  • TrkA receptor and TrkC receptor have been observed in the osteogenic region of the fractured mouse model. In addition, almost all osteoblast apoptosis agents are very advantageous. Expression of the TrkA receptor and TrkC receptor has been observed in the osteogenic region of the fractured mouse model. In addition, localization of NGF was observed in almost all osteoblasts. Recently, it was demonstrated that pan-Trk inhibitors in human hFOB osteoblasts inhibit tyrosine signaling activated by neurotrophic factors that bind to all three Trk receptors. This data supports the theory of using Trk inhibitors to treat bone remodeling diseases, such as bone metastases in cancer patients.
  • Larotrectinib (LOXO-101) is the first generation of Trk inhibitor developed by Loxo Oncology.
  • LOXO-101 began treating the first patient in March 2015; on July 13, 2016, it was awarded the breakthrough drug qualification by the FDA for Inoperable resection or metastatic solid tumors in adults and children with positive Trk fusion gene mutations; key enrollment completed in February 2017.
  • the Trk gene of cancer patients may produce some mutations (such as NTRK1 G595R, NTRK3 G623R and other mutations), resulting in drug resistance.
  • LOXO-195 (chemical name (6R, 15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7, 12, 0 21, 25] pentacosa-1 (24), 7,9,11,18 (25), 19,22- heptaen-17-one, having the following structure formula) are developed by the company Loxo Oncology
  • the second-generation Trk inhibitor can effectively resist the drug resistance produced by Larotrectinib.
  • ADME ulcerative co-oxidative desorption, distribution, metabolism, and/or excretion
  • Many of the drugs currently on the market also limit their range of applications due to poor ADME properties.
  • the rapid metabolism of drugs can lead to the inability of many drugs that could be effectively treated to treat diseases because they are too quickly removed from the body.
  • Frequent or high-dose medications may solve the problem of rapid drug clearance, but this approach can lead to problems such as poor patient compliance, side effects caused by high-dose medications, and increased treatment costs.
  • rapidly metabolizing drugs may also expose patients to undesirable toxic or reactive metabolites.
  • the present invention discloses a novel hydrazine-substituted 9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentene ring [16.5.2.0 2,6 , 0 7,12 ,0 21,25 ]dipentadecane-1(24), 7,9,11,18(25),19,22-hepten-17-one and its stereoisomers (for example, Compound ⁇ , compound ⁇ -a and compound ⁇ -b, structural formula below), and compositions and uses thereof, have better Trk kinase inhibitory activity, lower side effects, better pharmacodynamics/pharmacokinetics Performance, can be used to treat Trk kinase mediated diseases.
  • the term "compound of the invention” refers to formula (A), formula (A-1), formula (A-2), formula (Aa), formula (Aa-1), formula (Aa-2), formula (I), a compound of the formula (II), the formula (III), the formula (IV), the formula (Ia), the formula (IIa), the formula (IIIa) and the formula (IVa).
  • the term also includes formula (A), formula (A-1), formula (A-2), formula (Aa), formula (Aa-1), formula (Aa-2), formula (I), formula (II).
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 is deuterium;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 Each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 At least one of Y 3 , Y 4 and Y 5 is ⁇ ;
  • the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient.
  • a compound of the invention is provided in the pharmaceutical composition in an effective amount.
  • the compounds of the invention are provided in a therapeutically effective amount.
  • the compounds of the invention are provided in a prophylactically effective amount.
  • the present invention provides a process for the preparation of a pharmaceutical composition as described above, comprising the steps of: mixing a pharmaceutically acceptable excipient with a compound of the present invention to form a pharmaceutical composition.
  • the invention also relates to a method of treating a disease mediated by Trk kinase in a subject.
  • the method comprises administering to the subject a therapeutically effective amount of a compound of the invention.
  • the cancer is mediated by TrkA; TrkB; or TrkA and TrkB.
  • the patient is diagnosed or identified as having a Trk-related cancer.
  • the compound is administered orally, subcutaneously, intravenously or intramuscularly.
  • the compound is administered chronically.
  • the Trk kinase mediated disease is selected from the group consisting of pain, cancer, inflammation, a neurodegenerative disease, or a trypanosome infection.
  • deuterated means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuterated is used interchangeably with “one or more deuterated”.
  • non-deuterated compound means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
  • pharmaceutically acceptable salt means that, within the scope of sound medical judgment, it is suitable for contact with tissues of humans and lower animals without excessive toxicity, irritation, allergies, etc., and with reasonable benefits/dangers. Those salts that are proportionate.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., pharmaceutically acceptable salts as described in detail in J. Pharmaceutical Sciences (1977) 66: 1-19.
  • Pharmaceutically acceptable salts of the compounds of the invention include those derived from suitable inorganic and organic acids and bases.
  • the invention also includes isotopically labeled compounds, equivalent to the original compounds disclosed herein.
  • isotopes which may be listed as compounds of the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, respectively. , 31 P, 32 P, 35 S, 18 F and 36 Cl. a compound, or an enantiomer, a diastereomer, an isomer, or a pharmaceutically acceptable salt or solvate of the present invention, wherein an isotope or other isotopic atom containing the above compound is within the scope of the present invention .
  • isotopically-labeled compounds of the present invention such as the radioisotopes of 3 H and 14 C, are also among them, useful in tissue distribution experiments of drugs and substrates. ⁇ , ie 3 H and carbon 14, ie 14 C, are easier to prepare and detect and are preferred in isotopes.
  • isotopically labeled compounds can be prepared in a conventional manner by substituting a readily available isotopically labeled reagent with a non-isotopic reagent using the protocol of the examples.
  • the compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of "stereoisomer" forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the invention may be in the form of individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of a mixture of stereoisomers, A racemic mixture and a mixture rich in one or more stereoisomers are included.
  • the isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of a chiral salt; or preferred isomers can be passed Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • the compounds of the invention may be in amorphous or crystalline form. Furthermore, the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • crystalline form refers to a different arrangement of chemical drug molecules, generally expressed as the presence of a pharmaceutical material in a solid state. A drug may exist in a plurality of crystalline forms, and different crystal forms of the same drug may have different dissolution and absorption in the body, thereby affecting the dissolution and release of the formulation.
  • solvent compound refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio.
  • Hydrophilic compound means a complex formed by the coordination of a compound of the invention with water.
  • prodrug refers to a compound that is converted in vivo to an active form having its medical effect by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ACSSymposium Series Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: Solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each introduction This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention which, when administered to a patient, releases the parent compound in vivo.
  • Prodrugs are typically prepared by modifying functional groups in such a way that the modifications can be cleaved by routine manipulation or in vivo to yield the parent compound.
  • Prodrugs include, for example, a compound of the invention wherein a hydroxy, amino or thiol group is bonded to any group which, when administered to a patient, can be cleaved to form a hydroxy, amino or thiol group.
  • representative examples of prodrugs include, but are not limited to, the hydroxy, thiol and amino functional acetate/amide, formate/amide and benzoate/amide derivatives of the compounds of the invention.
  • an ester such as a methyl ester, an ethyl ester or the like can be used.
  • the ester itself may be active and/or may hydrolyze under conditions in humans.
  • Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups which readily decompose in the human body to release the parent acid or a salt thereof.
  • crystalline form refers to a different arrangement of chemical drug molecules, generally expressed as the presence of a pharmaceutical material in a solid state.
  • a drug may exist in a plurality of crystalline forms, and different crystal forms of the same drug may have different dissolution and absorption in the body, thereby affecting the dissolution and release of the preparation.
  • the term "subject” includes, but is not limited to, a human (ie, a male or female of any age group, eg, a pediatric subject (eg, an infant, a child, adolescent) or an adult subject (eg, Young adults, middle-aged adults or older adults) and/or non-human animals, for example, mammals, for example, primates (eg, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses , sheep, goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • treatment includes the effect of a subject having a particular disease, disorder, or condition that reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder. Or the development of a condition ("therapeutic treatment"), but also the effect that occurs before the subject begins to have a particular disease, disorder or disease (“prophylactic treatment”).
  • an "effective amount" of a compound refers to an amount sufficient to cause a target biological response.
  • an effective amount of a compound of the invention can vary depending on, for example, the biological target, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the age of the subject. Health conditions and symptoms. Effective amounts include therapeutically and prophylactically effective amounts.
  • a “therapeutically effective amount” of a compound, as used herein, is a quantity sufficient to provide a therapeutic benefit, or one or more associated with a disease, disorder, or condition, in the course of treating a disease, disorder, or condition, unless otherwise stated. Symptoms are delayed or minimized.
  • a therapeutically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other therapies that provides a therapeutic benefit in the treatment of a disease, disorder or condition.
  • the term "therapeutically effective amount” can include an amount that improves overall treatment, reduces or avoids the symptoms or causes of a disease or condition, or enhances the therapeutic efficacy of other therapeutic agents.
  • a “prophylactically effective amount” of a compound is an amount sufficient to prevent a disease, disorder, or condition, or a quantity sufficient to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent disease, unless otherwise stated. The number of relapses of a disorder or condition.
  • a prophylactically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other agents that provides a prophylactic benefit in the prevention of a disease, disorder or condition.
  • the term “prophylactically effective amount” can include an amount that improves the overall amount of prevention, or enhances the prophylactic efficacy of other prophylactic agents.
  • Combination and related terms mean the simultaneous or sequential administration of a therapeutic agent of the invention.
  • a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or together with another therapeutic agent in a single unit dosage form.
  • the invention provides a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 Each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 At least one of Y 3 , Y 4 and Y 5 is ⁇ .
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or
  • the technical scheme of ⁇ includes R 1 is selected from hydrogen or hydrazine, R 2 is selected from hydrogen or hydrazine, R 3 is selected from hydrogen or hydrazine, and so on, until R 12 is selected from hydrogen or hydrazine; more specifically, It includes a technical scheme in which R 1 is hydrogen or R 1 is deuterium, R 2 is hydrogen or R 2 is deuterium, R 3 is hydrogen or R 3 is deuterium, and so on, until R 12 is hydrogen or R 12 is deuterium.
  • the technical scheme that "Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine" includes Y 1 being selected from hydrogen or hydrazine, Y 2 being selected from hydrogen or ⁇ , Y 3 is selected from hydrogen or hydrazine, and so on, until Y 5 is selected from hydrogen or hydrazine; more specifically, Y 1 is hydrogen or Y 1 is hydrazine, Y 2 is hydrogen or Y 2 is ⁇ Y 3 is hydrogen or Y 3 is ruthenium, and so on, until Y 5 is hydrogen or Y 5 is ruthenium.
  • the technical scheme "X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D" includes X being CH 3 , X being CD 3 , X being CHD 2 or X being CH 2 D Technical solutions.
  • the invention relates to a compound of formula (A-1), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 Each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 At least one of Y 3 , Y 4 and Y 5 is ⁇ .
  • the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 Each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 At least one of Y 3 , Y 4 and Y 5 is ⁇ .
  • the invention relates to a compound of formula (II), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 Each independently selected from hydrogen or hydrazine;
  • X is selected from CH 3 , CD 3 , CHD 2 or CH 2 D;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 At least one of Y 3 , Y 4 and Y 5 is ⁇ .
  • the compound of the formula (A), the formula (A-1), the formula (I) and the formula (II) contains at least one halogen atom, more preferably one germanium atom, more preferably two.
  • Helium atom more preferably three helium atoms, more preferably four helium atoms, more preferably five helium atoms, more preferably six helium atoms, more preferably seven helium atoms, more preferably eight helium atoms More preferably nine helium atoms, more preferably ten helium atoms, more preferably eleven helium atoms, more preferably twelve helium atoms, more preferably thirteen helium atoms, more preferably fourteen Helium atoms, preferably fifteen helium atoms.
  • the cerium isotope content of cerium in the deuterated position is at least 0.015%, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than the natural strontium isotope content.
  • the ground is greater than 95%, more preferably greater than 99%.
  • the strontium isotope content in each deuterated position is at least 5%, preferably more than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25 More preferably, more than 30%, more preferably more than 35%, more preferably more than 40%, more preferably more than 45%, more preferably more than 50%, more preferably more than 55%, more preferably more than 60% More preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, and even more preferably greater than 95%, More preferably greater than 99%.
  • the fifteenth place contains ⁇ , preferably 16 ⁇ , more preferably seventeen ⁇ , more preferably eighteen ⁇ , more preferably nineteen ⁇ , more preferably twenty deuterium.
  • the compounds of formula (A), formula (A-1), formula (I) and formula (II) contain at least one, two, three, four, five, six, seven, eight , nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty thorium atoms.
  • R 1 is selected from hydrogen or hydrazine.
  • R 1 is hydrogen
  • R 1 is deuterium
  • R 2 , R 3 , R 4 , R 5 are each independently selected from hydrogen or hydrazine.
  • R 2 and R 3 are the same.
  • R 4 and R 5 are the same.
  • both R 2 and R 3 are deuterium.
  • both R 2 and R 3 are hydrogen.
  • R 4 and R 5 are both deuterium.
  • R 4 and R 5 are both hydrogen.
  • R 2 , R 3 , R 4 and R 5 are both deuterium.
  • R 2 , R 3 , R 4 and R 5 are all hydrogen.
  • R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or hydrazine.
  • R 7 and R 8 are the same.
  • R 9 and R 10 are the same.
  • R 11 and R 12 are the same.
  • R 6 , R 7 and R 8 are both deuterium.
  • R 6 , R 7 and R 8 are all hydrogen.
  • R 9 and R 10 are both deuterium.
  • both R 9 and R 10 are hydrogen.
  • R 11 and R 12 are both deuterium.
  • R 11 and R 12 are both hydrogen.
  • X is selected from the group consisting of CH 3 , CD 3 , CHD 2 or CH 2 D.
  • X is selected from CH 3 or CD 3.
  • Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine.
  • X is CD 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine.
  • X is CD 3
  • Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are all hydrogen
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or hydrazine.
  • X is CD 3
  • R 1 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are all hydrogen
  • R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or hydrazine.
  • X is CD 3
  • R 1 , R 2 , R 3 , R 4 , R 5 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are all hydrogen
  • R 6 , R 7 , R 8 , R 9 , R 10 , R 11 and R 12 are each independently selected from hydrogen or hydrazine.
  • R 2 , R 3 , R 4 and R 5 are both ⁇ , R 1 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , and R 8 are both deuterium
  • R 1 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine
  • X is selected from CH 3 or CD 3 .
  • R 2 , R 3 , R 4 , R 5 , R 9 and R 10 are both deuterium
  • R 6 , R 7 , R 8 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine
  • X is selected from CH 3 or CD 3 .
  • R 2 , R 3 , R 4 , R 5 , R 11 and R 12 are both deuterium
  • R 6 , R 7 , R 8 , R 9 , R 10 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine
  • X is selected from CH 3 or CD 3 .
  • R 6 , R 7 and R 8 are both ⁇ , R 1 , R 2 , R 3 , R 4 , R 5 , R 9 , R 10 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • R 9 and R 10 are both ⁇ , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 11 , R 12 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • R 11 and R 12 are both fluorene
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , Y 1 , Y 2 , Y 3 , Y 4 and Y 5 are each independently selected from hydrogen or hydrazine
  • X is selected from CH 3 or CD 3 .
  • the invention relates to a compound of formula (Aa), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 - R 12 and X are as defined above.
  • the invention relates to a compound of formula (Aa-1), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 - R 12 and X are as defined above.
  • the present invention is also directed to a compound of formula (Ia), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 - R 12 and X are as defined above.
  • the present invention is also directed to a compound of formula (IIa), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, polymorph, stereoisomer or isotopic variation thereof:
  • R 1 - R 12 and X are as defined above.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 11 and R 12 are selected from hydrogen, R 1 -R 10 Each is independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 9 and R 10 are selected from hydrogen, R 1 -R 8 And R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 9 -R 12 are selected from hydrogen, R 1 -R 8 Each is independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 is selected from H, and R 2 -R 12 are each independently It is selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 , R 11 and R 12 are selected from hydrogen, R 2 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 , R 9 and R 10 are selected from hydrogen, R 2 -R 8 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 , R 9 -R 12 are selected from hydrogen, R 2 -R 8 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from CH 3 and R 1 -R 12 are each independently It is selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from CH 3 and R 11 and R 12 are selected from hydrogen R 1 - R 10 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from CH 3 and R 9 and R 10 are selected from hydrogen R 1 - R 8 and R 11 - R 12 are each independently selected from hydrogen or hydrazine.
  • X is selected from CH 3 and R 9 and R 10 are selected from hydrogen R 1 - R 8 and R 11 - R 12 are each independently selected from hydrogen or hydrazine.
  • the additional condition is that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from CH 3 and R 9 -R 12 is selected from hydrogen R 1 - R 8 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from CH 3 and R 1 is selected from H, R 2 -R 12 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from the group consisting of CH 3 , R 1 , R 11 , and R 12 It is selected from H, R 2 - R 10 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from the group consisting of CH 3 , R 1 , R 9 , and R 10 It is selected from H, R 2 - R 8 and R 11 - R 12 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein X is selected from the group consisting of CH 3 , R 1 , R 9 -R 12 It is selected from H, R 2 - R 8 are each independently selected from hydrogen or hydrazine, with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 are selected from hydrogen, R 1 and R 6 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 11 -R 12 are selected from hydrogen And R 1 and R 6 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 9 -R 10 are selected from hydrogen R 1 , R 6 -R 8 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 9 -R 12 are selected from hydrogen R 1 , R 6 - R 8 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 are selected from hydrogen, R 6 -R 12 Each is independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 11 -R 12 are selected from hydrogen R 6 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 9 -R 10 are selected from hydrogen R 6 -R 8 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 9 -R 12 are selected from hydrogen R 6 - R 8 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 are selected from hydrogen, R 1 and R 6 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 11 -R 12 are selected from hydrogen And R 1 and R 6 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 9 -R 10 are selected from hydrogen R 1 , R 6 - R 8 and R 11 - R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 2 -R 5 and R 9 -R 12 are selected from hydrogen R 1 , R 6 - R 8 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 are selected from hydrogen, R 6 -R 12 Each is independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 11 -R 12 are selected from hydrogen R 6 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 9 -R 10 are selected from hydrogen R 6 -R 8 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 -R 5 and R 9 -R 12 are selected from hydrogen R 6 - R 8 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 with the proviso that the above compound contains at least one hydrazine.
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 1 -R 5 And R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 11 -R 12 Selected from hydrogen, R 1 -R 5 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 10 Selected from hydrogen, R 1 -R 5 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 12 Selected from hydrogen, R 1 -R 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 Selected from hydrogen, R 1 and R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 And R 11 -R 12 are selected from hydrogen, R 1 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 And R 9 -R 10 are selected from hydrogen, R 1 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 11 -R 12 is selected from hydrogen, R 2 -R 5 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 10 is selected from hydrogen, R 2 -R 5 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 12 is selected from hydrogen, R 2 -R 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 is selected from hydrogen, R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 11 -R 12 are selected from hydrogen, R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 9 -R 10 are selected from hydrogen, R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 or CD 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 1 -R 5 And R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 11 -R 12 Selected from hydrogen, R 1 -R 5 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 10 Selected from hydrogen, R 1 -R 5 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 12 Selected from hydrogen, R 1 -R 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 Selected from hydrogen, R 1 and R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 And R 11 -R 12 are selected from hydrogen, R 1 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 And R 9 -R 10 are selected from hydrogen, R 1 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 11 -R 12 is selected from hydrogen, R 2 -R 5 and R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 10 is selected from hydrogen, R 2 -R 5 and R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 9 -R 12 is selected from hydrogen, R 2 -R 5 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 is selected from hydrogen, R 9 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 11 -R 12 are selected from hydrogen, R 9 -R 10 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the invention provides a compound of formula (Aa), formula (Aa-1), formula (Ia), and formula (IIa), wherein R 1 and R 6 -R 8 are selected from the group consisting of ruthenium, R 2 -R 5 and R 9 -R 10 are selected from hydrogen, R 11 -R 12 are each independently selected from hydrogen or hydrazine, and X is selected from CH 3 .
  • the present invention is directed to a compound of formula (Aa-2), formula (IIIa) and formula (IVa), or a pharmaceutically acceptable salt, prodrug, hydrate or solvent compound thereof, polymorph, Stereoisomers or isotopic variants:
  • R 1 - R 12 , Y 1 - Y 5 and X are as defined above.
  • the compound is selected from the group consisting of:
  • the compound does not include a non-deuterated compound.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of the active component.
  • the pharmaceutical composition comprises a therapeutically effective amount of the active component.
  • the pharmaceutical composition comprises a prophylactically effective amount of the active component.
  • compositions of the present invention comprise a safe or effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier.
  • safe and effective amount it is meant that the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical compositions contain from 0.5 to 2000 mg of the compound of the invention per agent, more preferably from 1 to 500 mg of the compound of the invention per agent.
  • the "one dose" is a capsule or tablet.
  • “Pharmaceutically acceptable excipient” means a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants, or vehicles that can be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (eg, human serum albumin) ), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based material, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene-embedded Seg
  • compositions of the present invention can be prepared by combining the compounds of the present invention with suitable pharmaceutically acceptable excipients, for example, as solid, semi-solid, liquid or gaseous preparations such as tablets, pills, capsules. , powders, granules, ointments, emulsions, suspensions, solutions, suppositories, injections, inhalants, gels, microspheres and aerosols.
  • suitable pharmaceutically acceptable excipients for example, as solid, semi-solid, liquid or gaseous preparations such as tablets, pills, capsules. , powders, granules, ointments, emulsions, suspensions, solutions, suppositories, injections, inhalants, gels, microspheres and aerosols.
  • Typical routes of administration of a compound of the invention or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, transmucosal, enteral administration, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, ocular Internal, intramuscular, intramuscular, subcutaneous, intravenous administration.
  • the pharmaceutical composition of the present invention can be produced by a method well known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a drag coating method, a grinding method, an emulsification method, a freeze drying method, and the like.
  • the pharmaceutical composition can be formulated by mixing the active compound with pharmaceutically acceptable excipients which are well known in the art. These excipients enable the compounds of the present invention to be formulated into tablets, pills, troches, dragees, capsules, liquids, gels, slurries, suspensions and the like for oral administration to a patient.
  • Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. For example, it can be obtained by mixing the active compound with a solid excipient, optionally milling the resulting mixture, adding other suitable adjuvants if necessary, and then processing the mixture into granules. The core of a tablet or dragee.
  • Suitable excipients include, but are not limited to, binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
  • microcrystalline cellulose glucose solution, gum arabic, gelatin solution, sucrose and starch paste; talc, starch, calcium stearate or stearic acid; lactose, sucrose, starch, mannitol, sorbitol or phosphoric acid Calcium; silica; cross-linked hydroxymethylcellulose sodium, pre-treated starch, sodium starch glycolate, alginic acid, corn starch, potato starch, methyl cellulose, agar, hydroxymethyl cellulose, cross-linked poly Vinyl pyrrolidone and the like.
  • the core of the dragee may optionally be coated according to methods well known in the ordinary pharmaceutical practice, especially using enteric coatings.
  • compositions may also be suitable for parenteral administration, such as sterile solutions, suspensions or lyophilized products in a suitable unit dosage form.
  • suitable excipients such as fillers, buffers or surfactants can be used.
  • the compounds of the invention may be administered by any route and method of administration, for example by oral or parenteral (e.g., intravenous) administration.
  • a therapeutically effective amount of a compound of the invention is from about 0.0001 to 20 mg/kg body weight per day, such as from 0.001 to 10 mg/kg body weight per day.
  • the dosage frequency of the compounds of the invention is determined by the needs of the individual patient, for example, once or twice daily, or more times per day. Administration may be intermittent, for example, wherein the patient receives a daily dose of a compound of the invention over a period of several days, followed by a patient's daily dose of the compound of the invention for a period of several days or more. .
  • the compounds of the present invention exhibit Trk family protein tyrosine kinase inhibition, and the compounds are useful for the treatment of pain, cancer, inflammation, neurodegenerative diseases or trypanosomal infections and the like.
  • Some embodiments comprise a compound of the invention for use in the treatment of a condition and a disease (eg, a TrkA, TrkB, and/or TrkC mediated condition, such as one or more described herein) that can be treated by inhibition of TrkA, TrkB, and /TrkC kinases
  • a disease eg, a TrkA, TrkB, and/or TrkC mediated condition, such as one or more described herein
  • TrkA, TrkB, and/TrkC kinases e.g, a TrkA, TrkB, and/or TrkC mediated condition, such as one or more described herein
  • the compounds of the invention are also useful in the treatment of pain, including chronic and acute pain.
  • the compounds of the invention are useful for treating various types of pain, neuropathic pain, surgical pain, and pain associated with cancer, surgery, and fractures.
  • the invention provides a method of treating or preventing inflammation in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound of the invention.
  • the method comprises a method of treating the inflammation in a subject.
  • the method comprises a method of preventing the inflammation in a subject.
  • the invention provides a method of treating a neurodegenerative disease in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound of the invention.
  • the neurodegenerative disease is a demyelinating disease.
  • the neurodegenerative disease is a myelination disorder.
  • the neurodegenerative disease is multiple sclerosis.
  • the neurodegenerative disease is Parkinson's disease.
  • the neurodegenerative disease is Alzheimer's disease.
  • the invention provides a method of treating an infectious disease in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound of the invention.
  • the infectious disease is a trypanosome infection.
  • a Trk-related cancer can be selected from the group consisting of: non-small cell lung cancer, papillary thyroid cancer, glioblastoma multiforme, acute myeloid leukemia, colorectal cancer, large cell neuroendocrine cancer, prostate cancer, colon Cancer, acute myeloid leukemia, sarcoma, pediatric glioma, intrahepatic cholangiocarcinoma, hairy cell astrocytoma, low grade glioma, lung adenocarcinoma, salivary gland cancer, secretory breast cancer, fibrosarcoma, Kidney tumors and breast cancer.
  • the Trk-related cancer is selected from the group consisting of: a non-limiting example of a TRK-associated cancer comprising: Spitzoid melanoma, Spitz tumor (eg, metastatic Spitz tumor), non-small cell lung cancer (NSCLC), Thyroid cancer (eg, papillary thyroid tumor (PTC), acute myeloid leukemia (AML), sarcoma (eg, undifferentiated sarcoma or adult soft tissue sarcoma), pediatric glioma, colorectal cancer (CRC), polymorphism Glioblastoma (GBM), large cell neuroendocrine cancer (LCNEC), thyroid cancer, intrahepatic cholangiocarcinoma (LCC), hairy cell astrocytoma, low grade glioma, head and neck squamous epithelium Cell carcinoma, renal tumor, melanoma, bronchial cancer, B-cell cancer, Bronchus cancer, oral or pharynge
  • the compounds of the invention are useful for treating Trk-related cancer in pediatric patients.
  • the compounds provided herein can be used to treat infantile sarcoma, neuroblastoma, congenital mesoderm nephroma, cerebral low grade glioma, and pons glioma.
  • the compounds of the invention may be used in combination with one or more other therapeutic agents or therapies that act by the same or different mechanisms of action for the treatment of Trk-associated cancers.
  • the compounds of the invention are inhibitors of Trk kinase and are useful for treating, preventing or ameliorating a disease or disorder modulated or otherwise affected by one or more of Trk kinase domain mutants, or Ways to effectively treat, prevent or ameliorate one or more of its symptoms or causes.
  • the Trk kinase is selected from the group consisting of TrkA, TrkB, or TrkC.
  • Point mutations in the NTRK1 gene, the NTRK2 gene, and the NTRK3 gene were found in Trk inhibitor-resistant cancer cells. Point mutations in the NTRK1/2/3 gene can produce TrkA/B/C proteins, including amino acids in wild-type TrkA/B/C proteins that are substituted with different amino acids.
  • the compounds of the invention may be used to treat diseases mediated by at least one point mutation in the NTRK gene that results in the expression of the Trk protein.
  • the compounds of the invention are useful in the manufacture of a medicament for the treatment of a disease mediated by at least one point mutation in the NTRK gene that expresses the Trk protein.
  • At least one point mutation in the NTRK gene that results in expression of a Trk protein comprising a mutation at one or more amino acid positions can be selected from (i) at least one point mutation in the NTRK1 gene, which results in expression including TrkA proteins mutated at one or more amino acid positions selected from the group consisting of: 517, 542, 568, 573, 589, 595, 599, 600, 602, 646, 656, 657, 667, and 676, and/or ( Ii) at least one point mutation in the NTRK2 gene, which results in expression comprising a TrkB protein mutated at one or more amino acid positions selected from the group consisting of 545, 570, 596, 601, 617, 623, 624, 628, 630, 672, 682, 683, 693 and 702, and/or (iii) at least one point mutation in the NTRK3 gene, which results in expression of a TrkC protein comprising a mutation at one or more amino acid
  • the Trk protein comprises one or more of the following amino acid substitutions: G517R, A542V, V573M, F589L, F589C, G595S, G595R, D596V, D596V, F600L, F646V, C656Y, C656F, L657V, G667S, G667C And Y676S.
  • the TrkB protein comprises one or more of the following amino acid substitutions: G545R, A570V, Q596E, Q596P, V601G, F617L, F617C, F617I, G623S, G623R, D624V, R630K, C682Y, C682F, L683V, G693S And G713S.
  • the TrkC protein comprises one or more of the following amino acid substitutions: G545R, A570V, F617L, G623R, D624V, C685Y, C685F, L686VHE G696A.
  • the additional therapeutic agent is selected from the group consisting of: a therapeutic agent that targets a receptor tyrosine kinase, including cabozantinib, crizotinib, erlotinib, gefitinib, imatin Nipa, lapatinib, nilotinib, pazotinib, pertuzumab, regotinib, sunitinib and trastuzumab.
  • a therapeutic agent that targets a receptor tyrosine kinase including cabozantinib, crizotinib, erlotinib, gefitinib, imatin Nipa, lapatinib, nilotinib, pazotinib, pertuzumab, regotinib, sunitinib and trastuzumab.
  • the additional therapeutic agent is selected from a signal transduction pathway inhibitor, including, for example, a Ras-Raf-MEK-ERK pathway inhibitor (eg, sorafenib, trimetinib, or welluo) Fini), a PI3K-Akt-mTOR-S6K pathway inhibitor (eg, everolimus, rapamycin, perifosine, or sirolimus) and a modulator of the apoptotic pathway (eg, Obak) La (obataclax)).
  • a Ras-Raf-MEK-ERK pathway inhibitor eg, sorafenib, trimetinib, or welluo
  • Fini a PI3K-Akt-mTOR-S6K pathway inhibitor
  • a modulator of the apoptotic pathway eg, Obak) La (obataclax)
  • the additional therapeutic agent is selected from the group consisting of: a cytotoxic chemotherapeutic agent, including, for example, arsenic trioxide, bleomycin, cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, A Cytosine, dacarbazine, daunorubicin, docetaxel, doxorubicin, etoposide, fluorouracil, gemcitabine, irinotecan, lomustine, methotrexate, mitomycin C, Oxaliplatin, paclitaxel, pemetrexed, temozolomide and vincristine.
  • a cytotoxic chemotherapeutic agent including, for example, arsenic trioxide, bleomycin, cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, A Cytosine, dacarbazine, daunor
  • the additional therapeutic agent is selected from the group consisting of an angiogenesis-targeted therapy, including, for example, aboxicept and bevacizumab.
  • the additional therapeutic agent is selected from the group consisting of immunologically targeted agents, including, for example, aldesleukin, iprezumab, lammtuzumab, navizumab, and cyprus -T.
  • the additional therapeutic agent is selected from agents that are effective against the downstream Trk pathway, including, for example, biopharmaceuticals that target NGF, such as NGF antibodies and panTrk inhibitors.
  • the additional therapeutic agent or therapy is radiation therapy, including, for example, radioiodine therapy, external beam radiation, and radium 223 therapy.
  • the additional therapeutic agent comprises any of the therapies or therapeutic agents listed above, wherein the therapy or therapeutic agent is a disorder in which the cancer has a NTRK gene, a Trk protein, or an expression or activity or level thereof.
  • the therapy or therapeutic agent is a disorder in which the cancer has a NTRK gene, a Trk protein, or an expression or activity or level thereof.
  • the standard of care for cancer is any of the therapies or therapeutic agents listed above, wherein the therapy or therapeutic agent is a disorder in which the cancer has a NTRK gene, a Trk protein, or an expression or activity or level thereof.
  • the at least one additional therapeutic or therapeutic agent is selected from the group consisting of radiation therapy (eg, radioiodin therapy, external radiation, or radium 223 treatment), cytotoxic chemotherapeutic agents (eg, arsenic trioxide, bleomycin) , cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, cytarabine, dacarbazine, daunorubicin, docetaxel, doxorubicin, etoposide, fluorouracil, gemcitabine , irinotecan, lomustine, methotrexate, mitomycin C, oxaliplatin, paclitaxel, pemetrexed, temozolomide, or vincristine), ty
  • the additional therapeutic agent is a different Trk inhibitor.
  • Trk inhibitors include (R)-2-phenylpyrrolidine substituted imidazoxazine, AZD6918, GNF-4256, GTX-186, GNF-5837, AZ623, AG-879, altiratinib, CT327 , AR-772, AR-523, AR-786, AR-256, AR-618, AZ-23, AZD7451, cabozantinib, CEP-701, CEP-751, PHA-739358, dovetinib, entrectinib , PLX7486, GW441756, MGCD516, ONO-5390556, PHA-848125AC, regorafenib, sorafenib, sunitinib, TSR-011, VM-902A, K252a, 4-aminopyrazolylpyrimidine and substituted Pyrazolo[1,5-a]
  • These other therapeutic agents can be administered together with one or more of the compounds provided herein as part of the same or separate dosage forms via the same or different routes of administration and based on the same or different dosing schedules according to standards known to those skilled in the art. Drug practice is administered.
  • the compounds of the present invention have a number of advantages over non-deuterated compounds known in the art. Advantages of the present invention include: First, the compounds and compositions employing the technical solutions of the present invention provide a more favorable treatment for pain, cancer, inflammation, neurodegenerative diseases or certain infectious diseases, particularly Trk-related diseases. Therapeutic tools. Second, the metabolism of the compound in the organism is improved, giving the compound better pharmacokinetic parameter characteristics. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability. Third, the drug concentration of the compound in the animal is increased, and the drug efficacy is improved. Fourth, certain metabolites are inhibited and the safety of the compounds is increased.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C).
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
  • Example 1 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-13,13,14,14-d 4 (compound 13);
  • 3-Bromo-5-fluoro-2-methoxypyridine (3.09 g, 15 mmol) was dissolved in anhydrous THF (50 mL), and isopropyl magnesium chloride solution (7.0 mL, 14.0 mmol) was slowly added dropwise at 0 °C. After the dropwise addition, the mixture was warmed to 0 ° C and stirred for 1 hour, and then a solution of N-tert-butoxycarbonyl-2-pyrrolidone (1.85 g, 10.0 mmol) in anhydrous tetrahydrofuran (10 mL) was slowly added dropwise at -15 ° C, and stirred at room temperature for 30 min.
  • the compound 11 (224 mg, 0.52 mmol) was dissolved in 3 ml of methanol and 2 ml of water, lithium hydroxide (109.2 mg, 2.6 mmol) was added, the temperature was raised to 50 ° C, and the reaction was stirred for 4-6 h. After the reaction was completed by TLC, the mixture was cooled to room temperature. Dilute hydrochloric acid to adjust the pH to acidity, concentrate to remove the solvent, and directly put into the next reaction.
  • the racemic compound 13 was isolated using a chiral preparative column to give the compounds L-1-a and L-1-b.
  • Example 2 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-5,5,6-d 3 (compound 22);
  • the compound 15 (400 mg, 1.03 mmol) was dissolved in 4M aqueous hydrogen chloride (dichlorohexane) (8 ml, 32 mmol), and the mixture was heated to 110 ° C and stirred for 24 h. After the TLC detection reaction was completed, the solvent was concentrated and removed, and used directly in the next step.
  • 4M aqueous hydrogen chloride dichlorohexane
  • the compound 18 (289 mg, 0.52 mmol) was dissolved in a mixed solution of 5 ml of methanol and 5 ml of tetrahydrofuran, and a catalytic amount of Pd/C was added. The reaction was stirred at room temperature for 2-4 h. After TLC detection, the catalyst was removed by filtration. Concentrate to dryness and directly into the next reaction.
  • the compound 20 (223 mg, 0.52 mmol) was dissolved in 3 ml of methanol and 2 ml of water, and lithium hydroxide (109.2 mg, 2.6 mmol) was added thereto, and the mixture was heated to 50 ° C and stirred for 4-6 hours. After the reaction was completed by TLC, the mixture was cooled to room temperature. Dilute hydrochloric acid to adjust the pH to acidity, concentrate to remove the solvent, and directly put into the next reaction.
  • the racemic compound 22 was isolated using a chiral preparative column to give the compounds L-2-a and L-2-b.
  • Example 3 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-5,5,6,13,13,14,14-d 7 (compound 26);
  • the compound 24 (225.1 mg, 0.52 mmol) was dissolved in 3 ml of methanol and 2 ml of water, and lithium hydroxide (109.2 mg, 2.6 mmol) was added thereto, and the mixture was heated to 50 ° C and stirred for 4-6 hours. After the reaction was completed by TLC, the mixture was cooled to room temperature. The pH was adjusted to be acidic with dilute hydrochloric acid, and the solvent was concentrated to remove the mixture, which was directly applied to the next reaction.
  • the racemic compound 26 was isolated using a chiral preparative column to give the compounds L-3-a and L-3-b.
  • Example 4 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-4,4-d 2 (compound 40);
  • the succinimide (9.0 g, 90.9 mmol) was dissolved in 60 mL of deuterated methanol, potassium carbonate (1.44 g, 10.4 mmol) was added, and the mixture was stirred under microwave heating at 120 ° C for 30 min. The reaction mixture was concentrated, and the obtained solid was Compound 27, which was used directly to the next step.
  • the solid obtained in the above step was dispersed in 400 mL of anhydrous tetrahydrofuran, and lithium aluminum hydride (3.05 g, 80.3 mmol) was added portionwise in an ice bath, and the mixture was stirred for 15 min.
  • the reaction mixture was quenched with sodium sulfate decahydrate, filtered, and filtered, washed with ethyl acetate (200 mL), and the filtrate was concentrated and concentrated by column chromatography (PE/EA, 25% to 50%) to give 1.44 g of colorless oily liquid as compound 28 Directly invest in the next step.
  • the compound 30 (1.88 g, 5.97 mmol) was dissolved in toluene (20 mL), and 1.1 mL of concentrated hydrochloric acid was added, and the mixture was warmed to 65 ° C, and the reaction was stirred overnight, and the temperature was lowered to room temperature with 2M sodium hydroxide, and stirring was continued for 1 hour.
  • the reaction was completed by TLC.
  • the organic phase is separated, and the aqueous phase is extracted with ethyl acetate three times. The organic phase is combined, washed with brine, concentrated, and then purified by column chromatography.
  • the compound 33 (201 mg, 0.52 mmol) was dissolved in 4M hydrogen chloride dioxane (5 ml, 20 mmol), and the mixture was heated to 110 ° C and stirred for 24 h. After the TLC detection reaction was completed, the solvent was concentrated and removed, and used directly in the next step.
  • the racemic compound 40 was isolated using a chiral preparative column to give the compounds L-4-a and L-4-b.
  • Example 5 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-3,3-d 2 (compound 53);
  • the succinimide (3.0 g, 30.3 mmol) was dispersed in 130 mL of anhydrous tetrahydrofuran, and LiAlD 4 (1.02 g, 24.3 mmol) was added portionwise in an ice bath, and the mixture was stirred for 15 min.
  • the reaction mixture was quenched with sodium sulfate decahydrate, filtered, and the residue was washed with ethyl acetate (ethyl acetate), and the filtrate was concentrated and concentrated by column chromatography (PE/EA, 25% to 50%) to give 1.44 g of colorless oily liquid as compound 41 And go directly to the next step.
  • the compound 43 (1.87 g, 5.97 mmol) was dissolved in toluene (20 mL), and 1.1 mL of concentrated hydrochloric acid was added, and the mixture was warmed to 65 ° C, and the reaction was stirred overnight, and the mixture was adjusted to room temperature with 2M sodium hydroxide, and stirring was continued for 1 hour.
  • the reaction was completed by TLC.
  • the organic phase was separated, and the aqueous phase was extracted three times with ethyl acetate. The organic phase was combined, washed with brine, and concentrated.
  • the compound 50 (287 mg, 0.52 mmol) was dissolved in 10 ml of methanol, and hydrazine hydrate (130 mg, 2.6 mmol) was added thereto, and the mixture was heated to reflux for 1-2 h. After the reaction was completed by TLC, the solvent was concentrated and purified by column chromatography to give a pale yellow solid. That is, compound 51.
  • the compound 51 (224 mg, 0.52 mmol) was dissolved in 3 ml of methanol and 2 ml of water, lithium hydroxide (109.2 mg, 2.6 mmol) was added, and the mixture was heated to 50 ° C to stir the reaction for 4-6 h. After the reaction was completed by TLC, the mixture was cooled to room temperature. Dilute hydrochloric acid to adjust the pH to acidity, concentrate to remove the solvent, and directly put into the next reaction.
  • the racemic compound 53 was isolated using a chiral preparative column to give the compounds L-5-a and L-5-b.
  • Example 6 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-4,4,13,13,14,14-d 6 (compound 57);
  • the compound 55 (226 mg, 0.52 mmol) was dissolved in 3 ml of methanol and 2 ml of water, and lithium hydroxide (109.2 mg, 2.6 mmol) was added thereto. The mixture was heated to 50 ° C and stirred for 4-6 h. After the reaction was completed by TLC, the mixture was cooled to room temperature. Dilute hydrochloric acid to adjust the pH to acidity, concentrate to remove the solvent, and directly put into the next reaction.
  • the racemic compound 57 was isolated using a chiral preparative column to give the compounds L-6-a and L-6-b.
  • Example 7 (15R) -9- fluoro-15-methylerythromycin -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21, 25 ] Teflon -1(24),7,9,11,18(25),19,22-hepten-17-one-3,3,13,13,14,14-d 6 (compound 61);
  • the racemic compound 61 was isolated using a chiral preparative column to give the compounds L-7-a and L-7-b.
  • Example 8 9-difluoro-15 (meth -d 3) -2,11,16,20,21,24- dioxolane hexaazatetracyclo [16.5.2.0 2,6, 0 7,12, 0 21 ,25 ] 25 -alkane -1(24),7,9,11,18(25),19,22-hepten-17-one-15-d (compound 70);
  • the compound 66 (194 mg, 0.37 mmol) was dissolved in 10 ml of methanol, and a catalytic amount of Pd/C was added thereto. The reaction was carried out at room temperature for 2-4 hours after hydrogenation. After the reaction was completed by TLC, the catalyst was concentrated to remove the residue, and the filtrate was concentrated to dryness. The next step is to react.
  • the racemic compound 70 was isolated by a chiral preparative chromatography column to give compound compounds L-8-a, L-8-b, L-8-c, and L-8-d.
  • Test compounds were dissolved in DMSO to make a 20 mM stock solution. Compounds were diluted to 0.1 mM in DMSO (100 times the final concentration of the dilution) before use and diluted in 3 folds at 11 concentrations. Dilute to 4 times the final concentration of the dilution solution with the buffer.
  • the compounds of the present invention provides significant protein kinase inhibitory activity, typically have IC 50 values of less than 1nM.
  • the compound of the present invention exhibited strong inhibitory activity against TRKA/B/C as compared with the compound ⁇ which was not deuterated and the compound ⁇ -a.
  • RPMI-1640 medium (GIBCO, catalog number A10491-01), fetal bovine serum (GIBCO, catalog number 10099141), antibiotic (Penicillin-Streptomycin), IL-3 (PeproTech), puromycin; living cells
  • the assay kit CellTiter-Glo4 (Promega, Cat. No. G7572), 96-well black-wall clear flat bottom cell culture plate (Corning, Cat. No. 3340).
  • Example compound KM12IC 50 (nM) Compound ⁇ -a 2.56 Compound L-2-a 2.51
  • the compounds of the present invention all exhibited better properties for inhibiting the proliferation of KM12 cells.
  • Microsomal experiments human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate buffer Agent (pH 7.4).
  • phosphate buffer 100 mM, pH 7.4.
  • the pH of the solution was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
  • NADPH regeneration system containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride was prepared and placed on wet ice before use.
  • Formulation stop solution acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. 25057.5 ⁇ L of phosphate buffer (pH 7.4) was taken into a 50 mL centrifuge tube, and 812.5 ⁇ L of SD rat liver microsomes were added and mixed to obtain a liver microsome dilution having a protein concentration of 0.625 mg/mL.
  • the corresponding compound had a reaction concentration of 1 ⁇ M and a protein concentration of 0.5 mg/mL.
  • 100 ⁇ L of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper, and the reaction was terminated by vortexing for 3 min.
  • the plate was centrifuged at 5000 x g for 10 min at 4 °C.
  • 100 ⁇ L of the supernatant was taken into a 96-well plate to which 100 ⁇ L of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
  • the metabolic stability of human and rat liver microsomes was evaluated by simultaneously testing the compounds of the present invention and their compounds without deuteration.
  • the undeuterated compound LOXO-195 was used as a control.
  • the compounds of the invention significantly improved metabolic stability by comparison with the undeuterated compound LOXO-195.
  • the results of liver microsome experiments of human and rat of representative example compounds are shown in Table 3 below:
  • Rats were fed a standard diet and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the eyelids at a time point of 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
  • Rats were briefly anesthetized after inhalation of ether, and 300 ⁇ L of blood samples were collected from the eyelids in test tubes. There was 30 ⁇ L of 1% heparin salt solution in the test tube. The tubes were dried overnight at 60 ° C before use. After the blood sample collection was completed at the last time point, the rats were anesthetized with ether and sacrificed.
  • Plasma samples were centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate plasma from red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube, indicating the name and time of the compound. Plasma was stored at -80 °C prior to analysis. The concentration of the compound of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.

Abstract

本发明提供了一种取代的吡唑并[1,5-a]嘧啶类大环化合物的药物组合物及其用途,所述的吡唑并[1,5-a]嘧啶类大环化合物如式(Aa)所示化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体。本发明化合物为Trk激酶的抑制剂,并且可以用于治疗疼痛、癌症、炎症、神经退行性疾病和某些传染病。

Description

取代的吡唑并[1,5-a]嘧啶类的大环化合物 技术领域
本发明属于医药技术领域,尤其涉及一种取代的吡唑并[1,5-a]嘧啶类的大环化合物及包含该化合物的组合物及其用途。更具体而言,本发明涉及某些氘取代的9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷-1(24),7,9,11,18(25),19,22-七烯-17-酮的化合物及其立体异构体,这些氘取代的化合物显示Trk家族蛋白酪氨酸激酶抑制,且可用于治疗疼痛、炎症、癌症和某些传染病的用途,且这些氘取代的化合物具有更优良的药代动力学性质。
背景技术
Trk是由成为神经营养因子(NT)的一组可溶性生长因子激活的高亲和性受体酪氨酸激酶。Trk受体家族具有3个成员,即TrkA、TrkB和TrkC。神经营养因子中有(1)可激活TrkA的神经生长因子(NGF),(2)可激活TrkB的脑源性神经营养因子(BDNF)和NT-4/5,和(3)可激活TrkC的NT3。Trk在神经元组织中广泛表达且与神经元细胞的维持、信号传导和存活有关。
文献也显示Trk的过度表达、激活、扩增和/或突变与许多癌症有关,该癌症包括神经细胞瘤、卵巢癌、乳腺癌、***癌、胰腺癌、多发性骨髓瘤、星形细胞瘤与成神经管细胞瘤、神经胶质瘤、黑素瘤、甲状腺癌、胰腺癌、大细胞神经内分泌瘤和结肠直肠癌。此外,已证明Trk/神经营养因子途径的抑制剂在治疗疼痛和炎性疾病的多种临床前动物模型中有效。
神经营养因子/Trk途径,特别是BDNF/TrkB途径也已经牵涉神经变性疾病的病因,该疾病包括多发性硬化、帕金森金氏(Parkinson’s disease)和阿尔茨海默病(Alzheimer’s disease)。调节神经营养因子/Trk途径可用于治疗这些疾病和相关疾病。
据认为TrkA受体对于克氏锥虫(查加斯氏病)在人类宿主中的寄生虫感染中的疾病过程至关重要。因此,TrkA抑制剂可用于治疗查加斯氏病和有关的原生动物感染。
Trk抑制剂也可用于治疗与骨重建调节失衡有关的疾病,例如骨质疏松症、类风湿性关节炎和骨转移。骨转移是癌症的常见的并发症,在晚期乳腺癌或***癌患者中高达70%和在肺癌、结肠癌、胃癌、膀胱癌、子宫癌、直肠癌、甲状腺癌或肾癌患者中为约15至30%。蓉骨性转移可造成严重的疼痛、病理性骨折、危及生命的高钙血症、脊髓压迫症和其它神经压迫综合征。出于这些原因,骨 转移是花费高的严重的癌症并发症。因此,可诱导增殖性骨细胞凋亡的药剂是非常有优势的。已在骨折的小鼠模型的成骨区域中观察到TrkA受体和TrkC受体的表达。另外,在几乎所有的成骨细胞凋亡的药剂是非常有优势的。已在骨折的小鼠模型的成骨区域中观察到TrkA受体和TrkC受体的表达。另外,在几乎所有的成骨细胞中均观察到NGF的定位。最近,证明在人类hFOB成骨细胞中pan-Trk抑制剂可抑制由与所有3个Trk受体结合的神经营养因子所激活的酪氨酸信号传导。该数据支持使用Trk抑制剂治疗骨重建疾病(例如癌症患者中的骨转移)的理论。
Larotrectinib(LOXO-101)由Loxo Oncology公司研发的第一代Trk抑制剂,LOXO-101在2015年3月开始治疗第一名患者;2016年7月13日被FDA授予突破性药物资格,用于Trk融合基因突变阳性的成人及儿童的不可手术切除或转移性实体瘤;2017年2月完成关键入组。但是在接受Larotrectinib抑制剂治疗后,癌症患者的Trk基因可能产生一些突变(如NTRK1 G595R,NTRK3 G623R等位点的突变),导致耐药性的产生。LOXO-195(化学名称为(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷-1(24),7,9,11,18(25),19,22-七烯-17-酮,具有如下结构式)是Loxo Oncology公司研发的第二代Trk抑制剂,能有效抵制Larotrectinib产生的耐药性。已证明一名结直肠癌的成年患者和纤维肉瘤的儿童患者产生Larotrectinib耐药性后,接受LOXO-195来治疗能够延长Trk基因突变患者的病程,患者都有缓释效应,且几乎没有副作用(Drilon.A.,et al.,Cancer Discov.2017,7(9),1-10)。目前,FDA已经正式批准LOXO-195作为实验性新药,开展临床I期和II期试验。
Figure PCTCN2019072833-appb-000001
已知较差的吸收、分布、代谢和/或***(ADME)性质是导致许多候选药物临床试验失败的主要原因。当前上市的许多药物也由于较差的ADME性质限制了它们的应用范围。药物的快速代谢会导致许多本来可以高效治疗疾病的药物由于过快的从体内代谢清除掉而难以成药。频繁或高剂量服药虽然有可能解决药物快速清除的问题,但该方法会带来诸如病人依从性差、高剂量服药引起的副作用及治疗成本上升等问题。另外,快速代谢的药物也可能会使患者暴露于不良的毒性或反应性代谢物中。
虽然LOXO-195作为Trk抑制剂能有效治疗多种癌症等病症,但是发现具有治疗癌症等病症且 具有很好的口服生物利用度且有成药性的新型化合物还是具有挑战性的工作。因此,本领域仍需开发对适用作治疗剂的Trk激酶介导疾病具有选择性抑制活性或更好地药效学/药代动力学的化合物,本发明提供了这样的化合物。
发明概述
针对以上技术问题,本发明公开了一种新的氘取代的9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷-1(24),7,9,11,18(25),19,22-七烯-17-酮及其立体异构体(例如,化合物Φ,化合物Φ-a和化合物Φ-b,结构式如下),及其组合物和用途,其具有更好地Trk激酶抑制活性、更低的副作用、更好地药效学/药代动力学性能,可用于治疗Trk激酶介导的疾病。
Figure PCTCN2019072833-appb-000002
如本文所用,术语“本发明化合物”指式(A)、式(A-1)、式(A-2)、式(Aa)、式(Aa-1)、式(Aa-2)、式(I)、式(II)、式(III)、式(IV)、式(Ia)、式(IIa)、式(IIIa)和式(IVa)所示的化合物。该术语还包括式(A)、式(A-1)、式(A-2)、式(Aa)、式(Aa-1)、式(Aa-2)、式(I)、式(II)、式(III)、式(IV)、式(Ia)、式(IIa)、式(IIIa)和式(IVa)化合物的药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体。
对此,本发明采用以下技术方案:
本发明的第一方面,提供了式(Aa)化合物:
Figure PCTCN2019072833-appb-000003
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12中至少一个是氘;
或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体。
本发明的另一方面,提供了式(I))化合物:
Figure PCTCN2019072833-appb-000004
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5中至少一个是氘;
或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体。
在另一方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物。在具体实施方案中,本发明化合物以有效量提供在所述药物组合物中。在具体实施方案中,本发明化合物 以治疗有效量提供。在具体实施方案中,本发明化合物以预防有效量提供。
在另一方面,本发明提供了一种如上所述的药物组合物的制备方法,包括以下步骤:将药学上可接受的赋形剂与本发明化合物进行混合,从而形成药物组合物。
在另一方面,本发明还涉及提供一种在受试者中治疗由Trk激酶介导的疾病的方法。该方法包括向该受试者给药治疗有效量的本发明化合物。在具体实施方案中,所述癌症通过TrkA;TrkB;或TrkA和TrkB介导。在具体实施方案中,患者诊断或鉴定为患有Trk-相关的癌症。在具体实施方案中,口服、皮下、静脉内或肌肉内给药所述化合物。在具体实施方案中,长期给药所述化合物。在具体的实施方案中,Trk激酶介导的疾病选自疼痛、癌症、炎症、神经退行性疾病或锥虫感染。
由随后的具体实施方式、实施例和权利要求,本发明的其它目的和优点将对于本领域技术人员显而易见。
发明详述
定义
本文中,如无特别说明,“氘代”指化合物或基团中的一个或多个氢被氘所取代;氘代可以是一取代、二取代、多取代或全取代。术语“一个或多个氘代的”与“一次或多次氘代”可互换使用。
本文中,如无特别说明,“非氘代的化合物”是指含氘原子比例不高于天然氘同位素含量(0.015%)的化合物。
术语“药学上可接受的盐”是指,在可靠的医学判断范围内,适合与人和低等动物的组织接触而没有过度毒性、刺激性、***反应等等,并且与合理的益处/危险比例相称的那些盐。药学上可接受的盐在本领域是众所周知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1-19中详细描述的药学上可接受的盐。本发明化合物的药学上可接受的盐包括衍生自合适无机和有机酸和碱的盐。
本发明还包括同位素标记的化合物,等同于原始化合物在此公开。可以列为本发明的化合物同位素的例子包括氢,碳,氮,氧,磷,硫,氟和氯同位素,分别如 2H, 3H, 13C, 14C, 15N, 17O, 18O, 31P, 32P, 35S, 18F以及 36Cl。本发明中的化合物,或对映体,非对映体,异构体,或药学上可接受的盐或溶剂化物,其中含有上述化合物的同位素或其他其他同位素原子都在本发明的范围之内。本发明中某些同位素标记化合物,例如 3H和 14C的放射性同位素也在其中,在药物和底物的组织分布实验中是有用的。氚,即 3H和碳14,即 14C,它们的制备和检测比较容易,是同位素中的首选。同位素标记的化合物可以用一般的方法,通过用易得的同位素标记试剂替换为非同位素的试剂,用示例中的方案可以制备。
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种“立体异构体”形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本发明化合物可以是无定形或结晶形式。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“晶型”是指化学药物分子的不同排列方式,一般表现为药物原料在固体状态下的存在形式。一种药物可以多种晶型物质状态存在,同一种药物的不同晶型,在体内的溶解和吸收可能不同,从而会对制剂的溶出和释放产生影响。
术语“溶剂化合物”指本发明化合物与溶剂分子配位形成特定比例的配合物。“水合物”指本发明化合物与水进行配位形成的配合物。
术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)本发明化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
术语“晶型”是指化学药物分子的不同排列方式,一般表现为药物原料在固体状态下的存在形式。一种药物可以多种晶型物质状态存在,同一种药物的不同晶型,在体内的溶解和吸收可能不同,从 而会对制剂的溶出和释放产生影响。
如本文所用,术语“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在另一些实施方案中,受试者是非人动物。
“疾病”、“障碍”和“病症”在本文中可以互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括受试者开始患有具体疾病、障碍或疾病之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药物动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗和预防性治疗有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗有益处的数量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化。化合物的治疗有效量是指单独使用或与其他疗法联用的治疗剂的数量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其他治疗剂的治疗效能的数量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的数量,或足以预防与疾病、障碍或病症有关的一或多种症状的数量,或防止疾病、障碍或病症复发的数量。化合物的预防有效量是指单独使用或与其它药剂联用的治疗剂的数量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的数量,或增强其它预防药剂的预防效能的数量。
“组合”以及相关术语是指同时或依次给药本发明的治疗剂。例如,本发明化合物可以与另一治疗剂以分开的单位剂型同时或依次给药,或与另一治疗剂一起呈单一单位剂型同时给药。
具体实施方式
化合物
在一个实施方案中,本发明提供了式(A)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000005
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5中至少一个是氘。
在一个具体实施方案中,“R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘”的技术方案包括R 1选自氢或氘、R 2选自氢或氘、R 3选自氢或氘,以此类推,直至R 12选自氢或氘的技术方案;更具体地,包括R 1为氢或R 1为氘、R 2为氢或R 2为氘、R 3为氢或R 3为氘,以此类推,直至R 12为氢或R 12为氘的技术方案。在另一个具体实施方案中,“Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘”的技术方案包括Y 1选自氢或氘、Y 2选自氢或氘、Y 3选自氢或氘,以此类推,直至Y 5选自氢或氘的技术方案;更具体地,包括Y 1为氢或Y 1为氘、Y 2为氢或Y 2为氘、Y 3为氢或Y 3为氘,以此类推,直至Y 5为氢或Y 5为氘的技术方案。
在另一个具体实施方案中,“X选自CH 3、CD 3、CHD 2或CH 2D”的技术方案包括X为CH 3、X为CD 3、X为CHD 2或X为CH 2D的技术方案。
在一个具体实施方案中,本发明涉及式(A-1)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000006
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5中至少一个是氘。
在另一个具体实施方案中,本发明涉及式(I)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000007
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5中至少一个是氘。
在另一个具体实施方案中,本发明涉及式(II)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000008
其中,
R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘;
X选自CH 3、CD 3、CHD 2或CH 2D;
条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5中至少一个是氘。
作为本发明的优选实施方案,式(A)、式(A-1)、式(I)和式(II)的化合物中至少含有一个氘原子,更佳地一个氘原子,更佳地二个氘原子,更佳地三个氘原子,更佳地四个氘原子,更佳地五个氘原子,更佳地六个氘原子,更佳地七个氘原子,更佳地八个氘原子,更佳地九个氘原子,更佳地十个氘原子,更佳地十一个氘原子,更佳地十二个氘原子,更佳地十三个氘原子,更佳地十四个氘原子,更佳地十五个氘原子。
作为本发明的优选实施方案,氘在氘代位置的氘同位素含量至少是大于天然氘同位素含量0.015%,较佳地大于30%,更佳地大于50%,更佳地大于75%,更佳地大于95%,更佳地大于99%。
具体地说,在本发明中R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4、Y 5和X,各氘代位置中氘同位素含量至少是5%,较佳地大于10%,更佳地大于15%,更佳地大于20%,更佳地大于25%,更佳地大于30%,更佳地大于35%,更佳地大于40%,更佳地大于45%,更佳地大于50%,更佳地大于55%,更佳地大于60%,更佳地大于65%,更佳地大于70%,更佳地大于75%,更佳地大于80%,更佳地大于85%,更佳地大于90%,更佳地大于95%,更佳地大于99%。
在另一具体实施方案中,式(A)、式(A-1)、式(I)和式(II)化合物的R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4、Y 5和X,至少其中一个含氘,更佳地两个含氘,更佳地三个含氘,更佳地四个含氘,更佳地五个含氘,更佳地六个含氘,更佳地七个含氘,更佳地八个含氘,更佳地九个含氘,更佳地十个含氘,更佳地十一个含氘,更佳地十二个含氘,更佳地十三个含氘,更佳地十四个含氘,更佳地十五个含氘,更佳地十六个含氘,更佳地十七个含氘,更佳地十八个含氘,更佳地十九个含氘,更佳地二十个含氘。具体而言,式(A)、式(A-1)、式(I)和式(II)中化合物至少含有一个、两个、三个、四个、五个、六个、七个、八个、九个、十个、十一个、十二个、十三个、十四个、十五个、十六个、十七个、十八个、十九个、二十个氘原子。
作为本发明的具体实施方案,R 1选自氢或氘。
在另一具体实施方案中,R 1是氢。
在另一具体实施方案中,R 1是氘。
作为本发明的具体实施方案,R 2、R 3、R 4、R 5各自独立地选自氢或氘。
在另一具体实施方案中,R 2和R 3是相同的。
在另一具体实施方案中,R 4和R 5是相同的。
在另一具体实施方案中,R 2和R 3均为氘。
在另一具体实施方案中,R 2和R 3均为氢。
在另一具体实施方案中,R 4和R 5均为氘。
在另一具体实施方案中,R 4和R 5均为氢。
在另一具体实施方案中,R 2、R 3、R 4和R 5均为氘。
在另一具体实施方案中,R 2、R 3、R 4和R 5均为氢。
作为本发明的具体实施方案,R 6、R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自氢或氘。
在另一具体实施方案中,R 7和R 8是相同的。
在另一具体实施方案中,R 9和R 10是相同的。
在另一具体实施方案中,R 11和R 12是相同的。
在另一具体实施方案中,R 6、R 7和R 8均为氘。
在另一具体实施方案中,R 6、R 7和R 8均为氢。
在另一具体实施方案中,R 9和R 10均为氘。
在另一具体实施方案中,R 9和R 10均为氢。
在另一具体实施方案中,R 11和R 12均为氘。
在另一具体实施方案中,R 11和R 12均为氢。
作为本发明的具体实施方案,X选自CH 3、CD 3、CHD 2或CH 2D。
在另一具体实施方案中,X选自CH 3或CD 3
作为本发明的具体实施方案,Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘。
作为本发明的具体实施方案,X为CD 3,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘。
在另一具体实施方案中,X为CD 3,Y 1、Y 2、Y 3、Y 4和Y 5均为氢,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘。
在另一具体实施方案中,X为CD 3,R 1、Y 1、Y 2、Y 3、Y 4和Y 5均为氢,R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘。
在另一具体实施方案中,X为CD 3,R 1、R 2、R 3、R 4、R 5、Y 1、Y 2、Y 3、Y 4和Y 5均为氢,R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘。
作为本发明的具体实施方案,R 2、R 3、R 4和R 5均为氘,R 1、R 6、R 7、R 8、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,R 2、R 3、R 4、R 5、R 6、R 7、R 8均为氘,R 1、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,R 2、R 3、R 4、R 5、R 9和R 10均为氘,R 6、R 7、R 8、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,R 2、R 3、R 4、R 5、R 11和R 12均为氘,R 6、R 7、R 8、R 9、R 10、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
作为本发明的具体实施方案,R 6、R 7和R 8均为氘,R 1、R 2、R 3、R 4、R 5、R 9、R 10、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
作为本发明的具体实施方案,R 9和R 10均为氘,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 11、R 12、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
作为本发明的具体实施方案,R 11和R 12均为氘,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、Y 1、Y 2、Y 3、Y 4和Y 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一个实施方案中,本发明涉及式(Aa)化合物,或其药学上可接受的盐、前药、水合物或溶 剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000009
其中,R 1-R 12和X如上文所定义。
在另一个实施方案中,本发明涉及式(Aa-1)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000010
其中,R 1-R 12和X如上文所定义。
在另一个具体实施方案中,本发明还涉及式(Ia)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000011
其中,R 1-R 12和X如上文所定义。
在另一个具体实施方案中,本发明还涉及式(IIa)化合物,或其药学上可接受的盐、前药、水合 物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000012
其中,R 1-R 12和X如上文所定义。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 11和R 12选自氢,R 1-R 10各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 9和R 10选自氢,R 1-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 9-R 12选自氢,R 1-R 8各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1选自H,R 2-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1、R 11和R 12选自氢,R 2-R 10各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1、R 9和R 10选自氢,R 2-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1、R 9-R 12选自氢,R 2-R 8各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 1-R 12各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 11和R 12选自氢,R 1-R 10各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 9和R 10选自氢,R 1-R 8和R 11-R 12各自独立地选自氢或氘附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 9-R 12选自氢,R 1-R 8各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 1选自H,R 2-R 12各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 1、R 11和R 12选自H,R 2-R 10各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 1、R 9和R 10选自H,R 2-R 8和R 11-R 12各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,X选自CH 3,R 1、R 9-R 12选自H,R 2-R 8各自独立地选自氢或氘,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5选自氢,R 1和R 6-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 11-R 12选自氢,R 1和R 6-R 10各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 9-R 10选自氢,R 1、R 6-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 9-R 12选自氢,R 1、R 6-R 8各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5选自氢,R 6-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 11-R 12选自氢,R 6-R 10各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 9-R 10选自氢,R 6-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 9-R 12选自氢,R 6-R 8各自独立地选自氢或氘,X选自CH 3或CD 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5选自氢,R 1和R 6-R 12各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 11-R 12选自氢,R 1和R 6-R 10各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 9-R 10选自氢,R 1、R 6-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 2-R 5和R 9-R 12选自氢,R 1、R 6-R 8各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5选自氢,R 6-R 12各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 11-R 12选自氢,R 6-R 10各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 9-R 10选自氢,R 6-R 8和R 11-R 12各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含 有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1-R 5和R 9-R 12选自氢,R 6-R 8各自独立地选自氢或氘,X选自CH 3,附加条件是上述化合物至少含有一个氘。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 1-R 5和R 9-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 11-R 12选自氢,R 1-R 5和R 9-R 10各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 9-R 10选自氢,R 1-R 5和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 9-R 12选自氢,R 1-R 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5选自氢,R 1和R 9-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5和R 11-R 12选自氢,R 1和R 9-R 10各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5和R 9-R 10选自氢,R 1和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5和R 9-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 11-R 12选自氢,R 2-R 5和R 9-R 10各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 9-R 10选自氢,R 2-R 5和R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 9-R 12选自氢,R 2-R 5各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5选自氢,R 9-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5和R 11-R 12选自氢,R 9-R 10各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5和R 9-R 10选自氢,R 11-R 12各自独立地选自氢或氘,X选自CH 3或CD 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 1-R 5和R 9-R 12各自独立地选自氢或氘,X选自CH 3,。
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 11-R 12选自氢,R 1-R 5和R 9-R 10各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 9-R 10选自氢,R 1-R 5和R 11-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 9-R 12选自氢,R 1-R 5各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5选自氢,R 1和R 9-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5和R 11-R 12选自氢,R 1和R 9-R 10各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 6-R 8选自氘,R 2-R 5和R 9-R 10选自氢,R 1和R 11-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5和R 9-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 11-R 12选自氢,R 2-R 5和R 9-R 10各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 9-R 10选自氢,R 2-R 5和R 11-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 9-R 12选自氢,R 2-R 5各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5选自氢,R 9-R 12各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8选自氘,R 2-R 5和R 11-R 12选自氢,R 9-R 10各自独立地选自氢或氘,X选自CH 3
在另一具体实施方案中,本发明提供式(Aa)、式(Aa-1)、式(Ia)和式(IIa)化合物,其中,R 1和R 6-R 8 选自氘,R 2-R 5和R 9-R 10选自氢,R 11-R 12各自独立地选自氢或氘,X选自CH 3
在另一个实施方案中,本发明涉及式(Aa-2)、式(IIIa)和式(IVa)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
Figure PCTCN2019072833-appb-000013
其中,R 1-R 12、Y 1-Y 5和X如上文所定义。
作为本发明的优选实施方案中,所述化合物选自下组化合物:
Figure PCTCN2019072833-appb-000014
Figure PCTCN2019072833-appb-000015
Figure PCTCN2019072833-appb-000016
作为本发明的优选实施方案中,所述化合物不包括非氘代化合物。
药物组合物和施用方法
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的活性组分。在一些实施方案中,所述药物组合物包含治疗有效量的活性组分。在一些实施方案中,所述药物组合物包含预防有效量的活性组分。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有0.5-2000mg本发明化合物/剂,更佳地,含有1-500mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可接受的赋形剂”是指不会破坏一起调配的化合物的药理学活性的无毒载体、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载体、佐剂或媒剂包括(但不限于)离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明的药物组合物可通过将本发明的化合物与适宜的药学上可接受的赋形剂组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、溶液剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本发明的化合物或其药物组合物的典型途径包括但不限于口服、直肠、透黏膜、经肠给药,或者局部、经皮、吸入、肠胃外、舌下、***内、鼻内、眼内、抚摸内、肌内、皮下、静脉内给药。
本发明的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等等。
对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的赋形剂混合来配置该药物组合物。这些赋形剂能使本发明的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述的活性化合物与固体赋形剂混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅剂,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂、甜味剂或矫味剂等。如微晶纤维素、葡萄糖溶液、***胶浆、明胶溶液、蔗糖和淀粉糊;滑石、淀粉、硬脂酸钙或硬脂酸;乳糖、蔗糖、淀粉、甘露糖醇、山梨糖醇或磷酸二钙;二氧化硅;交联羟甲基纤维素钠、预交化淀粉、淀粉羟乙酸钠、藻酸、玉米淀粉、马铃薯淀粉、甲基纤维素、琼脂、羟甲基纤维素、交联聚乙烯吡咯烷酮等。可以根据通常药物实践中公知的方法任选地对糖衣剂的核心进行包衣,尤其使用肠溶包衣。
药物组合物还可适用于肠胃外给药,如合适的单位剂型的无菌溶液剂、混悬剂或冻干产品。能够使用适当的赋形剂,例如填充剂、缓冲剂或表面活性剂。
本发明化合物可以通过任何使用途径和方法给药,例如通过口服或肠胃外(例如,静脉内)给药。本发明化合物的治疗有效量为从约0.0001到20mg/kg体重/天,例如从0.001到10mg/kg体重/天。
本发明化合物的剂量频率由患者个体的需求决定,例如,每天1次或2次,或每天更多次。给药可以是间歇性的,例如,其中在若干天的期间内,患者接受本发明化合物的每日剂量,接着在若干天或更多天的期间,患者不接受式本发明化合物的每日剂量。
本发明化合物的治疗适应症
本发明化合物展现Trk家族蛋白酪氨酸激酶抑制作用,且该化合物可用于治疗疼痛、癌症、炎症、神经退行性疾病或锥虫感染等。
一些实施方案包括本发明化合物用于治疗可通过抑制TrkA、TrkB、和/TrkC激酶治疗的病症及疾病(例如TrkA、TrkB和/或TrkC介导的病症,例如本文所述的一种或多种病症,包括Trk相关的癌症)的用途。在一些实施方案中,本发明化合物也可用于治疗疼痛(包括慢性及急性疼痛)。在一些实施方案中,本发明化合物可用于治疗多种类型的疼痛、神经性疼痛、手术疼痛及与癌症、手术及骨折相关的疼痛。
在一些实施方案中,本发明提供了治疗或预防受试者炎症的方法,该方法包括向包括向受试者给药治疗有效量的本发明化合物。在一个实施方案中,所述的方法包括治疗受试者所述炎症的方法。在一个实施方案中,所述的方法包括预防受试者所述炎症的方法。
在一些实施方案中,本发明提供了治疗受试者神经退行性疾病的方法,该方法包括向受试者给药治疗有效量的本发明化合物。在一个实施方案中,所述的神经退行性疾病为脱髓鞘疾病。在一个实施方案中,所述的神经退行性疾病为髓鞘形成障碍。在一个实施方案中,所述的神经退行性疾病为多发性硬化症。在一个实施方案中,所述的神经退行性疾病为Parkinson疾病。在一个实施方案中,所述的神经退行性疾病为Alzheimer疾病。
在一些实施方案中,本发明提供了治疗受试者传染病的方法,该方法包括向受试者给药治疗有效量的本发明化合物。在一个实施方案中,所述的传染病为锥虫感染。
在一些实施方案中,本文提供了治疗诊断患有Trk-相关的癌症的患者的方法,包括向患者给药治疗有效量的本发明化合物。例如,Trk-相关的癌症可选自:非小细胞肺癌、***状甲状腺癌、多形性成胶质细胞瘤、急性髓细胞性白血病、结肠直肠癌、大细胞神经内分泌癌、***癌、结肠癌、急性髓细胞性白血病、肉瘤、小儿神经胶质瘤、肝内胆管癌、毛细胞性星形细胞瘤、低级神经胶质瘤、肺腺癌、唾液腺癌、分泌型乳腺癌、纤维肉瘤、肾瘤和乳腺癌。
在一些实施方案中,Trk-相关的癌症选自:TRK-相关的癌症的非限制性实例包括:Spitzoid黑素瘤、Spitz肿瘤(例如,转移性Spitz肿瘤)、非小细胞肺癌(NSCLC)、甲状腺癌(例如,***状 甲状腺瘤(PTC)、急性髓细胞性白血病(AML)、肉瘤(例如,未分化肉瘤或成人软组织肉瘤)、小儿神经胶质瘤、结肠直肠癌(CRC)、多形性成胶质细胞瘤(GBM)、大细胞神经内分泌癌症(LCNEC)、甲状腺癌、肝内胆管癌(LCC)、毛细胞性星形细胞瘤、低级神经胶质瘤、头颈部鳞状上皮细胞癌、肾瘤、黑素瘤、支气管癌、B-细胞癌症、Bronchus癌症、口腔或咽部癌症、血液组织癌、子***、胃癌、肾癌、肝癌、多发性骨髓瘤、卵巢癌、胰腺癌、唾液腺癌、小肠或阑尾癌症、睾丸癌、泌尿膀胱癌、小细胞肺癌、炎性肌纤维母细胞癌、胃肠道间质瘤、非霍奇金淋巴瘤、成神经细胞瘤、小细胞肺癌、鳞状上皮细胞癌、食管-胃癌、皮肤癌、赘瘤(例如,黑色素细胞赘瘤)、Spitz痣、星形细胞瘤、成神经管细胞瘤、神经胶质瘤、大细胞神经内分泌肿瘤、骨癌和直肠癌。
在一些实施方案中,本发明化合物可用于治疗小儿患者的Trk相关的癌症。例如,本文所提供的化合物可用于治疗婴儿型肉瘤、成神经细胞瘤、先天性中胚层肾瘤、脑低分级神经胶质瘤及脑桥神经胶质瘤。
在一些实施方案中,本发明化合物可与一种或多种通过相同或不同的作用机制起作用的其他治疗剂或疗法组合用于治疗Trk相关的癌症。
在一些实施方案中,本发明化合物是Trk激酶的抑制剂,并且可用于治疗、预防或改善由Trk激酶结构域突变体中一个或多个调节或以其它方式影响的疾病或障碍,或以其它方式有效地治疗、预防或改善其一种或多种症状或病因。
在一些实施方案中,所述的Trk激酶选自TrkA、TrkB或TrkC。
人们在Trk抑制剂抗性癌细胞中发现了NTRK1基因、NTRK2基因、NTRK3基因中的点突变。NTRK1/2/3基因中的点突变可以产生TrkA/B/C蛋白,其包括野生型TrkA/B/C蛋白中的氨基酸被不同氨基酸取代。
本发明化合物可以用于治疗由导致表达Trk蛋白的NTRK基因中至少一个点突变介导的疾病。
本发明化合物可用于制备治疗由导致表达Trk蛋白的NTRK基因中至少一个点突变介导的疾病的药物中的用途。
在一些实施方案中,导致表达Trk蛋白(其包括位于一个或多个氨基酸位置处的突变)的NTRK基因中至少一个点突变可以选自(i)NTRK1基因中至少一个点突变,其导致表达包括在选自下组的一个或多个氨基酸位置处突变的TrkA蛋白:517、542、568、573、589、595、599、600、602、646、656、657、667和676,和/或(ii)NTRK2基因中至少一个点突变,其导致表达包括在选自下组的一个或多个氨基酸位置处突变的TrkB蛋白:545、570、596、601、617、623、624、628、630、672、682、683、693和702,和/或(iii)NTRK3基因中的至少一个点突变,其导致表达包括选自下组的一个或多个氨基酸位置处突变的TrkC蛋白:545、570、596、601、617、623、624、628、630、675、685、 686、696和705。在另一些实施方案中,Trk蛋白包括一个或多个下述氨基酸取代:G517R、A542V、V573M、F589L、F589C、G595S、G595R、D596V、D596V、F600L、F646V、C656Y、C656F、L657V、G667S、G667C和Y676S。在另一些实施方案中,TrkB蛋白包括一个或多个下述氨基酸取代:G545R、A570V、Q596E、Q596P、V601G、F617L、F617C、F617I、G623S、G623R、D624V、R630K、C682Y、C682F、L683V、G693S和G713S。在一些实施方案中,TrkC蛋白包括一个或多个下述氨基酸取代:G545R、A570V、F617L、G623R、D624V、C685Y、C685F、L686VHE G696A。在一些实施方案中,所述其它治疗剂选自:靶向受体酪氨酸激酶的治疗剂,包括卡博替尼、克唑替尼、埃罗替尼、吉非替尼、伊马替尼、拉帕替尼、尼洛替尼、帕唑替尼、帕妥珠单抗、瑞戈替尼、舒尼替尼和曲妥单抗。
在一些实施方案中,所述其它治疗剂选自信号转导途径抑制剂,包括,例如,Ras-Raf-MEK-ERK途径抑制剂(例如,索拉非尼,曲美替尼,或威罗非尼),PI3K-Akt-mTOR-S6K途径抑制剂(例如,依维莫司、雷帕霉素、哌立福辛,或西罗莫司)和凋亡途径的调节剂(例如,奥巴克拉(obataclax))。
在一些实施方案中,所述其它治疗剂选自:细胞毒素化疗剂,包括,例如,三氧化二砷、博来霉素、卡巴他赛、卡培他滨、卡铂、顺铂、环磷酰胺、阿糖胞苷、达卡巴嗪、柔红霉素、多西紫杉醇、多柔比星、依托泊苷、氟尿嘧啶、吉西他滨、伊立替康、洛莫司汀、甲氨碟呤、丝裂霉素C、奥沙利铂、紫杉醇、培美曲塞、替莫唑胺和长春新碱。
在一些实施方案中,所述其它治疗剂选自血管发生-靶向的治疗,包括,例如,阿柏西普和贝伐单抗。
在一些实施方案中,所述其它治疗剂选自免疫靶向的试剂,包括,例如,阿地白介素、易普利单抗、拉姆单抗(Iambrolizumab)、纳武单抗和西普鲁塞-T。
在一些实施方案中,其它治疗剂选自有效抵抗下游Trk路径的药剂,包括,例如靶向NGF的生物药物,例如NGF抗体及panTrk抑制剂。
在一些实施方案中,其它治疗剂或疗法为放射疗法,包括(例如)放射性碘化物疗法、外照射辐射及镭223疗法。
在一些实施方案中,其它治疗剂包括上文所列示疗法或治疗剂中的任一者,这些疗法或治疗剂为其中癌症具有NTRK基因、Trk蛋白或它们的表达或活性或水平的失调的癌症的护理标准。
在一些实施方案中,本文提供了治疗患者癌症(例如,Trk-相关的癌症)的方法,包括向所述患者给药本发明化合物。在一些实施方案中,该至少一种其它治疗或治疗剂选自放射治疗(例如,放射性碘化物治疗、外照射辐射、或镭223治疗)、细胞毒素化疗剂(例如,三氧化二砷、博来霉素、卡巴他赛、卡培他滨、卡铂、顺铂、环磷酰胺、阿糖胞苷、达卡巴嗪、柔红霉素、多西紫杉醇、多 柔比星、依托泊苷、氟尿嘧啶、吉西他滨、伊立替康、洛莫司汀、甲氨碟呤、丝裂霉素C、奥沙利铂、紫杉醇、培美曲赛、替莫唑胺、或长春新碱)、酪氨酸激酶靶向的治疗(例如,阿法替尼、卡博替尼、西妥昔单抗、克唑替尼、达拉菲尼、埃罗替尼、吉非替尼、伊马替尼、拉帕替尼、尼洛替尼、帕唑帕尼、帕唑帕尼、帕尼单抗、帕妥珠单抗、瑞戈非尼、舒尼替尼、或曲妥单抗)、凋亡调节剂和信号转导抑制剂(例如依维莫司、哌立福辛、雷帕霉素、索拉菲尼、西罗莫司、曲美替尼、或威罗菲尼)、免疫-靶向的治疗(例如,阿地白介素、干扰素a-2b、易普利单抗、拉姆单抗、纳武单抗、***、或西普鲁赛-T)和血管发生-靶向的治疗(例如,阿柏西普或贝伐单抗),其中当与其它治疗或治疗剂组合时,本发明化合物可有效治疗所述癌症。
在一些实施方案中,所述其它治疗剂为不同的Trk抑制剂。其它Trk抑制剂的非限制性实例包括(R)-2-苯基吡咯烷取代的咪唑并哒嗪、AZD6918、GNF-4256、GTX-186、GNF-5837、AZ623、AG-879、altiratinib、CT327、AR-772、AR-523、AR-786、AR-256、AR-618、AZ-23、AZD7451、卡博替尼、CEP-701、CEP-751、PHA-739358、多韦替尼、entrectinib、PLX7486、GW441756、MGCD516、ONO-5390556、PHA-848125AC、瑞戈非尼、索拉非尼、舒尼替尼、TSR-011、VM-902A、K252a、4-氨基吡唑基嘧啶和取代的吡唑并[1,5-a]嘧啶化合物。
这些其它治疗剂可与本文所提供的一或多种化合物一起作为相同或单独剂型的一部分经由相同或不同的给药途径且基于相同或不同的给药时间表根据本领域技术人员已知的标准药物实践来给药。
本发明的化合物与现有技术中已知的非氘代化合物相比,具有一系列优点。本发明的优点包括:第一,采用本发明技术方案的化合物和组合物为疼痛、癌症、炎症、神经退行性疾病或某些传染病,特别是Trk-相关的疾病的治疗提供了更有利的治疗工具。第二,改进了化合物在生物体中的代谢,使化合物具有更好的药代动力学参数特性。在这种情况下,可以改变剂量并形成长效制剂,改善适用性。第三,提高了化合物在动物体内的药物浓度,提高了药物疗效。第四,抑制了某些代谢产物,提高化合物的安全性。
实施例
下面结合具体实施例,作进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~ 80℃)下进行。反应时间通常为0.1-60小时,优选地为0.5-24小时。
实施例1:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-13,13,14,14-d 4(化合物13);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-13,13,14,14-d 4(化合物L-1-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-13,13,14,14-d 4(化合物L-1-b)的制备。
Figure PCTCN2019072833-appb-000017
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000018
Figure PCTCN2019072833-appb-000019
步骤1:化合物1的合成
取3-溴-5-氟-2-甲氧基吡啶(3.09g,15mmol)溶于无水THF(50mL)中,0℃下缓慢滴加异丙基氯化镁溶液(7.0mL,14.0mmol),滴毕自然升温至0℃搅拌1h,再于-15℃缓慢滴加N-叔丁氧羰基-2-吡咯烷酮(1.85g,10.0mmol)的无水四氢呋喃(10mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用30mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得2.86g浅黄色液体即为化合物1,收率:61.1%。LC-MS(APCI):m/z=313.2(M+1) +
步骤2化合物2的合成
取化合物1(1.87g,5.97mmol)溶于甲苯(20mL)中,加入1.1mL浓盐酸,升温至65℃搅拌反应过夜,降至室温,用2M氢氧化钠调PH至14,继续搅拌1h,TLC检测反应完毕。分出有机相,水相用乙酸乙酯萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,柱层析得到624mg黄色液体即为化合物2,收率:53.9%。LC-MS(APCI):m/z=195.1(M+1) +
步骤3化合物3的合成
取化合物2(624mg,3.21mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得620mg无色油状液体即为化合物3,收率:98.5%。LC-MS(APCI):m/z=197.3(M+1) +
步骤4化合物4的合成
取化合物3(162mg,0.82mmol)和5-氯吡唑并[1,5-a]嘧啶-3-羧酸乙酯(185.4mg,0.82mmol)溶于无水乙醇(6mL)中,室温下加入DIPEA(N,N-二异丙基乙胺,423.9mg,3.28mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得204mg淡黄色固体粉末即为化合物4,收率:64.7%。LC-MS(APCI):m/z=386.5(M+1) +
步骤5化合物5的合成
取化合物4(200mg,0.52mmol)溶于4M的氯化氢的二氧六环(5ml,20mmol)溶液中,密封加热至110℃搅拌反应24h。TLC检测反应完毕后浓缩除去溶剂,直接用于下一步。
步骤6化合物6的合成
取化合物5(1.78g,4.79mmol)和N-苯基双(三氟甲烷磺酰)亚胺(1.88g,5.27mmol)分散于25mL无水DMF中,加入三乙胺(581.6mg,5.75mmol),氮气保护下室温搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取三遍,合并有机相,用饱和食盐水洗涤,浓缩后柱层析(PE/EA:50%~66%)得2.05g黄色固体粉末即为化合物6,收率:85%。LC-MS(APCI):m/z=504.3(M+1) +
步骤7化合物7的合成
取(S)-3-丁炔-2-醇(280mg,4.0mmol)溶于10mL无水二氯甲烷中,加入三乙胺(445.2mg,4.4mmol),氮气保护下0℃下缓慢滴加对甲苯磺酰氯(762.6mg,4.0mmol)的二氯甲烷(5mL)溶液,滴毕室温搅拌1h。TLC检测反应完毕后,加入二氯甲烷稀释,依次用水和饱和食盐水洗涤,浓缩后柱层析得到806mg类白色固体即为化合物7,收率:90%。
步骤8化合物8的合成
取化合物7(448mg,2.0mmol)溶于10mL无水DMF中,加入酰酞亚胺钾(370.4mg,2.0mmol),室温下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,水相用乙酸乙酯萃取3次,合并有机相,饱和食盐水洗涤,浓缩柱层析得到243mg白色固体即为化合物8,收率:61%。LC-MS(APCI):m/z=200.2(M+1) +
步骤9化合物9的合成
取化合物6(503.1mg,1.0mmol)和化合物8(199mg,1.0mmol)溶于25mL无水四氢呋喃中,氮气保护下加入Pd(PPh 3) 2Cl 2(35mg,0.05mmol)和CuI(19mg,0.1mmol),室温下一次性加入三乙胺(202.4mg,2.0mmol),室温搅拌过夜。TLC检测反应完毕后,浓缩除去溶剂,柱层析得287mg淡黄色固体粉末即为化合物9,收率:52%。LC-MS(APCI):m/z=553.3(M+1) +
步骤10化合物10的合成
取化合物9(287mg,0.52mmol)溶于10ml氘代甲醇中,加入催化量的Pd/C,充氘气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤11化合物11的合成
取化合物10(287mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到181.2mg浅黄色固体,收率:81%。LC-MS(APCI):m/z=431.2(M+1) +
步骤12化合物12的合成
取化合物11(224mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤13化合物13的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(五氟苯基二苯基磷酸酯,240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到73.9mg类白色固体即为化合物13,收率:37%。LC-MS(APCI):m/z=385.3(M+1) +1H NMR(400MHz,DMSO)δ8.87(s,1H),8.42(s,1H),8.33(s,1H),8.04(d,J=2.3Hz,1H),7.52(s,1H),6.74(d,J=2.3Hz,1H),4.38(t,1H),3.61(dd,J=17.0,9.3Hz,2H),3.15(m,2H),2.06(m,2H),2.01–1.65(m,2H),1.22(d,J=4.5Hz,2H).
步骤14化合物L-1-a和L-1-b的制备
采用手性制备色谱柱将消旋体化合物13进行分离得到化合物L-1-a和L-1-b。
实施例2:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6-d 3(化合物22);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6-d 3(化合物L-2-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6-d 3(化合物L-2-b)的制备。
Figure PCTCN2019072833-appb-000020
采用以下路线进行合成:
步骤1化合物14的合成
取化合物2(1.25g,6.42mmol)溶于氘代甲醇(20mL)中,加入Pd/C(100mg),充氘气室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得1.21g无色油状液体即为化合物14,收率:95%。LC-MS(APCI):m/z=200.1(M+1) +
步骤2化合物15的合成
取化合物14(326mg,1.64mmol)和5-氯吡唑并[1,5-a]嘧啶-3-羧酸乙酯(370.8mg,1.64mmol)溶于无水乙醇(15mL)中,室温下加入DIPEA(847.8mg,6.56mmol),加热回流30min。浓缩反 应液,柱层析(PE/EA,30%~50%)得445mg淡黄色固体粉末即为化合物15,收率:70%。LC-MS(APCI):m/z=389.5(M+1) +
步骤3化合物16的合成
取化合物15(400mg,1.03mmol)溶于4M的氯化氢二氧六环(8ml,32mmol)溶液中,密封加热至110℃搅拌反应24h。TLC检测反应完毕后浓缩除去溶剂,直接用于下一步。
步骤4化合物17的合成
取化合物16(1.79g,4.79mmol)和N-苯基双(三氟甲烷磺酰)亚胺(1.88g,5.27mmol)分散于25mL无水DMF中,加入三乙胺(581.6mg,5.75mmol),氮气保护下室温搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取三遍,合并有机相,用饱和食盐水洗涤,浓缩后柱层析(PE/EA:50%~66%)得2.05g黄色固体粉末即为化合物17,收率:85%。LC-MS(APCI):m/z=507.8(M+1) +
步骤5化合物18的合成
取化合物17(506.1mg,1.0mmol)和化合物8(199mg,1.0mmol)溶于25mL无水四氢呋喃中,氮气保护下加入Pd(PPh 3) 2Cl 2(35mg,0.05mmol)和CuI(19mg,0.1mmol),室温下一次性加入三乙胺(202.4mg,2.0mmol),室温搅拌过夜。TLC检测反应完毕后,浓缩除去溶剂,柱层析得289mg淡黄色固体粉末即为化合物18,收率:52%。LC-MS(APCI):m/z=556.3(M+1) +
步骤6化合物19的合成
取化合物18(289mg,0.52mmol)溶于5ml甲醇和5ml四氢呋喃的混合溶液中,加入催化量的Pd/C,充氢气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤7化合物20的合成
取化合物20(289mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到181.2mg浅黄色固体,收率:81%。LC-MS(APCI):m/z=430.2(M+1) +
步骤8化合物21的合成
取化合物20(223mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤9化合物22的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到65mg类白色固体即为化合物22,收率:32.5%。
步骤10化合物L-2-a和L-2-b的制备
采用手性制备色谱柱将消旋体化合物22进行分离得到化合物L-2-a和L-2-b。
实施例3:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6,13,13,14,14-d 7(化合物26);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6,13,13,14,14-d 7(化合物L-3-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-5,5,6,13,13,14,14-d 7(化合物L-3-b)的制备。
Figure PCTCN2019072833-appb-000022
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000023
步骤1化合物23的合成
取化合物18(289mg,0.52mmol)溶于10ml氘代甲醇中,加入催化量的Pd/C,充氘气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤2化合物24的合成
取化合物23(290mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到153.2mg浅黄色固体,收率:68%。LC-MS(APCI):m/z=434.2(M+1) +
步骤3化合物25的合成
取化合物24(225.1mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤4化合物26的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到90.55mg类白色固体即为化合物26,收率:45%。LC-MS(APCI):m/z=388.3(M+1) +
步骤5化合物L-3-a和L-3-b的制备
采用手性制备色谱柱将消旋体化合物26进行分离得到化合物L-3-a和L-3-b。
实施例4:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4-d 2(化合物40);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4-d 2(化合物L-4-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4-d 2(化合物L-4-b)的制备。
Figure PCTCN2019072833-appb-000024
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000025
步骤1化合物27的合成
取琥珀酰亚胺(9.0g,90.9mmol)溶于60mL氘代甲醇中,加入碳酸钾(1.44g,10.4mmol),微波加热120℃搅拌30min。浓缩反应液,所得固体即为化合物27,直接用于下一步。
步骤2化合物28的合成
上步所得固体分散于400mL无水四氢呋喃,冰浴下分批加入氢化锂铝(3.05g,80.3mmol),加完后于冰浴下搅拌15min。反应液用十水硫酸钠淬灭,过滤,滤渣用200mL乙酸乙酯洗涤,合并滤液并浓缩,柱层析(PE/EA,25%~50%)得1.44g无色油状液体即为化合物28,直接投入下一步反应。
步骤3化合物29的合成
取化合物28(1.44g,16.2mmol)溶于20mL二氯甲烷,室温下加入DIPEA和DMAP(4-二甲 氨基吡啶),冰水浴下缓慢滴加Boc 2O(二碳酸二叔丁酯),加完后室温搅拌过夜。浓缩反应液,柱层析得586mg棕色油状液体即为化合物29,直接投入下一步。
步骤4化合物30的合成
取3-溴-5-氟-2-甲氧基吡啶(3.09g,15mmol)溶于无水THF(50mL)中,0℃下缓慢滴加异丙基氯化镁溶液(7.0mL,14.0mmol),滴毕自然升温至0℃搅拌1h,再于-15℃缓慢滴加化合物29(1.89g,10.0mmol)的无水四氢呋喃(10mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用30mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得浅黄色液体即为化合物30。
步骤5化合物31的合成
取化合物30(1.88g,5.97mmol)溶于甲苯(20mL)中,加入1.1mL浓盐酸,升温至65℃搅拌反应过夜,降至室温,用2M氢氧化钠调PH至14,继续搅拌1h,TLC检测反应完毕。分出有机相,水相用乙酸乙酯萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,柱层析得到650mg黄色液体即为化合物31.
步骤6化合物32的合成
取化合物31(650mg,3.28mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得650mg无色油状液体即为化合物32。
步骤7化合物33的合成
取化合物32(162mg,0.82mmol)和5-氯吡唑并[1,5-a]嘧啶-3-羧酸乙酯(185.4mg,0.82mmol)溶于无水乙醇(6mL)中,室温下加入DIPEA(423.9mg,3.28mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得淡黄色固体粉末即为化合物33。
步骤8化合物34的合成
取化合物33(201mg,0.52mmol)溶于4M的氯化氢二氧六环(5ml,20mmol),密封加热至110℃搅拌反应24h。TLC检测反应完毕后浓缩除去溶剂,直接用于下一步。
步骤9化合物35的合成
取化合物34(1.79g,4.79mmol)和N-苯基双(三氟甲烷磺酰)亚胺(1.88g,5.27mmol)分散于25mL无水DMF中,加入三乙胺(581.6mg,5.75mmol),氮气保护下室温搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取三遍,合并有机相,用饱和食盐水洗涤,浓缩后柱层析(PE/EA:50%~66%)得黄色固体粉末即为化合物35。
步骤10化合物36的合成
取化合物35(503.1mg,1.0mmol)和化合物8(199mg,1.0mmol)溶于25mL无水四氢呋喃中,氮气保护下加入Pd(PPh 3) 2Cl 2(35mg,0.05mmol)和CuI(19mg,0.1mmol),室温下一次性加入三乙胺(202.4mg,2.0mmol),室温搅拌过夜。TLC检测反应完毕后,浓缩除去溶剂,柱层析得淡黄色固体粉末即为化合物36。
步骤11化合物37的合成
取化合物36(288mg,0.52mmol)溶于5ml甲醇和5ml四氢呋喃的混合溶液中,加入催化量的Pd/C,充氢气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤12化合物38的合成
取化合物37(289mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到浅黄色固体即为化合物38。
步骤13化合物39的合成
取化合物38(224mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤14化合物40的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到类白色固体即为化合物40。
步骤15化合物L-4-a和L-4-b的制备
采用手性制备色谱柱将消旋体化合物40进行分离得到化合物L-4-a和L-4-b。
实施例5:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3-d 2(化合物53);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3-d 2(化合物L-5-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3-d 2(化合物L-5-b)的制备。
Figure PCTCN2019072833-appb-000026
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000027
步骤1化合物41的合成
取琥珀酰亚胺(3.0g,30.3mmol)分散于130mL无水四氢呋喃,冰浴下分批加入LiAlD 4(1.02g,24.3mmol),加完后于冰浴下搅拌15min。反应液用十水硫酸钠淬灭,过滤,滤渣用70mL乙酸 乙酯洗涤,合并滤液并浓缩,柱层析(PE/EA,25%~50%)得1.44g无色油状液体即为化合物41,直接投入下一步。
步骤2化合物42的合成
取化合物41(1.44g,16.5mmol)溶于20mL二氯甲烷,室温下加入DIPEA(8.55g,66.1mmol)和DMAP(404mg,3.3mmol),冰水浴下缓慢滴加Boc 2O(7.22g,33.1mmol),加完后室温搅拌过夜。浓缩反应液,柱层析得586mg棕色油状液体即为化合物42,直接投入下一步。
步骤3化合物43的合成
取3-溴-5-氟-2-甲氧基吡啶(3.09g,15mmol)溶于无水THF(50mL)中,0℃下缓慢滴加异丙基氯化镁溶液(7.0mL,14.0mmol),滴毕自然升温至0℃搅拌1h,再于-15℃缓慢滴加化合物42(1.85g,10.0mmol)的无水四氢呋喃(10mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用30mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得浅黄色液体即为化合物43。
步骤4化合物44的合成
取化合物43(1.87g,5.97mmol)溶于甲苯(20mL)中,加入1.1mL浓盐酸,升温至65℃搅拌反应过夜,降至室温,用2M氢氧化钠调PH至14,继续搅拌1h,TLC检测反应完毕。分出有机相,水相用乙酸乙酯萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,柱层析得到624mg黄色液体即为化合物44。
步骤5化合物45的合成
取化合物44(624mg,3.21mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得620mg无色油状液体即为化合物45。
步骤6化合物46的合成
取化合物45(162mg,0.82mmol)和5-氯吡唑并[1,5-a]嘧啶-3-羧酸乙酯(185.4mg,0.82mmol)溶于无水乙醇(6mL)中,室温下加入DIPEA(423.9mg,3.28mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得204mg淡黄色固体粉末即为化合物46。
步骤7化合物47的合成
取化合物46(200mg,0.52mmol)溶于4M的氯化氢二氧六环(5ml,20mmol),密封加热至110℃搅拌反应24h。TLC检测反应完毕后浓缩除去溶剂,直接用于下一步。
步骤8化合物48的合成
取化合物47(1.78g,4.79mmol)和N-苯基双(三氟甲烷磺酰)亚胺(1.88g,5.27mmol)分散 于25mL无水DMF中,加入三乙胺(581.6mg,5.75mmol),氮气保护下室温搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取三遍,合并有机相,用饱和食盐水洗涤,浓缩后柱层析(PE/EA:50%~66%)得黄色固体粉末即为化合物48。
步骤9化合物49的合成
取化合物48(503.1mg,1.0mmol)和化合物8(199mg,1.0mmol)溶于25mL无水四氢呋喃中,氮气保护下加入Pd(PPh 3) 2Cl 2(35mg,0.05mmol)和CuI(19mg,0.1mmol),室温下一次性加入三乙胺(202.4mg,2.0mmol),室温搅拌过夜。TLC检测反应完毕后,浓缩除去溶剂,柱层析得淡黄色固体粉末即为化合物49。
步骤10化合物50的合成
取化合物49(287mg,0.52mmol)溶于5ml甲醇和5ml四氢呋喃的混合溶液中,加入催化量的Pd/C,充氢气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤11化合物51的合成
取化合物50(287mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到浅黄色固体即为化合物51。
步骤12化合物52的合成
取化合物51(224mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤13化合物53的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到类白色固体即为化合物53。
步骤14化合物L-5-a和L-5-b的制备
采用手性制备色谱柱将消旋体化合物53进行分离得到化合物L-5-a和L-5-b。
实施例6:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4,13,13,14,14-d 6(化合物57);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4,13,13,14,14-d 6(化合物L-6-a);
(6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-4,4,13,13,14,14-d 6(化合物L-6-b)的制备。
Figure PCTCN2019072833-appb-000028
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000029
步骤1化合物54的合成
取化合物36(287mg,0.52mmol)溶于10ml氘代甲醇中,加入催化量的Pd/C,充氘气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤2化合物55的合成
取化合物54(289mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到浅黄色固体即为化合物55。
步骤3化合物56的合成
取化合物55(226mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤4化合物57的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到类白色固体即为化合物57。
步骤5化合物L-6-a和L-6-b的制备
采用手性制备色谱柱将消旋体化合物57进行分离得到化合物L-6-a和L-6-b。
实施例7:(15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3,13,13,14,14-d 6(化合物61);
(6R,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3,13,13,14,14-d 6(化合物L-7-a);
( 6S,15R)-9-氟-15-甲基-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-3,3,13,13,14,14-d 6(化合物L-7-b)的制备。
Figure PCTCN2019072833-appb-000030
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000031
步骤1化合物58的合成
取化合物49(287mg,0.52mmol)溶于10ml氘代甲醇中,加入催化量的Pd/C,充氘气室温下搅拌反应2-4h,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,直接投入下一步反应。
步骤2化合物59的合成
取化合物58(289mg,0.52mmol)溶于10ml甲醇中,加入水合肼(130mg,2.6mmol),加热回流反应1-2h,TLC检测反应完毕后,浓缩除去溶剂,柱层析纯化得到浅黄色固体即为化合物59。
步骤3化合物60的合成
取化合物59(226mg,0.52mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,浓缩除去溶剂,直接投入到下一步反应。
步骤4化合物61的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到类白色固体即为化合物61。
步骤5化合物L-7-a和L-7-b的制备
采用手性制备色谱柱将消旋体化合物61进行分离得到化合物L-7-a和L-7-b。
实施例8:9-氟-15-(甲基-d 3)-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-15-d(化合物70);
(6R,15R)-9-氟-15-(甲基-d 3)-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-15-d(化合物L-8-a);
(6S,15R)-9-氟-15-(甲基-d 3)-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-15-d(化合物L-8-b);
(6R,15S)-9-氟-15-(甲基-d 3)-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-15-d(化合物L-8-c);
(6S,15S)-9-氟-15-(甲基-d 3)-2,11,16,20,21,24-六氮杂戊环[16.5.2.0 2,6,0 7,12,0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-17-酮-15-d(化合物L-8-d)的制备。
Figure PCTCN2019072833-appb-000032
Figure PCTCN2019072833-appb-000033
采用以下路线进行合成:
Figure PCTCN2019072833-appb-000034
步骤1化合物62的合成
取丙氨酸-d 4(1.0g,10.74mmol)溶于20mL无水甲醇中,0℃下缓慢滴加氯化亚砜6.4g,53.7mmol),加毕,回流反应2-4小时,TLC检测反应完毕后,浓缩除去溶剂,加入甲苯带2-3次,加入二氯甲烷(20ml)溶解,加入三乙胺(2.17g,21.48mmol),氮气保护下0℃下缓慢滴加(Boc) 2O(2.81g,12.89mmol),滴毕室温搅拌5h。TLC检测反应完毕后,加入二氯甲烷稀释,依次用水和饱和食盐水洗涤,浓缩后柱层析得到1.82g无色油状液体,收率:82%。
步骤2化合物63的合成
取化合物62(1.82g,8.8mmol)溶于20mL无水二氯甲烷中,氮气保护下,降温至-78℃,缓慢滴加DIBAL-H(二异丁基氢化铝,8.8ml,8.8mmol),低温下搅拌反应过夜,TLC检测反应完毕 后,加入水稀释,水相用乙酸乙酯萃取3次,合并有机相,饱和食盐水洗涤,浓缩柱层析得到1.01g无色液体,收率:65%。LC-MS(APCI):m/z=178.2(M+1) +
步骤3化合物65的合成
取化合物63(1.01g,5.7mmol)和化合物64(1.1g,5.7mmol)溶于25mL无水甲醇中,加入碳酸钾(2.36g,17.1mol),室温搅拌过夜。TLC检测反应完毕后,浓缩除去溶剂,柱层析得759mg淡黄色固体粉末,收率:77%。LC-MS(APCI):m/z=174.3(M+1) +
步骤4化合物66的合成
取化合物6(287mg,0.52mmol)和化合物65(135mg,0.78mmol)溶于10ml无水四氢呋喃中,氮气保护下加入二三苯基膦二氯化钯(36.5mg,0.052mmol)和碘化亚铜(19mg,0.1mmol),再加入碳酸钾(107.8mg,0.78mmol),加热至100℃搅拌反应过夜,TLC检测反应完毕后,过滤除去催化剂,滤液浓缩至干,柱层析得到194mg淡黄色粉末,收率:71%。LC-MS(APCI):m/z=527.3(M+1) +
步骤5化合物67的合成
取化合物66(194mg,0.37mmol)溶于10ml甲醇中,加入催化量Pd/C,充氢气室温下反应2-4小时,TLC检测反应完毕后,浓缩除去催化剂,滤液浓缩至干,直接投入到下一步反应。
步骤6化合物68的合成
取化合物67(196mg,0.37mmol)溶于3ml甲醇和2ml水中,加入氢氧化锂(109.2mg,2.6mmol),升温至50℃搅拌反应4-6h,TLC检测反应完毕后,降至室温,用稀盐酸调PH至酸性,乙酸乙酯萃取3-4次,合并有机相,浓缩后柱层析纯化得到175mg淡黄色固体,收率:94%。LC-MS(APCI):m/z=503.1(M+1) +
步骤7化合物69的合成
取化合物68(175mg,0.35mmol),加入4M氯化氢二氧六环溶液(5ml,20mmol),室温下搅拌反应1-2小时,TLC检测反应完毕后,浓缩除去溶剂,直接投入到下一步反应。
步骤7化合物70的合成
将上步所得产物溶于10ml无水DMF中,室温下加入FDPP(240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到58.3mg类白色固体,收率:43.3%。LC-MS(APCI):m/z=385.3(M+1) +1H NMR(400MHz,DMSO)δ8.85(s,1H),8.42(s,1H),8.31(s,1H),8.06(d,J=2.3Hz,1H),7.51(s,1H),6.74(d,J=2.3Hz,1H),4.38(t,1H),3.61(dd,J=17.0,9.3Hz,2H), 3.15(m,2H),2.06(m,2H),2.01–1.65(m,2H),1.22(d,J=4.5Hz,2H).
步骤7化合物L-8-a、L-8-b、L-8-c和L-8-d的制备
采用手性制备色谱柱将消旋体化合物70进行分离得到化合物化合物L-8-a、L-8-b、L-8-c和L-8-d。
生物活性测试。
(1)激酶抑制作用
化合物配制:受试化合物溶于DMSO配成20mM母液。使用前将化合物在DMSO中稀释成0.1mM(100倍终浓度的稀释液),并做3倍梯度稀释,11个浓度。加药时用缓冲液稀释成4倍终浓度的稀释液。
激酶检测:配制缓冲液后,将酶与预先稀释配制的不同浓度化合物混合,室温放置30分钟,每个浓度双复孔。加入对应底物及ATP,室温反应60分钟(其中设置阴阳性对照)。反应完毕加入抗体检测,室温孵育60分钟后Evnvision检测,采集数据。通过Evnvision酶标仪检测,测定在各浓度的本发明化合物存在下的酶活力,并计算不同浓度的化合物对酶活力的抑制活性,之后根据四参数方程,根据Graphpad 5.0软件对不同浓度化合物下酶活力的抑制活性进行拟合,计算出IC 50值。
按照上述方法,测试本发明化合物对TRK A、TRK B、TRK C激酶的抑制活性。代表性的实施例化合物的激酶抑制作用结果如表1所示。表1:
实施例化合物 TRK A IC 50(nM) TRK B IC 50(nM) TRK C IC 50(nM)
化合物Φ 8.27 4.19 3.10
化合物Φ-a 0.29 0.08 0.11
化合物L-2-a 0.17 0.07 0.07
如表1所示,本发明化合物具有显著的蛋白激酶抑制活性,一般具有低于1nM的IC 50值。特别是,同没有氘代的化合物Φ和化合物Φ-a比较,本发明化合物对TRKA/B/C表现出强的抑制活性。
(2)细胞毒性实验
检测实施例化合物对KM12(TPM3-TRKA)细胞活性的抑制效应。
耗材及试剂:RPMI-1640培养基(GIBCO,目录号A10491-01)、胎牛血清(GIBCO,目录号10099141)、抗生素(Penicillin-Streptomycin),IL-3(PeproTech),嘌呤霉素;活细胞检测试剂盒CellTiter-Glo4(Promega,目录号G7572),96孔黑壁透明平底细胞培养板(Corning,目录号3340)。
实验方法:1.制备细胞板将KM12细胞分别种于96孔板中,并在KM12细胞中加入8ng/ml IL-3, 细胞板置于二氧化碳培养箱中过夜培养。2.用DMSO溶解被测化合物并进行3.16倍梯度稀释,9个化合物浓度,设置三复孔实验。3.化合物处理细胞将化合物转移到细胞板中,化合物起始浓度为10μM。细胞板置于二氧化碳培养箱中培养3天。4.检测向细胞板中加入CellTiter-Glo试剂,室温孵育30分钟使发光信号稳定。采用PerkinElmer Envision多标记分析仪读数。代表性的实施例化合物的抑制细胞增殖作用结果如表2所示。
表2
实施例化合物 KM12IC 50(nM)
化合物Φ-a 2.56
化合物L-2-a 2.51
如表2所示,本发明化合物都表现出更好的抑制KM12细胞增殖的性质。
(3)代谢稳定性评价
微粒体实验:人肝微粒体:0.5mg/mL,Xenotech;大鼠肝微粒体:0.5mg/mL,Xenotech;辅酶(NADPH/NADH):1mM,Sigma Life Science;氯化镁:5mM,100mM磷酸盐缓冲剂(pH为7.4)。
储备液的配制:精密称取一定量的实施例化合物的粉末,并用DMSO分别溶解至5mM。
磷酸盐缓冲液(100mM,pH7.4)的配制:取预先配好的150mL的0.5M磷酸二氢钾和700mL的0.5M磷酸氢二钾溶液混合,再用0.5M磷酸氢二钾溶液调节混合液pH值至7.4,使用前用超纯水稀释5倍,加入氯化镁,得到磷酸盐缓冲液(100mM),其中含100mM磷酸钾,3.3mM氯化镁,pH为7.4。
配制NADPH再生***溶液(含有6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D,3.3mM氯化镁),使用前置于湿冰上。
配制终止液:含有50ng/mL盐酸***和200ng/mL甲苯磺丁脲(内标)的乙腈溶液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL人肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL SD大鼠肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。
样品的孵育:用含70%乙腈的水溶液将相应化合物的储备液分别稀释至0.25mM,作为工作液,备用。分别取398μL的人肝微粒体或者大鼠肝微粒体稀释液加入96孔孵育板中(N=2),分别加入2μL0.25mM的的工作液中,混匀。
代谢稳定性的测定:在96孔深孔板的每孔中加入300μL预冷的终止液,并置于冰上,作为终 止板。将96孔孵育板和NADPH再生***置于37℃水浴箱中,100转/分钟震荡,预孵5min。从孵育板每孔取出80μL孵育液加入终止板,混匀,补充20μL NADPH再生***溶液,作为0min样品。再向孵育板每孔加入80μL的NADPH再生***溶液,启动反应,开始计时。相应化合物的反应浓度为1μM,蛋白浓度为0.5mg/mL。分别于反应10、30、90min时,各取100μL反应液,加入终止板中,涡旋3min终止反应。将终止板于5000×g,4℃条件下离心10min。取100μL上清液至预先加入100μL蒸馏水的96孔板中,混匀,采用LC-MS/MS进行样品分析。
数据分析:通过LC-MS/MS***检测相应化合物及内标的峰面积,计算化合物与内标峰面积比值。通过化合物剩余量的百分率的自然对数与时间作图测得斜率,并根据以下公式计算t 1/2和CL int,其中V/M即等于1/蛋白浓度。
Figure PCTCN2019072833-appb-000035
t 1/2(min);CL int(μL/min/mg)。
对本发明化合物及其没有氘代的化合物同时测验比较,评价其在人和大鼠肝微粒体的代谢稳定性。采用未经氘代的化合物LOXO-195作为对照品。在人和大鼠肝微粒体实验中,通过与未经氘代的化合物LOXO-195对照,本发明化合物可以明显改善代谢稳定性。代表性实施例化合物的人和大鼠的肝微粒体实验结果如下表3所示:
表3:
Figure PCTCN2019072833-appb-000036
(4)大鼠药代动力学实验
6只雄性Sprague-Dawley大鼠,7-8周龄,体重约210g,分成2组,每组3只,经静脉或口服单个剂量的化合物(口服10mg/kg),比较其药代动力学差异。
大鼠采用标准饲料饲养,给予水。试验前16小时开始禁食。药物用PEG400和二甲亚砜溶解。眼眶采血,采血的时间点为给药后0.083小时,0.25小时、0.5小时、1小时、2小时、4小时、6小时、8小时、12小时和24小时。
大鼠吸入***后短暂麻醉,眼眶采集300μL血样于试管。试管内有30μL 1%肝素盐溶液。使用前,试管于60℃烘干过夜。在最后一个时间点血样采集完成之后,大鼠***麻醉后处死。
血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血样在4℃5000rpm离心5分钟,将血浆与红细胞分离。用移液器吸出100μL血浆到干净的塑料离心管中,标明化合物的名称和时间点。血浆在进行分析前保存在-80℃。用LC-MS/MS测定血浆中本发明化合物的浓度。药代动力学参数基于每只动物在不同时间点的血药浓度进计算。
实验表明,本发明化合物在动物体内具有更好的药代动力学性质,因此具有更好的药效学和治辽效果。
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (14)

  1. 一种式(Aa)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体:
    Figure PCTCN2019072833-appb-100001
    其中,
    R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12各自独立地选自氢或氘;
    X选自CH 3、CD 3、CHD 2或CH 2D;
    条件是如果X是CH 3,那么R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11和R 12中至少一个是氘。
  2. 根据权利要求1所述的化合物,其为式(Aa-1)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体:
    Figure PCTCN2019072833-appb-100002
    其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12和X如权利要求1所定义。
  3. 根据权利要求2所述的化合物,其为式(Ia)或式(IIa)化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体:
    Figure PCTCN2019072833-appb-100003
    其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12和X如权利要求2所定义。
  4. 根据权利要求1-3中任一项所述的化合物,其中,R 11和R 12各自独立地选自氢。
  5. 根据权利要求1-4中任一项所述的化合物,其中,R 9和R 10各自独立地选自氢。
  6. 根据权利要求1-5中任一项所述的化合物,其中,R 1选自氢。
  7. 根据权利要求1-6中任一项所述的化合物,其中,X选自CH 3
  8. 根据权利要求1-7中任一项所述的化合物,其中,R 2、R 3、R 4和R 5各自独立地选自氢。
  9. 根据权利要求1-8中任一项所述的化合物,其中,R 6、R 7和R 8各自独立地选自氘。
  10. 根据权利要求1所述的化合物,其中所述化合物选自下述化合物,或其药学上可接受的盐、前药、水合物或溶剂化合物、多晶型、立体异构体或同位素变体:
    Figure PCTCN2019072833-appb-100004
    Figure PCTCN2019072833-appb-100005
    Figure PCTCN2019072833-appb-100006
  11. 一种药物组合物,其含有药学上可接受的赋形剂和权利要求1-10中任一项所述的化合物或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体。
  12. 权利要求1-10中任一项所述的化合物或其药学上可接受的盐、前药、水合物或溶剂化合物、晶型、立体异构体或同位素变体,或权利要求11的药物组合物在制备治疗由野生型和突变型Trk激酶介导的疾病的药物中用途。
  13. 根据权利要求12所述的用途,其中所述疾病通过TrkA、TrkB或rkA和TrkB介导。
  14. 根据权利要求12或13所述的用途,其中所述疾病选自疼痛、癌症、炎症、神经退行性疾病或锥虫感染。
PCT/CN2019/072833 2018-01-23 2019-01-23 取代的吡唑并[1,5-a]嘧啶类的大环化合物 WO2019144885A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP19743399.8A EP3744723B1 (en) 2018-01-23 2019-01-23 Substituted pyrazolo[1,5-a]pyrimidine macrocyclic compound
JP2020540580A JP7162915B2 (ja) 2018-01-23 2019-01-23 置換ピラゾロ[1,5-a]ピリミジン系大環状化合物
US16/964,069 US11345703B2 (en) 2018-01-23 2019-01-23 Substituted pyrazolo[1,5-a]pyrimidine macrocyclic compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810063754 2018-01-23
CN201810063754.X 2018-01-23

Publications (1)

Publication Number Publication Date
WO2019144885A1 true WO2019144885A1 (zh) 2019-08-01

Family

ID=65917013

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/072833 WO2019144885A1 (zh) 2018-01-23 2019-01-23 取代的吡唑并[1,5-a]嘧啶类的大环化合物

Country Status (5)

Country Link
US (1) US11345703B2 (zh)
EP (1) EP3744723B1 (zh)
JP (1) JP7162915B2 (zh)
CN (2) CN111848622A (zh)
WO (1) WO2019144885A1 (zh)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019144885A1 (zh) * 2018-01-23 2019-08-01 深圳市塔吉瑞生物医药有限公司 取代的吡唑并[1,5-a]嘧啶类的大环化合物
CA3117200A1 (en) 2018-10-22 2020-04-30 Esker Therapeutics, Inc. Tyk2 inhibitors and uses thereof
WO2021027503A1 (zh) * 2019-08-12 2021-02-18 罗欣药业(上海)有限公司 三环类化合物、其制备方法、中间体及应用
CN113004305B (zh) * 2019-12-19 2024-04-09 赛诺哈勃药业(成都)有限公司 大环化合物及其制备方法和用途
CN111777549A (zh) * 2020-07-07 2020-10-16 中瀚(齐河县)生物医药科技有限公司 一种2-甲氧基-3-溴-5-氟吡啶的合成工艺
CN114230553B (zh) * 2020-09-09 2023-05-05 凯特立斯(深圳)科技有限公司 一种左旋烟碱的不对称合成方法
CN111875620B (zh) * 2020-09-28 2020-12-11 上海美迪西生物医药股份有限公司 吡唑并嘧啶类大环衍生物及其应用
WO2024017305A1 (zh) * 2022-07-20 2024-01-25 广州嘉越医药科技有限公司 含氰基取代的大环类化合物的用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102971322A (zh) * 2010-05-20 2013-03-13 阵列生物制药公司 作为trk激酶抑制剂的大环化合物
WO2017075107A1 (en) * 2015-10-26 2017-05-04 Nanda Nisha Point mutations in trk inhibitor-resistant cancer and methods relating to the same

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6221335B1 (en) 1994-03-25 2001-04-24 Isotechnika, Inc. Method of using deuterated calcium channel blockers
US6334997B1 (en) 1994-03-25 2002-01-01 Isotechnika, Inc. Method of using deuterated calcium channel blockers
JP3696884B2 (ja) 1994-03-25 2005-09-21 アイソテクニカ、インコーポレーテッド ジュウテリウム化による薬物の効能の増強
US6440710B1 (en) 1998-12-10 2002-08-27 The Scripps Research Institute Antibody-catalyzed deuteration, tritiation, dedeuteration or detritiation of carbonyl compounds
EP1104760B1 (en) 1999-12-03 2003-03-12 Pfizer Products Inc. Sulfamoylheteroaryl pyrazole compounds as anti-inflammatory/analgesic agents
TW200413273A (en) 2002-11-15 2004-08-01 Wako Pure Chem Ind Ltd Heavy hydrogenation method of heterocyclic rings
AU2006299424A1 (en) 2005-10-06 2007-04-12 Auspex Pharmaceuticals, Inc. Deuterated inhibitors of gastric H+, K+-ATPase with enhanced therapeutic properties
US7750168B2 (en) 2006-02-10 2010-07-06 Sigma-Aldrich Co. Stabilized deuteroborane-tetrahydrofuran complex
EP1873157A1 (en) 2006-06-21 2008-01-02 Bayer Schering Pharma Aktiengesellschaft Pyrazolopyrimidines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
TWI458729B (zh) 2008-10-22 2014-11-01 Array Biopharma Inc 作為TRK激酶抑制劑之經取代吡唑并〔1,5-a〕嘧啶化合物
CA2866143C (en) 2012-03-09 2020-08-04 Lexicon Pharmaceuticals, Inc. Pyrazolo[1,5-a]pyrimidine-based compounds, compositions comprising them, and methods of their use
EA031863B1 (ru) 2014-01-24 2019-03-29 ТиПи ТЕРАПЬЮТИКС, ИНК. Диарильные макроциклы в качестве модуляторов протеинкиназ
PL3699181T3 (pl) 2014-11-16 2023-05-22 Array Biopharma, Inc. Postać krystaliczna wodorosiarczanu (s)-n-(5-((r)-2-(2,5-difluorofenylo) - pirolidyn-1-ylo)-pirazolo[1,5-a]pirimidyn-3-ylo)-3-hydroksypirolidyno-1-karboksyamidu
WO2016161572A1 (en) * 2015-04-08 2016-10-13 Merck Sharp & Dohme Corp. TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF
US10316044B2 (en) 2015-07-02 2019-06-11 Tp Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
JP6812059B2 (ja) 2015-07-07 2021-01-13 塩野義製薬株式会社 TrkA阻害活性を有する複素環誘導体
CN106008639B (zh) 2016-03-11 2019-01-08 深圳市塔吉瑞生物医药有限公司 用于预防或治疗fxr-介导疾病的胆烷酸化合物
SG11201808559PA (en) 2016-04-04 2018-10-30 Loxo Oncology Inc Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2019144885A1 (zh) * 2018-01-23 2019-08-01 深圳市塔吉瑞生物医药有限公司 取代的吡唑并[1,5-a]嘧啶类的大环化合物

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102971322A (zh) * 2010-05-20 2013-03-13 阵列生物制药公司 作为trk激酶抑制剂的大环化合物
WO2017075107A1 (en) * 2015-10-26 2017-05-04 Nanda Nisha Point mutations in trk inhibitor-resistant cancer and methods relating to the same

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
D. FLEISHERS. RAMONH. BARBRA: "Improved oral drug delivery: Solubility limitations overcome by the use of prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, no. 2, 1996, pages 115 - 130
DRILON. A. ET AL., CANCER DISCOV., vol. 7, no. 9, 2017, pages 1 - 10
J. A. ELWICH ,ZHANG QINGLIAN : "3. Mechanism research", BASIC CHEMISTRY AND APPLICATIONS OF ISOTOPES, 31 January 1987 (1987-01-31), CN, pages 206 - 207, XP009522636 *
T. HIGUCHIV. STELLA: "Prodrugs as Novel Delivery Systems", vol. 14, 1987, A.C.S. SYMPOSIUM SERIES, article "Bioreversible Carriers in Drug Design"

Also Published As

Publication number Publication date
CN109575025B (zh) 2020-09-11
EP3744723B1 (en) 2023-10-18
JP2021511353A (ja) 2021-05-06
JP7162915B2 (ja) 2022-10-31
EP3744723A4 (en) 2021-05-19
CN111848622A (zh) 2020-10-30
EP3744723A1 (en) 2020-12-02
US20210047330A1 (en) 2021-02-18
CN109575025A (zh) 2019-04-05
US11345703B2 (en) 2022-05-31

Similar Documents

Publication Publication Date Title
CN109575025B (zh) 取代的吡唑并[1,5-a]嘧啶类的大环化合物
AU2013296627B2 (en) Deuterated ibrutinib
CN111362949B (zh) 一种取代的吡唑并[1,5-a]嘧啶化合物及其药物组合物及用途
CN110914277B (zh) 咪唑并[1,2-b]嘧啶并[4,5-d]哒嗪-5(6H)-酮类化合物及其应用
CN109851638B (zh) 取代的二氨基嘧啶化合物
US11512074B2 (en) Substituted diamino heterocyclic carboxamide compound and a composition containing the compound and use thereof
EP3560921A1 (en) Quinazoline compound and preparation method, application, and pharmaceutical compostion thereof
JP2023538090A (ja) Btk阻害剤としての架橋二環式化合物
JP2022521491A (ja) タンパク質アルギニンメチルトランスフェラーゼ5(prmt5)の選択的阻害剤
WO2019134573A1 (zh) 一种氘代二苯氨基嘧啶类化合物的制备方法及其晶型
CN109776544A (zh) 吡唑并[3,4-d]嘧啶类化合物及其制备方法和用途
WO2019223671A1 (zh) 取代的氨基喹唑啉类化合物及其药物组合物及其用途
CN113336774B (zh) 作为trk抑制剂的取代的手性二芳基大环化合物
JP6523259B2 (ja) イコチニブリン酸塩の新規な多形及びその使用
EP3816162A1 (en) Diarylpyrazole compound, composition comprising same, and use thereof
KR101804449B1 (ko) 아이코티닙의 다형체 형태 및 이의 용도
TW202406904A (zh) Cdk抑制劑、醫藥組成物及其治療應用
WO2020168963A1 (zh) 取代的芳香稠合环衍生物及其组合物及用途
WO2019015689A1 (zh) 手性3-奎宁环酮类化合物、制备方法及用途
WO2021185348A1 (zh) 取代的丙烯酰胺衍生物及其组合物及用途
WO2019015690A1 (zh) 氘代3-奎宁环酮类化合物、制备方法及用途
TW202200589A (zh) Mll1抑制劑及抗癌劑
WO2019034147A1 (zh) 1-甲基-色氨酸类化合物及其制备方法和用途
JP2016520624A (ja) イコチニブマレイン酸塩の多形及びその使用

Legal Events

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

Ref document number: 19743399

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020540580

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019743399

Country of ref document: EP

Effective date: 20200824