CN112480100A - Pyrrolidone derivatives - Google Patents

Pyrrolidone derivatives Download PDF

Info

Publication number
CN112480100A
CN112480100A CN201910856406.2A CN201910856406A CN112480100A CN 112480100 A CN112480100 A CN 112480100A CN 201910856406 A CN201910856406 A CN 201910856406A CN 112480100 A CN112480100 A CN 112480100A
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
compounds
acceptable salt
equiv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201910856406.2A
Other languages
Chinese (zh)
Other versions
CN112480100B (en
Inventor
余宁辉
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Conway Guangzhou Biotechnology Co ltd
Original Assignee
Conway Guangzhou Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Conway Guangzhou Biotechnology Co ltd filed Critical Conway Guangzhou Biotechnology Co ltd
Priority to CN201910856406.2A priority Critical patent/CN112480100B/en
Publication of CN112480100A publication Critical patent/CN112480100A/en
Application granted granted Critical
Publication of CN112480100B publication Critical patent/CN112480100B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D471/02Heterocyclic 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 system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to pyrrolidone derivatives and pharmaceutically acceptable compositions thereof which are useful as inhibitors of methionine aminopeptidase.

Description

Pyrrolidone derivatives
Technical Field
The present invention relates generally to pyrrolidone derivatives as inhibitors of methionyl aminopeptidase 2 and pharmaceutical compositions containing them and their use for the treatment of diseases, disorders and conditions associated with methionyl aminopeptidase 2 including cancer and obesity.
Background
In 1990 Judah Folkman and its group published a pioneering finding about the anti-angiogenic activity of the natural compound fumagillin (Nature 1990; 348: 555-. The molecular target of fumagillin was validated as methionyl aminopeptidase 2(MetAP-2) (Proc Natl Acad Sci USA 1997; 94: 6099-. (Am J Pathol 2001; 159: 721-. Experimental studies have demonstrated that MetAP-2 plays an important role in the proliferation and apoptotic pathways of Cancer cells (Cancer Res 2003; 63: 7861-7869; J Biol Chem 1993; 268: 10796-10801). The reduction of tumor metastasis by inhibition of MetAP-2 is not only associated with its anti-angiogenic effect, but also with the physical interaction between MetAP-2 and the metastasis associated protein (i.e., S100A4) (J Biol Chem 2002; 277: 26396-26402). Furthermore, inhibition of MetAP-2 may impair mRNA telomerase expression, contributing to a reduction in tumor proliferation (Am J Pathol 2001; 159: 721-.
Different structures of MetAP-2 inhibitors have been developed, among which irreversible MetAP-2 inhibitors are represented by fumagillin and its analogs such as TNP-470, CKD-732 and PPI-2458, which can inactivate enzymes by covalently modifying histidine residues in the active site (Curr Med Chem 2012; 19: 1021-. TNP-470 has been introduced into phase II clinical trials in renal cell carcinoma, breast, cervical and pancreatic Cancer and glioblastoma (Clin Cancer Res 2001; 7: 1198-. However, clinical trials found that TNP-470 has the following disadvantages: poor pharmacokinetics, low oral bioavailability and dose-limiting CNS side effects (Cancer Chemother Pharmacol 2004; 54: 308-. To overcome these disadvantages of TNP-470, modification of the side chain of C6 can lead to promising analogs such as CDK-732 and PPI-2458(Curr Med Chem 2012; 19: 1021-. HMPA co-TNP-470 was found to significantly enhance and prolong the activity of TNP-470 while preventing crossing the blood-brain barrier to eliminate CNS-related toxic side effects (Nat Med 2004; 10: 255-. Recently, the nano-polymer micelle of TNP-470 was found to significantly inhibit tumor growth in mice without causing nerve damage (Nat Biotechnol 2008; 26: 799-.
Over the past few years, a number of reversible inhibitors with different branches have been developed. These analogs are characterized by a core structure that interacts with two metal ions in the active site, and in addition has one or two chains that can occupy the protein (Curr Med Chem 2012; 19: 1021-. 2-hydroxy-3-aminoamide is the core structure of ubenimex, a natural inhibitor of aminopeptidases. Based on their framework, a series of MetAP-2 inhibitors have been developed (Bioorg Med Chem Lett 2004; 14: 865-868). The Bengamides compounds comprise a series of marine natural products, and the fusion keto-amino acid derivatives are non-selective inhibitors of MetAP-1 and MetAP-2, have strong antiproliferative effects and can induce cell arrest in G1 and G2/M phases (J Biol Chem 2003; 278: 52964-52971). LAF389 is a synthetic analogue of bengamide that has entered phase I clinical trials, but further studies have been hampered by severe cardiovascular dose-limiting toxicities (Anticancer Drugs 2007; 18: 219-225). Validation of reversibly selective MetAP-2 inhibitors suitable for oral administration led to the discovery of anthranilate sulfonamides (Bioorg Med Chem Lett 2006; 16: 3574-. A-800141 is a highly selective and reversible inhibitor against MetAP-2 and can cause cell cycle arrest in the G1 phase (Proc Natl Acad Sci USA 2008; 105: 1838-. High throughput screening of CO 2+ has led to the discovery of 1,2, 4-triazole inhibitors that are selective for MetAP-2 (J Med Chem 2007; 50: 3777-3785). A group of compounds with pyridyl pyrimidine core was found to strongly inhibit MatAp1 and MetAP-2 with very low selectivity (Angew Chem Int Ed Engl 2006; 45: 3772-3775). Nitrogoline, an antibiotic used for the treatment of urinary tract infections, was identified from HTS libraries as a MetAP-2 inhibitor (J Natl Cancer Inst 2010; 102: 1855- -. Other reversible inhibitors are less potent on MetAP-2 or also have other protease inhibitory activity, making these inhibitors unsuitable as clinical drug candidates. However, these studies lay the foundation for the rational design of more specific, more potent, less toxic and better pharmacological MetAP-2 inhibitors.
Disclosure of Invention
The present invention provides pyrrolidone derivatives and pharmaceutically acceptable salts thereof. The invention also provides pharmaceutical compositions comprising the pyrrolidone derivatives, and their use in the treatment of diseases, disorders and conditions associated with MetAP-2, including cancer and obesity.
One aspect of the present invention provides a compound represented by the following formula I:
Figure BDA0002198418950000031
wherein X is selected from C or N,
a is selected from the following groups:
Figure BDA0002198418950000032
b is selected from the following groups:
Figure BDA0002198418950000033
n is 0,1, 2,3 or 4;
R1and R2Each independently selected from:
halogen and-CN;
or a pharmaceutically acceptable salt thereof.
Detailed Description
1.General description of the Compounds of the invention
In certain embodiments, the present invention provides inhibitors of MetAP-2. In some embodiments, the compositions include a compound described by a formula herein, or a pharmaceutically acceptable salt thereof, with the differences being specifically described in the definitions below.
2.Compounds and definitions
The compounds of the present invention include the compounds described generally above and are further illustrated by the classes, subclasses, and species disclosed herein. The following definitions are followed herein unless otherwise indicated. For the purposes of the present invention, chemical elements are defined according to the periodic Table of the elements (CAS version, Handbook of Chemistry and Physics, 75 th edition). In addition, the general principles of Organic Chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry", 5th Ed. Ed.: smith, m.b. and March, j., John Wiley & Sons, New York: 2001, the entire contents of which are incorporated herein by reference.
The term "alkylene" refers to a divalent alkyl group. "alkylene chain" refers to a polymethylene group, i.e., - (CH)2)nN is a positive integer, preferably 1 to 6,1 to 4,1 to 3, 1 to 2 or 2 to 3. A substituted alkylene chain refers to a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include the aliphatic substituents described below.
The term "halogen" denotes F, Cl, Br or I.
The term "pharmaceutically acceptable salt" as used herein, means a salt, which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals at a reasonable benefit/risk ratio without excessive toxicity, irritation, or allergic response. Pharmaceutically acceptable salts are well known in the art. Pharmaceutically acceptable salts are described in detail, for example, in j.pharmaceutical Sciences, 1977,66,1-19, to s.m.berge et al, which is incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. By way of example, pharmaceutically acceptable non-toxic acid addition salts are salts formed with inorganic acids (such as hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric acids), organic acids (such as acetic, oxalic, maleic, tartaric, citric, succinic, or malonic acids), or by other methods known in the art, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, caproic acid, hydroiodic acid, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, dodecylsulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, phosphates, pivalates, propionates, Stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, pentanoates, and the like.
Suitable bases for derivatization to form salts include alkali metals, alkaline earth metals, ammonium, and N+(C1-4Alkyl radical)4And (3) salt. Representative alkali or alkaline earth metals include sodium, lithium, potassium, calcium, magnesium, and the like. Other pharmaceutically acceptable salts include, where appropriate, non-toxic ammonium, quaternary ammonium, and cationic amines formed using counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkyl sulfonates, and aryl sulfonates.
Unless otherwise indicated, structures described herein are also meant to include all isomeric forms (e.g., enantiomers, diastereomers, and geometric isomers or conformations) of the structure; for example, the R and S configurations, Z and E double bond isomers, and Z and E conformational isomers of the various asymmetric centers. Thus, single stereochemical isomers as well as enantiomeric, diastereomeric and geometric (or conformational) isomeric mixtures of the compounds of the invention are within the scope of the invention. Unless otherwise indicated, all isomeric forms of the compounds of the invention are within the scope of the present invention.
In addition, unless otherwise indicated, structures described herein are also meant to include compounds that differ only in the substitution of one or more isotopically enriched atoms. For example, compounds having the structure of the invention, include replacement of hydrogen with deuterium or tritium, or with deuterium or tritium13C-or14C-rich carbon substitution is within the scope of the present invention. In some embodiments, the group contains one or more deuterium atoms.
Still further, the compounds of formula I also include isotopically labeled forms thereof. The isotopically labeled form of the compound of formula I is identical to the compound, in that only one or more atoms are replaced by an atom having a different atomic mass or mass number than the atom usually found in nature. Are readily commercially available by well known methods andexamples of isotopes that can be incorporated into compounds of formula I include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example2H、3H、13C、14C、15N、18O、17O、31P、32P、35S、18F and36and (4) Cl. Compounds of formula I containing one or more of the foregoing isotopes and/or other isotopes of other atoms, as well as prodrugs or pharmaceutically acceptable salts thereof, are part of this invention. Isotopically-labelled compounds of formula I can be used in a variety of advantageous ways. For example, isotopically-labelled compounds of formula I (e.g. having incorporated therein a compound such as3H or14A radioisotope of C) may be used in tissue distribution assays of drugs and/or substrates. Tritium (A) among these radioactive isotopes because of its simple preparation and excellent detectability3H) And carbon-14 (14C) Is particularly preferred. Heavier isotopes (e.g. deuterium (ll) (ll)) due to higher metabolic stability2H) The isotopically labeled compounds formed by the introduction of the compounds of formula I are more therapeutically advantageous. Higher metabolic stability translates directly into increased in vivo half-life or lower doses, which in most cases represent preferred embodiments of the invention. Isotopically-labelled compounds of formula I can be prepared by replacing a non-isotopically-labelled reactant with a readily available isotopically-labelled reactant in the synthetic schemes and associated disclosure described in the examples and preparations section herein.
Deuterium (b) may also be used in order to control the oxidative metabolism of compounds by kinetic first-order isotopic effect2H) Incorporated into the compounds of formula I. Kinetic first-order isotopic effects refer to the effect of a change in the rate of chemical reaction caused by the exchange of isotopic nuclei, which in turn is caused by a change in the ground-state energy required for covalent bond formation after isotopic exchange. Exchange of heavier isotopes generally results in a reduction in the ground state energy of the chemical bonds, resulting in a reduction in the rate in rate-limited bond cleavage reactions. If bond breakage occurs at or near the saddle point region corresponding to the multi-product reaction, the product distribution ratio will change significantly. In particular to: if deuterium is bonded to a carbon atom in a non-exchangeable position, kM/kDRate differences of 2-7 are common. If this rate difference is successfully applied to the easily oxidizable compound of formula I, the in vivo properties of this compound can be drastically altered and its pharmacokinetic properties improved.
During drug discovery and development, one skilled in the art is able to optimize pharmacokinetic parameters while maintaining desirable in vitro properties. It is reasonable to assume that many compounds with poor pharmacokinetic profiles are susceptible to oxidative metabolism. The currently available in vitro liver microsomal assays provide useful information about this type of oxidative metabolic processes, which in turn allows for the rational design of deuterated compounds of formula I, i.e., improved stability by counteracting such oxidative metabolism. The pharmacokinetic profile of the compound of formula I thus obtained is significantly improved, quantitatively expressed as the in vivo half-life (t/2), maximum effective concentration (C)max) Increase in area under the dose response curve (AUC) and F, and decrease in clearance, dose and material costs.
The following is for the purpose of illustrating the foregoing: compounds of formula I having multiple potential sites of oxidative metabolic attack (e.g., benzylic hydrogen atoms and hydrogen atoms bonded to nitrogen atoms) can be prepared as a series of analogs in which various combinations of hydrogen atoms are replaced with deuterium atoms, so that some, most, or all of these hydrogen atoms have been replaced with deuterium atoms. The half-life measurement enables an efficient and accurate determination of the degree of improvement in oxidative metabolic resistance. From this determination, it is clear that the aforementioned deuterium-hydrogen exchange results in a 100% increase in half-life of the original compound.
Deuterium-hydrogen exchange of a compound of formula I may also be effective to alter the metabolic profile of the original compound, thereby reducing or eliminating undesirable toxic metabolites. For example, if a toxic metabolite is produced by oxidative hydrocarbon (C-H) bond cleavage, it can be reasonably assumed that the deuterated analog will greatly reduce or eliminate the production of the undesired metabolite even if its oxidation reaction is not rate-dependent. Further information on the prior art of deuterium-hydrogen exchange can be found in the following documents: hanzlik et al, J.org.chem.55,3992-3997,1990; reider et al, j.org.chem.52,3326-3334,1987; foster, adv. drug res.14,1-40,1985; gillette et al, Biochemistry 33(10)2927-2937, 1994; and Jarman et al Carcinogenesis 16(4),683-688, 1993.
Any definitions set forth herein for different chemical groups are inclusive of their definitions as any single group or combination of groups. References herein to different embodiments include the embodiment as a stand-alone embodiment or in combination with any other embodiments/portions of embodiments.
3.Description of exemplary Compounds
As an embodiment, the present invention provides a compound represented by the following formula I:
Figure BDA0002198418950000071
wherein X is selected from C or N,
a is selected from the following groups:
Figure BDA0002198418950000072
b is selected from the following groups:
Figure BDA0002198418950000073
n is 0,1, 2,3 or 4;
R1and R2Each independently selected from:
halogen and-CN;
or a pharmaceutically acceptable salt thereof.
As an embodiment, the present invention provides a compound represented by the following formula II:
Figure BDA0002198418950000074
wherein X is selected from C or N,
a is selected from the following groups:
Figure BDA0002198418950000081
b is selected from the following groups:
Figure BDA0002198418950000082
n is 0,1, 2,3 or 4;
R1and R2Each independently selected from:
halogen and-CN;
or a pharmaceutically acceptable salt thereof.
As a still further embodiment, the present invention provides a compound (compound 1) having the structure:
Figure BDA0002198418950000083
as another still further embodiment, the present invention provides a compound (compound 2) having the structure:
Figure BDA0002198418950000084
as a still further embodiment, the present invention provides a compound (compound 3) having the structure:
Figure BDA0002198418950000091
as another still further embodiment, the present invention provides a compound (compound 4) having the structure:
Figure BDA0002198418950000092
as a still further embodiment, the present invention provides a compound (Compound 5) having the structure:
Figure BDA0002198418950000093
as another still further embodiment, the present invention provides a compound (compound 6) having the structure:
Figure BDA0002198418950000094
as yet another embodiment, the present invention relates generally to compositions comprising the above compounds, which have therapeutic or prophylactic effects on cancers such as liver cancer, cholangiocarcinoma and malignant mesothelioma, pancreatic cancer, head and neck cancer, and hemangioma.
As yet another embodiment, the present invention is generally directed to a unit dosage form comprising a pharmaceutical composition disclosed herein.
As yet another embodiment, the present invention relates generally to methods of treating type II diabetes in a mammal in need thereof with compositions comprising the above compounds.
4.Drugs, medicaments and administration
Pharmaceutically acceptable compositions
According to another embodiment, the present invention provides a composition comprising a compound of the present invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant or vehicle. The amount of a compound of the invention in a composition is an effective amount to measurably modulate MetAP-2 or a mutant thereof in a biological sample or patient. In certain embodiments, the amount of compound in the compositions of the invention is effective to measurably modulate MetAP-2 or a mutant thereof in a biological sample or patient. In certain embodiments, the compositions of the present invention are prepared as medicaments and used to administer to a patient in need of such compositions.
The term "patient" or "subject" as used herein refers to an animal, preferably a mammal, most preferably a human.
The term "pharmaceutically acceptable carrier, adjuvant or vehicle" refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound with which it forms a medicament. Pharmaceutically acceptable carriers, adjuvants or vehicles for use in the compositions of the invention include, but are not limited to: ion exchangers, aluminum oxide, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances, such as phosphates, glycine, sorbic acid, potassium sorbate, water and partial glyceride mixtures of salts or electrolytes and vegetable fatty acids (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
"pharmaceutically acceptable derivative" refers to any non-toxic salt, ester salt or other derivative of a compound of the present invention. These derivatives are capable of providing, directly or indirectly, a compound of the invention, or an inhibitory active metabolite or residue thereof, upon administration to a subject.
Modes of administration of the compositions of the present invention include oral, parenteral, inhalation spray, topical, rectal, nasal, buccal, vaginal or implant administration. The term "parenteral administration" as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the composition is administered orally, intraperitoneally, or intravenously. Sterile injectable forms of the compositions of the present invention include aqueous or oleaginous suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation is also a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable carriers and solvents include water, ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
For the purposes described above, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids (such as oleic acid and its glyceride derivatives) and natural pharmaceutically acceptable oils (such as olive oil or castor oil, especially their polyoxyethylated versions) are useful in the preparation of injectables. These oil solutions or suspensions also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents, which are commonly used in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants may also be used for pharmaceutical purposes, such as tweens, spans, and other emulsifying agents or bioavailability enhancers, which are commonly used to prepare pharmaceutically acceptable solid, liquid, or other dosage forms.
The pharmaceutically acceptable compositions of the present invention are administered orally in any orally acceptable dosage form, exemplary oral dosage forms include capsules, tablets, aqueous suspensions, and solutions. In the case of oral tablets, commonly used carriers include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in capsule form, useful diluents include lactose and dried corn starch. When the oral route is carried out using aqueous suspensions, the active ingredient is mixed with emulsifying and suspending agents. If desired, sweetening, flavoring or coloring agents may also be optionally added.
Alternatively, the pharmaceutically acceptable compositions of the present invention are administered rectally in the form of suppositories which can be prepared by mixing the active ingredient with suitable non-irritating excipients. These excipients are solid at room temperature, but liquid at rectal temperature and will therefore melt in the rectum to release the drug, including cocoa butter, beeswax and polyethylene glycols.
The pharmaceutically acceptable compositions of the present invention may also be administered topically, particularly where the target of treatment contains areas or organs (including ocular, dermal or lower intestinal disorders) that are susceptible to topical application. Suitable topical agents can be readily prepared for each of these regions or organs.
Topical administration for the lower intestinal tract may be achieved by rectal suppository dosage forms (see above) or in suitable enema dosage forms. Topical transdermal patches are also available.
For topical administration, the pharmaceutically acceptable compositions of the present invention may be formulated in a suitable ointment by suspending or dissolving the active ingredient in one or more carriers. Exemplary carriers for topical administration include mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutically acceptable compositions of the present invention may be formulated in a suitable lotion or cream containing the active ingredient suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
The pharmaceutically acceptable compositions of the present invention may also optionally be administered by nasal aerosol or inhalation. These compositions may be prepared as aqueous salt solutions according to techniques well known in the art of pharmaceutical formulation, wherein benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons and/or other conventional solubilizing or dispersing agents may also be used.
Most preferably, the pharmaceutically acceptable compositions of the present invention are administered by oral administration. Such agents may be taken with or without meals. In some embodiments, the pharmaceutically acceptable compositions of the present invention are not taken with a meal; in other embodiments, the pharmaceutically acceptable compositions of the present invention are taken with meals.
When used in combination with a carrier material to produce a single dosage form, the amount of a compound of the present invention may vary depending on the subject being treated and the corresponding mode of administration. Preferably, the compositions of the invention should be in a dosage form that enables administration of an effective dose of the compound of 0.01-100mg/kg body weight/day to a patient receiving administration.
It will also be understood that the specific dose and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the compound employed, the age, body weight, general health, sex, diet, time of administration, rate of drug excretion, drug compatibility, the judgment of the treating physician, and the severity of the disease undergoing therapy. The amount of the compound of the present invention in the composition also depends on the particular compound in the composition.
Use of compounds and pharmaceutically acceptable compositions
The compounds provided according to the invention can be used as inhibitors of MetAP-2.
The compounds provided according to the invention can be used for the production of medicaments, in particular as MetAP-2 inhibitors.
The compounds provided according to the invention can be used to treat or prevent any cancer with a significant degree of angiogenesis (e.g., lung, breast, prostate, head and neck, esophagus, pancreas, liver, colon, or kidney cancer) or to induce metastatic cancer (e.g., colon, breast, liver, head and neck, stomach, and melanoma). These compounds may be used in monotherapy, or in combination therapy with radiation therapy or chemotherapy.
The compounds provided according to the invention can also be used for the treatment or prevention of liver cancer, cholangiocarcinoma, malignant mesothelioma, pancreatic cancer, head and neck cancer and hemangioma.
The compounds provided according to the invention may also be used for the treatment or prevention of obesity. The present invention provides a method of treating a type II diabetic patient in need of such treatment comprising administering to the patient an effective amount of a compound of formula II or a pharmaceutically acceptable salt thereof. Preferably, the patient is a human. The present invention provides a method of treating nonalcoholic steatohepatitis in a patient in need thereof, comprising administering to the patient an effective amount of a compound of formula II or a pharmaceutically acceptable salt thereof.
The present invention provides the use of a compound of formula I and formula II, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of obesity. The invention also provides the use of a compound of formulae I and II, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in a therapy to provide therapeutic weight loss.
Examples
As described in the examples below, the present invention describes, by certain exemplary embodiments, the steps of preparing certain compounds using general methods, but these general methods are not limited to the synthesis of the specific compounds in the examples. These general procedures, as well as others known to those of ordinary skill in the art, are equally applicable to the synthesis of all of the compounds described herein, as well as derivatives and analogs thereof.
The compound numbers used in the following examples correspond to the compound numbers described above.
1H-NMR and13C-NMR was measured using a 400MHz Varian Mercury nuclear magnetic instrument. Proton chemical shifts were referenced to residual proton resonance in CDCl3 (7.26ppm) as an internal standard. Carbon chemical shifts were referenced to the deuterated solvent signal (77.20ppm) in CDCl3 as an internal standard.
LC-MS spectra were determined by Shimadzu LC-MS2020 and separated using an Agilent C18 column (Eclipse XDB-C18,5um, 2.1X 50mm) at a flow rate of 1mL/min, mobile phase A: water containing 0.1% formic acid; mobile phase B: acetonitrile solution containing 0.1% formic acid. The gradient method shown in the table below was used.
Time (min) Mobile phase A Mobile phase B
0 95 5
3 0 100
4 0 100
4.05 95 5
HPLC was carried out using an Agilent 1200HPLC analyzer using a Zorbax Eclipse XDB C18 column (2.1X 150mm) at a flow rate of 1 mL/min. Mobile phase A: water with 0.1% TFA; mobile phase B: 0.1% TFA in acetonitrile. A general method with the following gradient was used.
Time (min) Mobile phase A Mobile phase B
0 95 5
15 0 100
16 0 100
16.5 95 5
16.5 End up
Example 1
Preparation of 3- (5- (3, 5-difluorobenzyl) -1,2, 4-oxadiazol-3-yl) -3-hydroxy-1- (1H-indol-5-yl)
Pyrrolidin-2-one (Compound 1)
Figure BDA0002198418950000141
The reaction formula of example 1 is as follows:
Figure BDA0002198418950000151
step 1: potassium tert-butoxide (720.72mg, 6.435mmol, 1.5 equiv.) was added to a solution of compound 1-1(700mg, 4.29mmol, 1.0 equiv.) in tetrahydrofuran (10 mL). The mixture was stirred at room temperature for 30 minutes, and compound 1-2(0.64mL, 5.15mmol, 1.2 equiv.) was added to the mixture. The mixture was stirred for 2 hours at room temperature. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was then diluted with water (10mL) and extracted with ethyl acetate (2X 10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate, 84:16) to give compound 1-3(1.23g, 95%) as a white solid. TLC conditions: petroleum ether ethyl acetate 5:1, UV254 nm. R of Compound 1-1f0.2, R of Compounds 1-3f=0.6。
Step 2: a solution of compounds 1-3(1.23g, 4.06mmol, 1.0 equiv.) in tetrahydrofuran (30mL) was hydrogenated catalyzed by Pd/C (0.246g, 20% wt) at room temperature for 3 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane: methanol, 96:4) to obtain compounds 1-4(1g, 91%) as pale yellow solids. TLC conditions: dichloromethane: methanol 10:1, UV254 nm. R of Compounds 1 to 3f0.9, R of Compounds 1-4f=0.6。
And step 3: compound 1-5(684.8mg, 4.028mmol, 1.1 equiv.) was added to a solution of compound 1-4(1.0g, 3.662mmol, 1.0 equiv.) in ethanol (30 mL). The mixture was refluxed at 100 ℃ for 5 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (dichloromethane: methanol, 10:1) to obtain compounds 1 to 6(450mg, 32%).
And 4, step 4: a solution of compounds 1-6(3.8g, 9.9mmol, 1.0 equiv.), ammonium chloride (1.587g, 29.6mmol, 3.0 equiv.), 2- (7-aza-1H-benzotriazol-1-yl) -1,1,3, 3-tetramethyluronium hexafluorophosphate (7.58g, 19.8mmol, 2.0 equiv.) and N, N-diisopropylethylamine (5.11g, 39.6mmol, 4.0 equiv.) in dimethylformamide (80mL) was stirred at room temperature under a nitrogen atmosphere for 2 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was diluted with ethyl acetate and water and extracted with ethyl acetate (2X 80 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (dichloromethane: methanol, 100: 1) to give compounds 1-7(2.43g, 64%) as a white solid. TLC conditions: dichloromethane: methanol 20:1, UV254 nm. R of Compounds 1 to 6f0.2, R of Compounds 1 to 7f=0.4。
And 5: phosphorus oxychloride (2.4g, 15.68mmol, 3.0 equiv.) and pyridine (2.48g, 31.36mmol, 6.0 equiv.) are added to compounds 1-7(2.0g, 5.16mmol, 1.0 equiv.)Dry acetonitrile (40 mL). The mixture was stirred at 70 ℃ for 1.5 hours under nitrogen atmosphere. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was cooled to room temperature, quenched with saturated sodium bicarbonate solution and extracted with ethyl acetate (3 × 40 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate, 3:1) to obtain compounds 1 to 8(1.36g, 72%) as a white solid. TLC conditions: petroleum ether: ethyl acetate ═ 1:1, UV254 nm. R of Compounds 1 to 7f0.05, R of Compounds 1 to 8f=0.6。
Step 6: a mixture of compounds 1-8(1.32g, 3.63mmol, 1.0 equiv.), hydroxylamine hydrochloride (1.25g, 18.04mmol, 5.0 equiv.) and sodium carbonate (1.91g, 18.04mmol, 5.0 equiv.) in ethanol/water (50mL/32.5mL) was stirred at reflux under nitrogen for 4 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was filtered and concentrated under reduced pressure. The residue was diluted with water and extracted with ethyl acetate (3X 100 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (dichloromethane: methanol, 100: 1)
Figure BDA0002198418950000171
80: 1) to give compounds 1-9(690mg, 48%) as white solids. TLC conditions: dichloromethane: methanol 15:1, UV254 nm. R of Compounds 1 to 8f0.9, R of Compounds 1 to 9f=0.3。
And 7: compounds 1-10(300.0mg, 1.73mmol, 1.0 equiv.), 2- (1H-benzotriazol-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate (555.0mg, 1.73mmol, 1.0 equiv.), 1-hydroxybenzotriazole (47.0mg, 0.35mmol, 0.2 equiv.) and N, N-diisopropylethylamine (1.1g, 8.65mmol, 5.0 equiv.) were added to a solution of compounds 1-9(690.0mg, 1.73mmol, 1.0 equiv.) in dimethylformamide (10 mL). The mixture was stirred at room temperature for 2 hours under nitrogen atmosphere, and then the mixture was stirred at 110 ℃ for 3 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was diluted with ethyl acetate and water (10mL),extract with ethyl acetate (2X 10 mL). The combined organic layers were washed 3 times with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate, 5: 2) to give compounds 1 to 11(240mg, 26%) as white solids. TLC conditions: petroleum ether: ethyl acetate ═ 3:1, UV254 nm. R of Compounds 1 to 9f0, R of Compounds 1 to 11f0.3. TLC conditions: dichloromethane: methanol 20:1, UV254 nm. R of Compounds 1 to 9f0.3, R of Compounds 1-11f=0.9。
And 8: tetrabutylammonium perchlorate (144mg, 1.12mmol, 3.0 equiv.) and sodium ethoxide (76mg, 1.12mmol, 3.0 equiv.) are added to a solution of compounds 1-11(200mg, 0.374mmol, 1.0 equiv.) in t-butanol/dichloromethane (6mL/2 mL). The mixture was stirred at 70 ℃ for 1 hour under nitrogen atmosphere. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was then cooled to 0 ℃, saturated sodium sulfite solution and ammonium chloride solution were added, and extracted with dichloromethane (2 × 10 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC (petroleum ether: ethyl acetate, 1: 1) to give compounds 1-12(53mg, 26%). TLC conditions: petroleum ether: ethyl acetate ═ 1:1, UV254 nm. R of Compounds 1 to 11f0.8, R of Compounds 1-12f=0.6。
And step 9: a solution of compounds 1-12(45mg, 0.082mmol, 1.0 equiv.) and DBU (62mg, 0.41mmol, 5.0 equiv.) in methanol (3mL) was heated to 80 deg.C and stirred for 4 h. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was then cooled to room temperature and extracted with ethyl acetate (2X 5 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC to give compound 1(5.2mg, 15%) as a yellow solid. LCMS (liquid Crystal Module) [ M +1 ]]:411;1H NMR(CDCl3,400MHz):δ8.24(s,1H),7.72(s,1H),7.41(m,1H),7.35(m,1H),7.23(m,1H),6.84(m,2H),6.673-6.71(m,1H),6.53(s,1H),4.19(s,2H),4.13-4.11(m,1H),3.99-3.97(m,1H),2.97-2.93(m,1H)and 2.57(m,1H).
Examples 2 and 3
Preparation of (S) -3- (5- (3, 5-difluorobenzyl) -1,2, 4-oxadiazol-3-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (Compound 2) and (R) -3- (5- (3, 5-difluorobenzyl) -1,2, 4-oxadiazol-3-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (Compound 3).
Figure BDA0002198418950000181
The equations for examples 2 and 3 are as follows:
Figure BDA0002198418950000182
the racemic compound 3-5-3, 5-difluorobenzyl) -1,2, 4-oxadiazol-3-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (compound 1) (50mg) was isolated by the following method: a Chiralcel OD column (5X 25cm, 20% u03BCm) and a solvent system of n-heptane/ethanol (70:30) were used with a flow rate of 100 ml/min. The first eluting enantiomer was collected and concentrated to give (S) -3- (3- (3, 5-difluorobenzyl) -1,2, 4-oxadiazol-5-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (compound 2). The enantiomer in the second fraction was collected and concentrated to give (R) -3- (3- (3, 5-difluorobenzyl) -1,2, 4-oxadiazol-3-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (Compound 3).
Example 4
Preparation of 3- (5- (3, 5-difluorobenzyl) -1,3, 4-oxadiazol-2-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (compound 4).
Figure BDA0002198418950000191
The reaction formula of example 4 is as follows:
Figure BDA0002198418950000192
step 1: potassium tert-butoxide (10.4g, 92.55mmol, 1.5 equiv.) and compound 4-2(13g, 74.07mmol, 1)2 equiv) was added to a solution of compound 4-1(10g, 61.7mmol, 1.0 equiv) in tetrahydrofuran (100 mL). The mixture was stirred at room temperature overnight. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was then diluted with water (100mL) and extracted with ethyl acetate (2X 100 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give compound 4-3(13.2g, 71%) as a white solid. TLC conditions: petroleum ether ethyl acetate 5:1, UV254 nm. R of Compound 4-1f0.2, R of Compound 4-3f=0.7。
Step 2: Pd/C (1.3g, 10% wt) was added to a solution of compound 4-3(13.2g, 43.7mmol, 1.0 equiv.) in tetrahydrofuran (150mL) and hydrogenated at room temperature for 16 h. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was filtered to give a filtrate and the solvent was removed in vacuo. The residue was recrystallized from petroleum ether and ethyl acetate to give compound 4-4(7.1g, 60%) as a white solid. TLC conditions: petroleum ether ethyl acetate 5:1, UV254 nm. R of Compound 4-3f0.7, R of Compound 4-4f=0.2。
And step 3: compound 4-5(4.9g, 28.7mmol, 1.1 equiv.) is added to a solution of compound 4-4(7.1g, 26.1mmol, 1.0 equiv.) in ethanol (70 mL). The mixture was refluxed at 100 ℃ for 5 hours. TLC analysis of the reaction mixture showed complete conversion to the desired product. The mixture was then filtered and concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography to give compound 4-6(3g, 30%). TLC conditions: dichloromethane methanol 15:1, UV254 nm. R of Compound 4-4fR of compound 4-6 ═ 1f=0.2。
And 4, step 4: the compounds 4-7(258mg, 1.3mmol, 1.0 equiv.), 2- (7-Aza) -1H-benzotriazol-1-yl) -1,1,3, 3-tetramethyluronium hexafluorophosphate (590mg, 1.56mmol, 1.2 equiv.) and N, N-diisopropylethylamine (335mg, 2.6mmol, 2.0 equiv.) were added to a solution of the compound 4-6(500mg, 1.3mmol, 1.0 equiv.) in succession in dimethylformamide (5 mL). The mixture was stirred at room temperature overnight. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then washed with water (10 m)L) and extracted with ethyl acetate (2X 10 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give compound 4-8(350mg, 51%) as a white solid. TLC conditions: dichloromethane methanol 15:1, UV254 nm. R of Compounds 4 to 6f0.2, R of Compounds 4 to 8f=0.8。
And 5: cerium (III) chloride heptahydrate (100mg, 0.27mmol, 0.4 equiv.) was added to a solution of compound 4-8(350mg, 0.66mmol, 1.0 equiv.) in isopropanol (20 mL). The mixture was stirred overnight under an oxygen atmosphere at 70 ℃. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then diluted with water (10mL) and extracted with ethyl acetate (2X 10 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give compounds 4-9(250mg, 70%). TLC conditions: dichloromethane methanol 20:1, UV254 nm. R of Compounds 4 to 8f0.6, R of Compounds 4-9f=0.5。
Step 6: a solution of compounds 4-9(250mg, 0.46mmol, 1.0 equiv.) in methanol (9mL) was added to a solution of sodium hydroxide (368mg, 9.2mmol, 20.0 equiv.) in water (9 mL). The mixture was stirred at reflux for 2 hours. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then cooled to room temperature and extracted with ethyl acetate (2 × 10 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC to give product 4(68mg, 37%) as a white solid. TLC conditions: dichloromethane methanol 20:1, UV254 nm. R of Compounds 4 to 9f0.5, R of compound 4f=0.3。LCMS:[M+1]:404.1H NMR(d6-DMSO,400MHz):δ11.08(s,1H),7.96(m,1H),7.68(s,1H),7.32(m,3H),7.02-6.99(m,4H),6.40(t,J=0.96Hz,1H),3.89-3.80(m,2H),3.12-3.04(m,2H),2.77-2.47(m,5H),2.33(m,1H),2.05(m,1H),1.87(m,2H)and.30-1.26(m,1H)。
Example 5
Preparation of 3-hydroxy-2-oxo-1- (1H-pyrrolo [2,3-b ] pyridin-5-yl) -N- ((1,2,3, 4-tetrahydronaphthalen-2-yl) methyl) pyrrolidine-3-carboxamide (Compound 5).
Figure BDA0002198418950000211
The reaction formula of this example 5 is as follows:
Figure BDA0002198418950000221
step 1: potassium tert-butoxide (720.72mg, 6.435mmol, 1.5 equiv.) is added to a solution of compound 5-1(700mg, 4.29mmol, 1.0 equiv.) in tetrahydrofuran (10 mL). After stirring at room temperature for 30 min, compound 5-2(0.64mL, 5.15mmol, 1.2 equiv.) was added to the mixture. The mixture was stirred at room temperature for 2 hours and TLC analysis of the reaction showed complete conversion to the desired product. The mixture was then diluted with water (10mL) and extracted with ethyl acetate (2X 10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting crude product was purified by silica gel chromatography (petroleum ether: ethyl acetate, 84:16) to give compound 5-3(1.23g, 95%) as a white solid. TLC conditions: petroleum ether ethyl acetate 5:1, UV254 nm. R of Compound 5-1f0.2, R of Compound 5-3f=0.6。
Step 2: Pd/C (0.246g, 20% wt) was added to a solution of compound 5-3(1.23g, 4.06mmol, 1.0 equiv) in tetrahydrofuran (30mL) and hydrogenated at room temperature for 3 hours. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was filtered to give a filtrate and the solvent was removed in vacuo. The residue was purified by silica gel column chromatography (dichloromethane: methanol, 96:4) to obtain compound 5-4(1g, 91%) as a pale yellow solid. TLC conditions: dichloromethane methanol 10:1, UV254 nm. R of Compound 5-3f0.9. R of Compound 5-4f=0.6。
And step 3: compound 5-5(684.8mg, 4.028mmol, 1.1 equiv.) is added to a solution of compound 5-3(1.0g, 3.662mmol, 1.0 equiv.) in ethanol (30 mL). The mixture was refluxed at 100 ℃ for 5 hours. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (dichloromethane: methanol, 10:1) to give compound 5-6(450mg, 32%).
And 4, step 4: compounds 5-7(462.15mg, 2.34mmol, 2.0 equiv.), 1- (3-dimethylaminopropyl)) -3-ethylcarbodiimide hydrochloride (2.242g, 11.7mmol, 10.0 equiv.) and 1-hydroxybenzotriazole (3.756g, 11.7mmol, 1.0 equiv.) are added sequentially to a solution of compounds 5-6(450mg, 1.17mmol, 1.0 equiv.) in dimethylformamide (5 mL). The mixture was stirred at room temperature overnight. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was diluted with water (10mL) and extracted with ethyl acetate (2X 10 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate, gradient elution, 90:10 to 45:55) to give compound 5-8(150mg, 24%) as a white solid. TLC conditions: ethyl acetate 1:1, UV254 nm. R of Compounds 5 to 6f0.5. R of Compounds 5 to 8f=0.3。
Step 5 Compounds 5-8 are converted to Compounds 5-9 in analogy to the preparation of Compounds 4-9.
Step 6: a solution of sodium hydroxide (110.2mg, 2.754mmol, 20.0 equiv.) in water (3mL) was added to a solution of compound 5-9(75mg, 0.138mmol, 1.0 equiv.) in methanol (3 mL). The mixture was then heated and stirred at reflux for 0.5 h. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then cooled to room temperature and extracted with ethyl acetate (2 × 5 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The resulting crude product was purified by preparative TLC (petroleum ether: ethyl acetate, 1: 1) to give compound 5(3mg, 5%) as a white solid. TLC conditions: ethyl acetate 1:1, UV254 nm. R of Compounds 5 to 9f0.4, R of compound 5f=0.1。LCMS:[M+1]:405.1H NMR(DMSO,400MHz):δ11.66(s,1H),8.46(m,1H),8.13(s,1H),8.00(s,1H),7.47(s,1H),7.01(s,4H),6.65-6.64(d,J=1.6Hz,1H),6.44(m,1H),3.91-3.86(m,2H),3.13-3.05(m,2H),2.77(m,2H),2.60(m,2H),2.17(m,1H),2.09(m,1H),1.87(m,2H)and1.32-1.26(m,1H)。
Example 6
Preparation of 3- (5- (3, 5-difluorobenzyl) -1,3, 4-oxadiazol-2-yl) -3-hydroxy-1- (1H-indol-5-yl) pyrrolidin-2-one (compound 6).
Figure BDA0002198418950000241
The reaction formula of example 6 is as follows:
Figure BDA0002198418950000242
step 1: thionyl chloride (1.03g, 8.72mmol, 3.0 equiv.) was added dropwise to a solution of compound 6-1(500mg, 2.9mmol, 1.0 equiv.) in methanol (10 mL). The mixture was heated and stirred at reflux for 4 hours. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was then quenched with sodium thiosulfate, water was added and extracted with ethyl acetate (2 × 10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate, 90:10) to give compound 6-2(540mg, 100%). TLC conditions: ethyl acetate 5:1, UV254 nm. R of Compound 6-1f0.3, R of Compound 6-2f=0.6。
And step 3: hydrazine hydrate (80mg, 1.61mmol, 3.0 equiv.) is added to a solution of compound 6-2(100mg, 0.54mmol, 1.0 equiv.) in methanol (10 mL). The mixture was heated and stirred at reflux for 2 hours. TLC analysis showed complete conversion of the reaction to the desired product. The reaction mixture was diluted with water, extracted with ethyl acetate (2X 20mL) and the combined organic phases concentrated under reduced pressure. The resulting crude product was purified by preparative TLC (petroleum ether: ethyl acetate, 3:1) to give compound 6-3(80mg, 80%). TLC conditions: ethyl acetate 3:1, UV254 nm. R of Compound 6-2f0.6, R of Compound 6-3f=0.3。
And 4, step 4: a mixture of compound 6-3(50mg, 0.27mmol, 1.0 equiv.), compound 6-4(103mg, 0.27mmol, 1.0 equiv.) and phosphorus oxychloride (3mL) was heated to 60 ℃ and stirred for 2 hours.TLC analysis showed complete conversion of the reaction to the desired product. The reaction was quenched with ice water, extracted with ethyl acetate (2 × 3mL), filtered and concentrated under reduced pressure. The resulting crude product was purified by preparative TLC (dichloromethane: methanol, 10:1) to give compound 6-5(40mg, 28%). TLC conditions: dichloromethane methanol 10:1, UV254 nm. R of Compound 6-3f0.4, R of Compound 6-5f=0.6。
And 5: cerium (III) chloride heptahydrate (84mg, 0.224mmol, 0.4 equiv.) was added to a solution of compound 6-5(300mg, 0.56mmol, 1.0 equiv.) in isopropanol (5 mL). The mixture was heated to 70 ℃ under an oxygen atmosphere and stirred overnight. TLC analysis showed complete conversion of the reaction to the desired product. The mixture was diluted with water (10mL), extracted with ethyl acetate (2X 10mL), and the organic layer was dried over sodium sulfate and concentrated under reduced pressure. The resulting crude product was purified by silica gel chromatography (dichloromethane: methanol, 96:4) to give compound 6-6(150mg, 68%). TLC conditions: dichloromethane methanol 10:1, UV254 nm. R of Compounds 6 to 5f0.5, R of Compound 6-6f=0.4。
And 5: a solution of sodium hydroxide (218mg, 5.45mmol, 20.0 equiv.) in water (6mL) was added to a solution of compound 6-6(150mg, 0.27mmol, 1.0 equiv.) in methanol (6 mL). The mixture was heated to 105 ℃ and stirred at reflux for 4 hours. TLC analysis showed complete conversion of the reaction to the desired product. The reaction solution was cooled to room temperature and extracted with ethyl acetate (2X 5 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The resulting crude product was purified by preparative TLC (dichloromethane: methanol, 10:1) to give compound 6(24mg, 22%) as a white solid. TLC conditions: dichloromethane methanol 10:1, UV254 nm. R of Compound 6-6f0.4, R of compound 6f=0.3。LCMS:[M+1]:411.1H NMR(d6-DMSO,400MHz):δ11.12(s,1H),7.67(s,1H),7.34(m,3H),7.16(m,1H),7.06(m,3H),6.41-6.40(d,J=2.0Hz,4H),4.36(s,2H),3.90(m,1H),3.80(m,1H),2.77(m,1H)and 2.38(m,1H)。
Example 7
Enzymatic Activity assay for MetAP-2
MetAP-2 activity was determined by an enzyme-coupled assay involving the use of the tripeptide Met-Ala-Ser (MAS) as substrate and recombinant human MetAP-2(His-Tev-MetAP-2, prepared internally). First, the released methionine is converted into Metox by L-Amino Acid Oxidase (AAO), and hydrogen peroxide is released. In the second step, the leukocyte dye dianisidine is oxidized into dianisidine ox by the catalysis of horseradish peroxidase under the combined action of hydrogen peroxide. As dianisidine ox is formed, an increase in absorbance at 450nm results, and thus MetAP-2 activity can be determined by measuring absorbance in the kinetic measurement mode. The release of one molecule of methionine corresponds to the production of one molecule of dianisidine ox. The MetAP-2 enzyme activity corresponds directly to the increase in absorbance per pass.
In examples 1,2 and 3, the IC50 for MetAP-2 was 1nM to 10. mu.M. In examples 4, 5 and 6, MetAP-2 was present at 10-100. mu.M.
Pharmaceutical preparation
(A) Injection preparation: 100g of the active ingredient according to the invention and 5g of disodium hydrogen phosphate are dissolved in 3L of double distilled water, adjusted to pH6.5 with 2N hydrochloric acid, sterile-filtered, transferred to injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contained 5mg of active ingredient.
(B) Suppository: 20g of the active ingredient according to the invention are mixed with 100g of soya lecithin and 1400g of cocoa butter and melted, poured into moulds and allowed to cool. Each suppository contains 20mg of active ingredient.
(C) Solution: a solution was prepared by dissolving 1g of the active ingredient of the present invention, 9.38g of sodium dihydrogen phosphate dihydrate, 28.48g of disodium hydrogen phosphate dodecahydrate, and 0.1g of benzalkonium chloride in 940mL of double distilled water in 940mL of double steam. The pH was adjusted to 6.8 and the solution was made up to 1 liter and sterilized by radiation. The solution can be used as eye drops.
(D) Ointment: 500mg of active ingredient according to the invention are mixed under sterile conditions with 99.5g of vaseline.
(E) And (3) tablet preparation: 1kg of the active ingredient of the invention, 4kg of lactose, 1.2kg of potato starch, 0.2kg of talc and 0.1kg of magnesium stearate are mixed and compressed in a conventional manner to give tablets. Such tablets contain 10 mg of active ingredient per tablet.
(F) Coating tablets: tablets were compressed analogously to example E and subsequently coated in a conventional manner with sucrose, potato starch, talc, tragacanth and dye.
(G) And (3) capsule preparation: 2kg of active ingredient of the invention are introduced into hard gelatin capsules in a conventional manner so that each capsule contains 20mg of active ingredient.
(H) Ampoule (2): a solution of 1kg of the active ingredient according to the invention in 60 l of double distilled water is sterile-filtered, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.
(I) Inhalation and atomization: 14g of active ingredient according to the invention are dissolved in 10 l of isotonic NaCl solution and the solution is transferred to a commercially available spray container with a pump mechanism. The solution may be sprayed into the oral or nasal cavity. One injection (about 0.1ml) corresponds to a dose of about 0.14 mg.
While a number of embodiments of the present invention are described herein, it will be apparent that the basic examples described above may be varied to provide further embodiments which utilize the compounds and methods of the present invention. It is, therefore, to be understood that the scope of the invention is defined by the appended claims rather than by the specific embodiments which have been presented by way of example.

Claims (15)

1. A compound represented by the following formula I:
Figure FDA0002198418940000011
wherein X is selected from C or N,
a is selected from the following groups:
Figure FDA0002198418940000012
b is selected from the following groups:
Figure FDA0002198418940000013
n is 0,1, 2,3 or 4;
R1and R2Each independently selected from:
halogen and-CN;
or a pharmaceutically acceptable salt thereof.
2. A compound represented by the following formula II:
Figure FDA0002198418940000014
wherein X is selected from C or N,
a is selected from the following groups:
Figure FDA0002198418940000021
b is selected from the following groups:
Figure FDA0002198418940000022
n is 0,1, 2,3 or 4;
R1and R2Each independently selected from:
halogen and-CN;
or a pharmaceutically acceptable salt thereof.
3. The compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000023
4. the compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000031
5. the compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000032
6. the compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000033
7. the compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000034
8. the compound of any one of claims 1 to 2, or a pharmaceutically acceptable salt thereof, wherein the compound is:
Figure FDA0002198418940000041
9. a method for treating or preventing cancer such as liver cancer, cholangiocarcinoma and malignant mesothelioma, pancreatic cancer, head and neck cancer, and hemangioma, comprising: administering an effective amount of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof.
10. A method of treating obesity in a mammal in need of such treatment, comprising: administering to the mammal an effective amount of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof.
11. A method of treating type II diabetes in a mammal in need of such treatment, comprising: administering to the mammal an effective amount of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof.
12. A compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, for use in therapy.
13. Use of a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament.
14. A pharmaceutical composition, comprising: a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof; and at least one pharmaceutically acceptable carrier, diluent or excipient.
15. A pharmaceutical composition, comprising: a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof; at least one pharmaceutically acceptable carrier, diluent and excipient; and at least one other pharmaceutically active agent.
CN201910856406.2A 2019-09-11 2019-09-11 Pyrrolidone derivatives Active CN112480100B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910856406.2A CN112480100B (en) 2019-09-11 2019-09-11 Pyrrolidone derivatives

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910856406.2A CN112480100B (en) 2019-09-11 2019-09-11 Pyrrolidone derivatives

Publications (2)

Publication Number Publication Date
CN112480100A true CN112480100A (en) 2021-03-12
CN112480100B CN112480100B (en) 2022-10-14

Family

ID=74920542

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910856406.2A Active CN112480100B (en) 2019-09-11 2019-09-11 Pyrrolidone derivatives

Country Status (1)

Country Link
CN (1) CN112480100B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100216815A1 (en) * 2007-10-04 2010-08-26 Duffy Joseph L N-substituted oxindoline derivatives as calcium channel blockers
CN102803252A (en) * 2009-06-11 2012-11-28 鲁汶天主教大学研究开发部 Indole amide derivatives and related compounds for use in the treatment of neurodegenerative diseases
CN103153951A (en) * 2010-10-13 2013-06-12 默克专利股份公司 Pyrrolidinones as MetAP-2 inhibitors
CN104245675A (en) * 2012-04-04 2014-12-24 默克专利股份公司 Cyclic amides as MetAP-2 inhibitors
CN106660989A (en) * 2014-08-04 2017-05-10 默克专利股份公司 Pyrrolidinone derivatives as metap-2 inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100216815A1 (en) * 2007-10-04 2010-08-26 Duffy Joseph L N-substituted oxindoline derivatives as calcium channel blockers
CN102803252A (en) * 2009-06-11 2012-11-28 鲁汶天主教大学研究开发部 Indole amide derivatives and related compounds for use in the treatment of neurodegenerative diseases
CN103153951A (en) * 2010-10-13 2013-06-12 默克专利股份公司 Pyrrolidinones as MetAP-2 inhibitors
CN104245675A (en) * 2012-04-04 2014-12-24 默克专利股份公司 Cyclic amides as MetAP-2 inhibitors
CN106660989A (en) * 2014-08-04 2017-05-10 默克专利股份公司 Pyrrolidinone derivatives as metap-2 inhibitors

Also Published As

Publication number Publication date
CN112480100B (en) 2022-10-14

Similar Documents

Publication Publication Date Title
WO2021129820A1 (en) Spiro ring-containing quinazoline compound
EP3184521B1 (en) Indazole compounds as fgfr kinase inhibitors, preparation and use thereof
WO2021129824A1 (en) New-type k-ras g12c inhibitor
WO2021190467A1 (en) Spiro ring-containing quinazoline compound
WO2021027911A1 (en) Novel spirocyclic k-ras g12c inhibitor
JP2024514703A (en) PARP inhibitor with piperazine structure, its preparation method and its pharmaceutical use
CN114846005A (en) SHP2 inhibitor and application thereof
CN107445896B (en) Phenyl hydroxamic acid compound with anti-tumor activity and application thereof
CN111662294A (en) Compound with activity of degrading Btk
CA3029086C (en) Chiral heterocyclic compound with hedgehog pathway antagonist activity, method and use thereof
CN110872285A (en) Heterocyclic compounds as receptor interacting protein 1(RIP1) kinase inhibitors
WO2023280182A1 (en) Compound serving as kat6 inhibitor
CN116322697A (en) Quinazoline compound and pharmaceutical composition thereof
KR19980703022A (en) 3- (bis-substituted phenylmethylene) oxindole derivatives
CN112480100B (en) Pyrrolidone derivatives
WO2023274246A1 (en) Amide compound and use thereof
WO2023097189A1 (en) Ampk activators
CN115322158A (en) As KRAS G12C Substituted quinazoline compounds of protein inhibitor
CN109651377B (en) Compound for treating cancer and application thereof
CN109748914B (en) Pyridopyrimidine compound and application thereof
CA3147471A1 (en) Inhibitors of human atgl
CN112538086B (en) Split compound of heterocyclic-triazole ring and indole carboxylic acid and salt thereof, preparation method and medical application thereof
WO2020177752A1 (en) 1,2,4-triazole compound, preparation method therefor and pharmaceutical use thereof
AU2017244139A1 (en) Compounds for the inhibition of cyclophilins and uses thereof
CN108558869B (en) For treating the compound and its preparation of liver cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant