WO2019118612A1 - Targeting compounds - Google Patents

Targeting compounds Download PDF

Info

Publication number
WO2019118612A1
WO2019118612A1 PCT/US2018/065225 US2018065225W WO2019118612A1 WO 2019118612 A1 WO2019118612 A1 WO 2019118612A1 US 2018065225 W US2018065225 W US 2018065225W WO 2019118612 A1 WO2019118612 A1 WO 2019118612A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
oxy
octan
oxaspiro
methylbut
Prior art date
Application number
PCT/US2018/065225
Other languages
French (fr)
Inventor
Robert Zahler
James E. Vath
Original Assignee
Zafgen, Inc.
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 Zafgen, Inc. filed Critical Zafgen, Inc.
Publication of WO2019118612A1 publication Critical patent/WO2019118612A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • 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

Definitions

  • Membrane transporters are found throughout the body and play an important role in drug disposition. The tissue distribution of a drug can have a significant impact on both the agents’ safety and efficacy. Of the over 400 human transporters about 20 of them are important for drug transport (Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010; 9(3):2l5-36).
  • SLC transporters are expressed in many tissues such as brain, liver, small intestine, kidney and are involved in the uptake of drugs and nutrients into cells. SLC transporters are categorized into three categories (i) the first type transport by facilitated diffusion via a concentration gradient, (ii) the second type utilizes Na + or H + cotransport and (iii) the third type involves exchanging substrates to aid transport.
  • the main SLC transporters include organic anion transporting polypeptides (OATP), organic anion transporters (OAT) and organic cation transporters (OCT).
  • the OATPs represent a superfamily of membrane transport proteins that mediate the sodium-independent transport of a diverse range of amphiphilic organic
  • OATP1B1 SCOlBl
  • 1B3 SLC01B3
  • 2B1 SLC02B1
  • SLC02B1 SLC02B1
  • OATP1B1 is expressed uniformly throughout the lobules.
  • the OATP1B1 transporters exhibit broad substrate selectivity that includes anionic (e.g. statins such as pravastatin, pitavastatin, and rosuvastatin), zwitterionic (e.g. rifampicin) and neutral lipophilic (e.g. paclitaxel) drugs.
  • anionic e.g. statins such as pravastatin, pitavastatin, and rosuvastatin
  • zwitterionic e.g. rifampicin
  • neutral lipophilic e.g. paclitaxel
  • OATP1B3 is in the sinusoidal membranes of hepatocytes located around the central vein. OATP1B3 has considerable substrate overlap with OATP1B1 but the gastrointestinal peptide Cholecystokinin is reported to be exclusively transported by OATP1B3.
  • Other specific OATP1B3 substrates include docetaxel, digoxin, paclitaxel and the toxin amanitin (Letschert, K., et al., Molecular characterization and inhibition of amanitin uptake into human hepatocytes. Toxicol Sci, 2006. 91(1): p. 140-9). CCK-8 is a specific inhibitor of OATP1B3.
  • Cyclosporine and CI-1034 are inhibitors of the three major OATP isoforms (Rizwan, AN. and Burckhardt, G, Organic anion transporters of the SLC22 family: biopharmaceutical, physiological, and pathological roles. Pharm Res, 2007. 24(3): p. 450-70; Sahi, T, et al., Metabolism and transporter-mediated drug-drug interactions of the endothelin-A receptor antagonist CI-1034. Chem Biol Interact, 2006. 159(2): p. 156-68).
  • human OATP1B1 and OAT1B3 are liver specific they may be used for liver targeting to enhance liver concentration and minimize drug exposure to the peripheral tissues (e.g. brain) to reduce toxicity and/or side effects.
  • Some medicinal chemistry design principles of orally active drugs for liver targeting through OATP transporters have been described (Tu M, Mathiowetz AM, Pfefferkom JA, Cameron KO, Dow RL, Litchfield J, Di L, Feng B, Liras S. Medicinal chemistry design principles for liver targeting through OATP transporters. Curr Top Med Chem. 2013; l3(7):857-66).
  • These principles include, e.g., efficient hepatocyte uptake via OATP1B1 and/or OATP1B3 transporters which generally require an acidic function in the molecule, low passive permeability to minimize distribution into peripheral tissues that lack liver-specific OATP transporters, and sufficient bioavailability to support oral
  • OATP1B1 and 1B3 have been primary human liver-specific transporter targets
  • OATP2B1 and Sodium Tauracholate Transporting Polypeptide (NCTP1) represent additional human liver uptake transporters which could contribute to a liver targeted approach.
  • OATP2B1 In addition to the sinusoidal membrane of the hepatocyte, OATP2B1 is widely expressed in tissues including apical membrane of enterocytes, the basal membrane of the syncytiotrophoblasts, the luminal membrane of brain capillary endothelial cells, small arteries and veins of the heart, and the lung. OATP2B1 has broad substrate specificity transporting mostly anionic organic endo- and xenobiotics. It exhibits some pH dependency in that it seems to transport some substrates more efficiently at lower pH.
  • OATP1B1 and 1B3 Its substrate specificity appears somewhat more restricted than OATP1B1 and 1B3, however it transports fexofenadine, statins, glibenclamide, glyburide, estrone-3 -sulfate (E3S), dehydroepiandrosterone-3-sulfate (DHEAS), prostaglandin E2, and taurocholate.
  • OATP2B1 in the liver mediates the hepatic uptake of many xenobiotics, including bromosulphophthalein (BSP), benzylpenicillin, fexofenadine, glibenclamide, pravastatin, atorvastatin, rosuvastatin, and fluvastatin.
  • BSP bromosulphophthalein
  • OATP2B1 inhibitors include many organic anions, mono- and dicarboxylic acids, steroid hormones and their derivatives, drugs, such as rifamycin SV, pravastatin cyclosporine A and gemfibrozil as well as constituents of citrus juices and herbal extracts.
  • OATP family members are poorly conserved evolutionarily and orthologues for human OATPs may not exist in rodents. The significant species differences for the
  • OATP/Oatp transporters make it challenging to translate pharmacokinetic or pharmacodynamic data from preclinical species to humans.
  • NTCP Sodium Taurocholate Co-Transporting Polypeptide
  • SLOC10A1 Sodium Taurocholate Co-Transporting Polypeptide
  • NTCP is primarily responsible for the uptake of bile acids from the sinusoids.
  • NTCP is one of the key transporters in the enterohepatic circulation of bile acids, which also include the canalicular efflux transporter BSEP, and ASBT and OSTa/b in the gastrointestinal tract (Doring, B., et ah, The SLC10 carrier family: transport functions and molecular structure. Curr Top Membr, 2012. 70: p. 105-68).
  • Other substrates include sulfated steroids (estrone-3 -sulfate, DHEAS) and thyroid hormones.
  • NTCP interacts with a relatively narrow panel of drugs and other xenobiotics.
  • a computational model of a substrate pharmacophore has been developed (Dong Z. et al. A Substrate Pharmacophore for the Human Sodium Taurocholate Co-transporting Polypeptide. Int J Pharm. 2015 Jan 15; 478(1): 88-95).
  • NTCP is important in the hepatic uptake of statins, particularly rosuvastatin, and is therefore a risk factor for transporter-mediated DDI for this drug.
  • Chronic liver diseases such as fatty liver disease and liver fibrosis can be risk factors that may dispose progression to hepatocellular carcinoma, and represent a common and difficult clinical challenge of worldwide importance.
  • liver fibrosis is the excessive accumulation of extracellular matrix proteins including collagen that occurs in most types of chronic liver diseases. Advanced liver fibrosis results in cirrhosis, liver failure, and portal hypertension and often requires liver transplantation.
  • Non-alcoholic steatohepatitis is a severe form of fatty liver disease that affects 2 to 5 percent of Americans.
  • Many people with NASH have risk factors also associated with HCC, e.g., obesity, diabetes and/or elevated blood cholesterol levels.
  • Alpha-l antitrypsin deficiency is an inherited disorder associated with an increased risk of liver and lung disease.
  • the disorder is associated with the retention of the liver-produced protein alpha-l antitrypsin in the liver and low levels of alpha-l antitrypsin in the serum.
  • Clinical features of alpha-l antitrypsin deficiency include chronic liver inflammation and liver fibrosis, which can progress in advanced forms to chronic hepatitis, cirrhosis and hepatocellular carcinoma.
  • Chronic kidney disease may increase the risk for recurrence and progression of other cancers such as non-muscle invasive bladder cancer, and can also limit the ability to receive and withstand cancer therapies.
  • Other kidney diseases such as lupus nephritis can lead to kidney failure and patients suffering from lupus nephritis are at a higher risk for cancers such as B-cell lymphoma, and for heart and blood vessel problems.
  • Methionine aminopeptidase 2 encodes a protein that functions at least in part by enzymatically removing the amino terminal methionine residue from certain newly translated proteins such as glyceraldehyde-3 -phosphate dehydrogenase. Increased expression of the MetAP2 gene has been historically associated with various forms of cancer. Molecules inhibiting the enzymatic activity of MetAP2 have been identified and have been explored for their utility in the treatment of various tumor types.
  • This disclosure is directed in part to, for example, carboxylic acid (and carboxylate salt) sidechains appended to the 6-position alcohol of fumagillol that are MetAP2 modulators selectively targeting the liver, intestine and/or kidney, their use as medicinal agents, processes for their preparation, and pharmaceutical compositions containing them as an active ingredient both alone or in combination with other agents, as well as provides for their use as
  • compositions comprising at least one disclosed compound and a pharmaceutically acceptable carrier.
  • L 1 , L 2 and T are as defined herein.
  • the liver disorder may be one or more of, for example, non-alcoholic steatohepatitis, alcoholic steatohepatitis, carcinoma (e.g., hepatocellular carcinoma), liver cirrhosis, and hepatitis B.
  • the kidney disorder may be one or more of chronic kidney disease, glomerular disease such as iGA nephropathy, lupus nephritis, polycystic kidney disease.
  • hepatic preneoplastic lesions in patient in need thereof, comprising administering an effective amount of a compound disclosed herein.
  • the patient may be at risk for but not yet suffering from NASH.
  • the patient may have at least one of, for example: minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation, and/or balloon degeneration.
  • the patient may suffer from type 2 diabetes and/or is obese.
  • the patient may suffer from alpha- 1 antitrypsin deficiency.
  • provided herein are methods of treating hepatocellular carcinoma in a patient in need thereof, comprising administering to the patient an effective amount of a disclosed compound.
  • Contemplated methods of treating may also include administering a disclosed compound in combination with one or more other agents (and/or in combination with weight loss surgery and/or weight loss therapy).
  • contemplated methods herein can include administering one or more additional agents, e.g. as disclosed herein, for example, an anti diabetic medication (metformin), a modulator of glucagon -like peptide -1 receptor such as exenatide and liraglutide, obeticholic acid, a PPAR agonist, and/or a thiazolidinedione such as pioglitazone.
  • additional agents e.g. as disclosed herein, for example, an anti diabetic medication (metformin), a modulator of glucagon -like peptide -1 receptor such as exenatide and liraglutide, obeticholic acid, a PPAR agonist, and/or a thiazolidinedione such as pioglitazone.
  • Treating includes any effect, e.g., lessening, reducing, modulating, or eliminating, that results in the improvement of the condition, disease, disorder and the like.
  • alkenyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond.
  • alkenyl groups include, but are not limited to, a straight or branched group of 2-6 or 3-4 carbon atoms, referred to herein as C 2-6 alkenyl, and C 3- 4alkenyl, respectively.
  • alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, etc.
  • alkoxy refers to a straight or branched alkyl group attached to oxygen (alkyl-O-).
  • exemplary alkoxy groups include, but are not limited to, alkoxy groups of 1-6 or 2-6 carbon atoms, referred to herein as Ci -6 alkoxy, and C 2-6 alkoxy, respectively.
  • Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, isopropoxy, etc.
  • alkoxyalkyl refers to a straight or branched alkyl group attached to oxygen, attached to a second straight or branched alkyl group (alkyl -O-alkyl-).
  • alkoxyalkyl groups include, but are not limited to, alkoxyalkyl groups in which each of the alkyl groups independently contains 1-6 carbon atoms, referred to herein as Ci. 6 alkoxy-Ci- 6 alkyl.
  • Exemplary alkoxyalkyl groups include, but are not limited to
  • alkyoxycarbonyl refers to a straight or branched alkyl group attached to oxygen, attached to a carbonyl group (alkyl -O-C(O)-).
  • alkoxycarbonyl groups include, but are not limited to, alkoxycarbonyl groups of 1-6 carbon atoms, referred to herein as Ci -6 alkoxycarbonyl.
  • Exemplary alkoxycarbonyl groups include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, t-butoxycarbonyl, etc.
  • alkenyloxy refers to a straight or branched alkenyl group attached to oxygen (alkenyl-O-).
  • exemplary alkenyloxy groups include, but are not limited to, groups with an alkenyl group of 3-6 carbon atoms, referred to herein as C3 -6 alkenyloxy.
  • Exemplary“alkenyloxy” groups include, but are not limited to allyloxy, butenyloxy, etc.
  • alkynyloxy refers to a straight or branched alkynyl group attached to oxygen (alkynyl-O).
  • exemplary alkynyloxy groups include, but are not limited to, groups with an alkynyl group of 3-6 carbon atoms, referred to herein as C 3-6 alkynyloxy.
  • Exemplary alkynyloxy groups include, but are not limited to, propynyloxy, butynyloxy, etc.
  • alkyl refers to a saturated straight or branched hydrocarbon.
  • exemplary alkyl groups include, but are not limited to, straight or branched hydrocarbons of 1-6, 1-4, or 1-3 carbon atoms, referred to herein as Ci -6 alkyl, Ci -4 alkyl, and Ci -3 alkyl, respectively.
  • Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l-butyl, 3 -methyl -2 -butyl, 2-methyl- 1 -pentyl, 3-methyl-l-pentyl, 4- methyl-l -pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2 -pentyl, 2,2-dimethyl- 1- butyl, 3, 3 -dimethyl -1 -butyl, 2-ethyl- 1 -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, etc.
  • alkylcarbonyl refers to a straight or branched alkyl group attached to a carbonyl group (alkyl-C(O)-).
  • exemplary alkylcarbonyl groups include, but are not limited to, alkylcarbonyl groups of 1-6 atoms, referred to herein as Ci -6 alkylcarbonyl groups.
  • exemplary alkylcarbonyl groups include, but are not limited to, acetyl, propanoyl, isopropanoyl, butanoyl, etc.
  • alkynyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond.
  • exemplary alkynyl groups include, but are not limited to, straight or branched groups of 2-6, or 3-6 carbon atoms, referred to herein as C 2-6 alkynyl, and C 3-6 alkynyl, respectively.
  • exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, etc.
  • carbonyl refers to the radical -C(O)-.
  • cycloalkoxy refers to a cycloalkyl group attached to oxygen (cycloalkyl-O-).
  • exemplary cycloalkoxy groups include, but are not limited to, cycloalkoxy groups of 3-6 carbon atoms, referred to herein as C 3-6 cycloalkoxy groups.
  • Exemplary cycloalkoxy groups include, but are not limited to, cyclopropoxy, cyclobutoxy, cyclohexyl oxy, etc
  • cycloalkyl or a“carbocyclic group” as used herein refers to a saturated or partially unsaturated hydrocarbon group of, for example, 3-6, or 4-6 carbons, referred to herein as C3-6cycloalkyl or C4 -6 cycloalkyl, respectively.
  • exemplary cycloalkyl groups include, but are not limited to, cyclohexyl, cyclopentyl, cyclopentenyl, cyclobutyl or cyclopropyl.
  • halo or“halogen” as used herein refer to F, Cl, Br, or I.
  • heteroaryl or“heteroaromatic group” as used herein refers to a monocyclic aromatic 5-6 membered ring system containing one or more heteroatoms, for example one to three heteroatoms, such as nitrogen, oxygen, and sulfur. Where possible, said heteroaryl ring may be linked to the adjacent radical though carbon or nitrogen. Examples of heteroaryl rings include but are not limited to furan, thiophene, pyrrole, thiazole, oxazole, isothiazole, isoxazole, imidazole, pyrazole, triazole, pyridine or pyrimidine etc.
  • heterocyclyl or“heterocyclic group” are art-recognized and refer to saturated or partially unsaturated, 4-10 membered ring structures, including spirocyclic, bridged or fused rings, and whose ring structures include one to three heteroatoms, such as nitrogen, oxygen, and sulfur. Where possible, heterocyclyl rings may be linked to the adjacent radical through carbon or nitrogen. Examples of heterocyclyl groups include, but are not limited to, pyrrolidine, piperidine, morpholine, thiomorpholine, piperazine, oxetane, azetidine, tetrahydrofuran or dihydrofuran etc.
  • heterocyclyloxy refers to a heterocyclyl group attached to oxygen (heterocyclyl -0-).
  • heteroaryloxy refers to a heteroaryl group attached to oxygen (heteroaryl-O-).
  • “Pharmaceutically or pharmacologically acceptable” include molecular entities and compositions that should not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
  • preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologies standards.
  • pharmaceutically acceptable carrier or“pharmaceutically acceptable excipient” as used herein refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical
  • compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • composition refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
  • Individual,”“patient,” or“subject” are used interchangeably and include any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the compounds of the disclosure can be administered to a mammal, such as a human, but can also be administered to other mammals such as an animal in need of veterinary treatment, e.g., domestic animals (e.g, dogs, cats, and the like), farm animals (e.g, cows, sheep, pigs, horses, and the like) and laboratory animals (e.g, rats, mice, guinea pigs, and the like).
  • “Modulation” includes antagonism (e.g, inhibition), agonism, partial antagonism and/or partial agonism.
  • the term“therapeutically effective amount” means the amount of the subject compound that will elicit the biological or medical response of a tissue, system or animal, (e.g. mammal or human) that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • the compounds of the disclosure are administered in therapeutically effective amounts to treat a disease.
  • a therapeutically effective amount of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect, such as an amount that results in substantially mitigating a liver or kidney disease or disorder.
  • salts refers to salts of acidic or basic groups that may be present in compounds used in the compositions.
  • Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • the acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including, but not limited to, malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, / oluenesulfonate and pamoate (i.e., 1 , 1 '-
  • Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations.
  • Examples of such salts include alkali metal or alkaline earth metal salts, particularly calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
  • Compounds included in the present compositions that include a basic or acidic moiety may also form pharmaceutically acceptable salts with various amino acids.
  • the compounds of the disclosure may contain both acidic and basic groups; for example, one amino and one carboxylic acid group. In such a case, the compound can exist as an acid addition salt, a zwitterion, or a base salt.
  • the compounds of the disclosure may contain one or more chiral centers and, therefore, exist as stereoisomers.
  • stereoisomers when used herein consist of all enantiomers or diastereomers. These compounds may be designated by the symbols“(+),”“(- ),”“R” or“S,” depending on the configuration of substituents around the stereogenic carbon atom, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
  • the present invention encompasses various stereoisomers of these compounds and mixtures thereof. Mixtures of enantiomers or diastereomers may be designated“( ⁇ )” in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
  • the compounds of the disclosure may contain one or more double bonds and, therefore, exist as geometric isomers resulting from the arrangement of substituents around a carbon-carbon double bond.
  • the symbol . denotes a bond that may be a single, double or triple bond as described herein.
  • Substituents around a carbon-carbon double bond are designated as being in the“Z’ or“£” configuration wherein the terms“Z’ and“£” are used in accordance with IUPAC standards.
  • structures depicting double bonds encompass both the“ E” and“Z” isomers.
  • Substituents around a carbon-carbon double bond alternatively can be referred to as“cis” or“trans,” where“cis” represents substituents on the same side of the double bond and“trans” represents substituents on opposite sides of the double bond.
  • Compounds of the disclosure may contain a carbocyclic or heterocyclic ring and therefore, exist as geometric isomers resulting from the arrangement of substituents around the ring.
  • the arrangement of substituents around a carbocyclic or heterocyclic ring are designated as being in the“Z” or“E” configuration wherein the terms“Z” and“E” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting carbocyclic or heterocyclic rings encompass both“Z” and“E” isomers.
  • Substituents around a carbocyclic or heterocyclic rings may also be referred to as“cis” or“trans”, where the term“cis” represents substituents on the same side of the plane of the ring and the term“trans” represents substituents on opposite sides of the plane of the ring.
  • Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated“cis/trans.”
  • Individual enantiomers and diasteriomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art.
  • Stereoselective syntheses a chemical or enzymatic reaction in which a single reactant forms an unequal mixture of stereoisomers during the creation of a new stereocenter or during the transformation of a pre-existing one, are well known in the art.
  • Stereoselective syntheses encompass both enantio- and diastereoselective transformations, and may involve the use of chiral auxiliaries. For examples, see Carreira and Kvaemo, Classics in Stereoselective Synthesis , Wiley-VCH: Weinheim, 2009
  • the compounds disclosed herein can exist in solvated as well as unsolvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
  • the compound is amorphous.
  • the compound is a single polymorph.
  • the compound is a mixture of polymorphs.
  • the compound is in a crystalline form.
  • the invention also embraces isotopically labeled compounds of the invention which are identical to those recited herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 3 ⁇ 4 3 ⁇ 4 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • a compound of the invention may have one or more H atom replaced with deuterium.
  • Certain isotopically-labeled disclosed compounds e.g ., those labeled with 3 H and 14 C are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-l4 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Isotopically labeled compounds of the invention can generally be prepared by following procedures analogous to those disclosed in the examples herein by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • prodrugs of disclosed compounds are also contemplated herein.
  • prodrug refers to compounds that are transformed in vivo to yield a disclosed compound or a
  • the transformation may occur by various mechanisms (such as by esterase, amidase, phosphatase, oxidative and or reductive metabolism) in various locations (such as in the intestinal lumen or upon transit of the intestine, blood or liver).
  • Prodrugs are well known in the art (for example, see Rautio,
  • a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as (Ci -8 )alkyl, (C 2-i2 )alkylcarbonyloxymethyl, 1 -(alkyl carbonyl oxy)ethyl having from 4 to 9 carbon atoms, 1- methyl-l-(alkylcarbonyloxy)-ethyl having from 5 to 10 carbon atoms,
  • alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms
  • l-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms
  • 1 -methyl- l-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms
  • N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms
  • a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (Ci -6 )alkylcarbonyloxymethyl, l-((Ci -6 )alkylcarbonyloxy)ethyl, 1 -methyl- l-((Ci. 6 )alkylcarbonyloxy)ethyl (C l-6 )alkoxycarbonyloxym ethyl, N-(Ci.
  • a prodrug can be formed, for example, by creation of an amide or carbamate, an N-alkylcarbonyloxyalkyl derivative, an (oxodioxolenyl)methyl derivative, an N-Mannich base, imine or enamine.
  • a secondary amine can be metabolically cleaved to generate a bioactive primary amine, or a tertiary amine can metabolically cleaved to generate a bioactive primary or secondary amine.
  • the present disclosure provides a pharmaceutically acceptable base salt of a compound represented by:
  • L 1 is -NR X R 2 ;
  • R 1 is H or Ci-3alkyl
  • R 2 is selected from the group consisting of: a 4-9 membered monocyclic heterocyclic ring, heteroaryl, -Ci -2 alkylene-heteroaryl, phenyl, -Ci -2 alkylene- phenyl and -Ci -6 alkylene; wherein -Ci -6 alkylene may be straight or branched and may optionally be substituted with one or more substituents each independently selected from R p ; or
  • R 1 and R 2 taken together with the nitrogen to which they are attached form a 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring; wherein the 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring is optionally substituted with one or more substituents each independently selected from R h ;
  • R a and R b are independently selected, for each occurrence, from the group consisting of hydrogen and C 1.3 alkyl.
  • L 1 is selected from the group consisting of:
  • p is 1 or 2
  • q is 1 or 2
  • r is 0, 1 or 2
  • Z is CH 2 or O
  • Y is N or CH.
  • L 1 is selected from the group consisting of:
  • L 1 is selected from the group consisting of: wherein independently for each occurrence p is 1 or 2, and q is 1 or 2. [0062] In some embodiments, L 1 is selected from the group consisting of:
  • L 2 is selected from the group consisting of: -CH 2 -, -CH 2 -CH 2 -, -0-CH 2 -, -0-CH 2 -CH 2 -, -NR a -CH 2 -, -CH 2 -C(CH 3 )(CH 3 )-, -0-CH 2 -C(CH 3 )(CH 3 )-,-0-CH 2 -C(CH 3 )(CH 3 )-,-0-0
  • the base salt is an alkali metal salt.
  • the alkali metal is selected from the group consisting of, e.g., sodium, potassium, and lithium.
  • T is selected from the group consisting of:
  • T may be selected from a base salt (e.g., a lithium, sodium or potassium salt) of a moiety selected from the group consisting of:
  • a base salt e.g., a lithium, sodium or potassium salt
  • compounds that may be selected from the group consisting of: sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)-3- methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide; sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetate
  • the disclosure provides a method of treating or mitigating a liver, intestine and/or kidney disorder in a patient in need thereof, comprising administering an effective amount of a disclosed compound to the patient.
  • the liver disorder is one or more of, e.g.: non-alcoholic steatohepatitis, alcoholic
  • steatohepatitis hepatocellular carcinoma, liver cirrhosis, and hepatitis B.
  • the kidney disorder may be one or more of chronic kidney disease, glomerular disease such as iGA nephropathy, lupus nephritis, polycystic kidney disease
  • Contemplated patients include not only humans, but other animals such as companion animals (e.g., dogs, cats).
  • companion animals e.g., dogs, cats.
  • Methods of treating hepatocellular carcinoma in a patient in need thereof comprising adminsetering an effective amount of a disclosed compound to the patient.
  • the patient suffers from type 2 diabetes, hepatitis B, cirrhosis, alpha-l antitrypsin deficiency, and/or is obese.
  • hepatic preneoplastic lesions in patient in need thereof, comprising administering an effective amount of a disclosed compound to the patient.
  • the patient is at risk for but not yet suffering from NASH.
  • the patient has at least one of, for example: minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation, and/or balloon degeneration.
  • the patient is diabetic (e.g. type 2 diabetes) or in pre-diabetic condition. The patient may or may not also suffer from obesity.
  • the patient has a risk of developing NASH (and/or alcoholic steatohepatitis or cirrhosis), but is not suffering from NASH and/or alcoholic steatohepatitis or cirrhosis, or in other embodiments, the patient has developed NASH, alcoholic steatohepatitis or cirrhosis, but has not suffered from hepacellular carcinoma.
  • a patient may have minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation and/or balloon degeneration.
  • Compounds may be administered in an amount insufficient to modulate or suppress angiogenesis in said patient.
  • the patient suffers from type 2 diabetes and/or is obese.
  • Contemplated patients may optionally be currently administering or have been administered amiodarone, antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
  • antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
  • a patient suscepted of suffering from NASH or one or more of NASH symptoms may be scored using a NASH scoring.
  • a NASH scored is the combined score of fibrosis (0-4), steatosis (0-3), lobular inflammation (0-3), and ballooning degeneration (0-2) as illustrated in table A (see Design and validation of a histological scoring system for nonalcoholic fatty liver disease, Kleiner et ak, Hepatology 41; 2005).
  • contemplated compounds disclosed herein may beneficially affect hepatic glucose uptake, insulin action, steatosis, inflammation and/or cell dysplasia.
  • Disclosed compounds may be administered to patients (animals and/or humans) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. It will be appreciated that the dose required for use in any particular application will vary from patient to patient, not only with the particular compound or composition selected, but also with the route of administration, the nature of the condition being treated, the age and condition of the patient, concurrent medication or special diets then being followed by the patient, and other factors which those skilled in the art will recognize, with the appropriate dosage ultimately being at the discretion of the attendant physician.
  • a compound of this invention may be administered orally, subcutaneously, topically, parenterally, by inhalation spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • administration may include subcutaneous injections, intravenous or intramuscular injections or infusion techniques.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a disclosed compound will be that amount of the compound which is the lowest dose effective to safely produce the maximal therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • a disclosed compound can be administered at about 0.01 mg/kg to about 200 mg/kg, at about 0.1 mg/kg to about 100 mg/kg, or at about 0.5 mg/kg to about 50 mg/kg.
  • the effective amount may be less than when the agent is used alone.
  • Contemplated methods may include administration of a disclosed compounds, for example, hourly, twice hourly, every three to four hours, daily, twice daily, 1, 2, 3 or 4 times a week, every three to four days, every week, or once every two weeks depending on half-life and clearance rate of the particular compound.
  • Treatment can be continued for as long or as short a period as desired.
  • the compositions may be administered on a regimen of, for example, one to four or more times per day.
  • a suitable treatment period can be, for example, at least about one week, at least about two weeks, at least about one month, at least about six months, at least about 1 year, or indefinitely.
  • a treatment period can terminate when a desired result disclosed herein, e.g., treating or mitigating a liver disorder disclosed herein, is achieved.
  • a treatment regimen can include a corrective phase, during which a dose sufficient to treat or mitigate a liver disorder disclosed herein, and can be followed by a maintenance phase, during which a e.g. a lower dose sufficient to prevent recurrence of the liver disorder is administered.
  • a suitable maintenance dose is likely to be found in the lower parts of the dose ranges provided herein, but corrective and maintenance doses can readily be established for individual subjects by those of skill in the art without undue experimentation, based on the disclosure here
  • Contemplated methods may also include administering a disclosed compound in combination with one or more other agents (and/or in combination with weight loss surgery and/or weight loss therapy).
  • other agents may be selected from an antidiabetic medication (e.g., metformin, a modulator of glucagon-like peptide -1 receptor such as exenatide, liraglutide, lixisenatide, albiglutide and dulaglutide; and/or a thiazolidinedione such as rosiglitazone, pioglitazone, troglitazone, and/or netoglitazone).
  • an antidiabetic medication e.g., metformin, a modulator of glucagon-like peptide -1 receptor such as exenatide, liraglutide, lixisenatide, albiglutide and dulaglutide
  • a thiazolidinedione such as rosiglitazone, pioglitazone, trogli
  • other agents may include a C-C chemokine receptor type 5/type 2 (CCR5/CCR2) modulator, such as but not limited to cenicriviroc.
  • the other agent may be fibroblast growth factor 21 (FGF21) modulator, such as but not limited to BMS-986036, an
  • the other agent is a peroxisome proliferation-activated receptor (PPAR) agonist, and/or may include agents such as elafibranor, clofibrate, gemfibrozil, ciprofibrate, bezafibrate, and/or fenofibrate,
  • PPAR peroxisome proliferation-activated receptor
  • the other agent is a obeticholic acid, aramchol, emricasan, silicab, GS-4997 (Gilead), GR-MD-02 (Galectin Therapeutics), Tipelukast, ARI-3037MO (Arisaph
  • a contemplated other agent is selected from an antioxidant (e.g., vitamin E, selenium or betaine), a blood pressure lowering medication (e.g., isinopril, losartan, metoprolol, amlodipine, aliskiren), and a cholesterol lowering medication (e.g., atorvastatin, rosuvastatin, or nicotinic acid).
  • a contemplated other agent is selected from amiodarone, antiviral drugs such as nucleoside analogues, aspirin, corticosteroids, methotrexate, tamoxifen, and tetracycline.
  • a patient with NASH may also have other conditions, such as diabetes, high blood pressure or high cholesterol levels.
  • contemplated herein are disclosed compounds in combination with at least one other agent that may be used to treat one or more of these conditions.
  • the second active agent can be, for example, an antidiabetic medication (e.g., metformin, and/or a thiazolidinedione such as rosiglitazone, pioglitazone, troglitazone, and/or netoglitazone), an antioxidant (e.g., vitamin E, selenium or betaine), a blood pressure lowering medication (e.g., lisinopril, losartan, metoprolol, amlodipine, aliskiren), or a cholesterol lowering medication (e.g., atorvastatin, rosuvastatin, or nicotinic acid), or another agent such as vitamin E.
  • an antidiabetic medication e.g., metformin, and/
  • contemplated methods further include administering to the patient an active agent such as one or more of: amiodarone, antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
  • an active agent such as one or more of: amiodarone, antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
  • compositions comprising compounds as disclosed herein formulated together with a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
  • These formulations include those suitable for oral, rectal, topical, buccal, parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) rectal, vaginal, or aerosol
  • compositions may be formulated as a unit dose, and/or may be formulated for oral or subcutaneous administration.
  • Exemplary pharmaceutical compositions may be used in the form of a pharmaceutical preparation, for example, in solid, semisolid or liquid form, which contains one or more of disclosed compounds, as an active ingredient, in admixture with an organic or inorganic carrier or excipient suitable for external, enteral or parenteral applications.
  • the active ingredient may be compounded, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use.
  • the active object compound is included in the pharmaceutical composition in an amount sufficient to produce the desired effect upon the process or condition of the disease.
  • the principal active ingredient may be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g, water, to form a solid preformulation
  • a pharmaceutical carrier e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g, water, to form a solid preformulation
  • compositions containing a homogeneous mixture of a disclosed compound, or a non-toxic pharmaceutically acceptable salt thereof.
  • a homogeneous mixture of a disclosed compound or a non-toxic pharmaceutically acceptable salt thereof.
  • the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • solid dosage forms for oral administration capsules, tablets, pills, dragees, powders, granules and the like
  • the subject composition is mixed with one or more
  • pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4)
  • disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents.
  • solution retarding agents such as paraffin
  • absorption accelerators such as quaternary ammonium compounds
  • wetting agents such as, for example, acetyl alcohol and glycerol monostearate
  • absorbents such as kaolin and bentonite clay
  • lubricants such a talc,
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the subject composition moistened with an inert liquid diluent.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, cyclodextrins and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate
  • Suspensions in addition to the subject composition, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing a subject composition with one or more suitable non irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the body cavity and release the active agent.
  • suitable non irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the body cavity and release the active agent.
  • Dosage forms for transdermal administration of a subject composition include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to a subject composition, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays may contain, in addition to a subject composition, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays may additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Disclosed compositions and compounds may alternatively be administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A non-aqueous (e.g., fluorocarbon propellant) suspension could be used.
  • Sonic nebulizers may be used because they minimize exposing the agent to shear, which may result in degradation of the compounds contained in the subject compositions.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of a subject composition together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular subject composition, but typically include non-ionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • compositions contemplated herein may be suitable for parenteral administration comprise a subject composition in combination with one or more
  • sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and non-aqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate and cyclodextrins.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate and cyclodextrins.
  • Proper fluidity may be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • the compounds described herein can be prepared in a number of ways based on the teachings contained herein and synthetic procedures known in the art. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated. The starting materials for the examples are either commercially available or are readily prepared by standard methods from known materials.
  • Reactions were monitored by LCMS and/or TLC on silica gel 60 HSGF254 percolated plates (0.15-0.2 mm Si02) and visualized using UV light (254 nm or 365 nm) and/or staining with phosphomolybdic acid ethanol solution (10 g in 100 mL ethanol) and subsequent heating.
  • Preparative HPLC were typically performed either by Method A: SHIMADZU LC- 8A (Column: YMC Pack ODS-A (l50*30mm, lOpm)) or Method B: LC-6AD (Column:
  • CDI l,l '-carbonyl diimidazole
  • DIPEA diisopropylethylamine
  • PE petroleum ether
  • TBTU 2-(lH -Benzoin azole- 1 -yl)- 1,1, 3 ,3 -tetram ethyl aminium tetrailuoroborate
  • Example 1 Preparation of sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)-3-methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide
  • Step 7 Preparation of sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)-3- methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide
  • Example 2 Preparation of sodium 2-(l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetate
  • Step 2 Preparation of 2-(l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetic acid
  • Step 3 Preparation of sodium 2-(l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetate
  • Example 3 Preparation of sodium l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4- carboxylate
  • Step 2 Preparation of sodium l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4- carboxylate
  • Example 4 Preparation of sodium 5-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)tetrazol-2-ide
  • Step 1 Preparation of tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate
  • Step 3 Preparation of tert-butyl 3-((2H-tetrazol-5-yl)methyl)azetidine-l-carboxylate
  • Step 5 Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl lH-imidazole-l-carboxylate
  • Step 6 Preparation of (3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((2H-tetrazol-5-yl)methyl)azetidine-l- carboxylate
  • Step 7 Preparation of Sodium 5-((l-(((( /?, S,5A,6/?)-5-methoxy-4-((2/?, /?)-2-methyl-3-
  • Example 5 Preparation of sodium 2-((l-((((( /?, S,5A,6/?)-5-methoxy-4-((2/?, /?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)acetate N
  • Step 2 Preparation of 2-((l-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetic acid
  • Step 3 Preparation of sodium 2-((l-((((3/?,7V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
  • Step 1 Preparation of 3-(l-(terf-butoxycarbonyl)azetidin-3-yl)propanoic acid
  • Step 3 Preparation of 3-(l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoic acid
  • Step 4 Preparation of sodium 3-(l-((((J ?, A,5A,6/?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoate
  • Example 7 Preparation of sodium 5-(2-(l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,.?/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)tetrazol-2-ide
  • Step 1 Preparation of terf-butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate Boc
  • Step 2 Preparation of tert- butyl 3-(2-(tosyloxy)ethyl)azetidine-l-carboxylate Boc
  • Step 3 Preparation of tert- butyl 3-(2-cyanoethyl)azetidine-l-carboxylate Boc
  • Step 4 Preparation of tert- butyl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate
  • Step 6 (J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate
  • Example 8 Preparation of sodium ( 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2 - methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetyl)(methylsulfonyl)amide
  • Step 1 Preparation of tert- butyl 3-formylazetidine-l-carboxylate
  • Step 3 Preparation of (J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(methylsulfonamido)-2- oxoethyl)azetidine-l-carboxylate
  • Step 4 Preparation of sodium(2-(l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetyl)(methylsulfonyl)amide
  • Example 9 Preparation of sodium ethyl(2-(l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate
  • Step 1 Preparation of tert-butyl 3-formylazetidine-l-carboxylate
  • Step 2 Preparation of (E)-tert-but ⁇ 3-(2-(diethoxyphosphoryl)vinyl)azetidine-l- carboxylate
  • Step 3 Preparation of tert- butyl 3-(2-(ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l- carboxylate
  • Step 5 Preparation of (3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2- (ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l-carboxylate
  • Step 6 Preparation of sodium ethyl(2-(l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate
  • reaction mixture was lyophilized to give sodium ethyl(2-(l-((((3i?, ⁇ ri',5ri', 6/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)phosphonate as a solid (57 mg).
  • Example 10 Preparation of sodium 2-((l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)-2-methylpropanoate
  • Step 1 Preparation of ferf-Z>Hiy/ 3-((l-methoxy-l-oxopropan-2-yl)oxy)azetidine-l- carboxylate
  • Step 5 Preparation of 2-((l-((((J ?, A,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)-2-methylpropanoic acid
  • Step 6 Preparation of sodium 2-(( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)-2-methylpropanoate
  • Example 11 Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-2,2-dimethylpropanoate
  • Step 1 Preparation of (E)-tert-butyl 3-(3-ethoxy-3-oxoprop-l-en-l-yl)azetidine-l- carboxylate
  • Step 2 Preparation of tert-butyl 3-(3-ethoxy-3-oxopropyl)azetidine-l-carboxylate
  • Step 3 Preparation of tert-butyl 3-(3-ethoxy-2,2-dimethyl-3-oxopropyl)azetidine-l- carboxylate
  • Step 6 Preparation of 3-(3-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)cyclobutyl)-2,2- dimethylpropanoic acid
  • Step 7 Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
  • Example 12 Preparation of sodium (3-(l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoyl)(methylsulfonyl)amide
  • Step 1 Preparation of terf-butyl3-(3-(methylsulfonamido)-3-oxopropyl)azetidine-l- carboxylate Boc
  • Step 3 Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(3-(methylsulfonamido)-3- oxopropyl)azetidine-l-carboxylate
  • Step 4 Preparation of Sodium (3-( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
  • Example 13 Preparation of sodium 5-((l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate
  • Step 1 Preparation of tert-butyl 3-(4-ethoxy-2,4-dioxobutyl)azetidine-l-carboxylate
  • Step 2 Preparation of tert-butyl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l- carboxylate
  • Step 4 Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((3-hydroxyisoxazol-5- yl)methyl)azetidine-l-carboxylate
  • Step 5 Preparation of sodium 5-((l-((((3/?, ,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
  • Lyophilization provided 5 -( ( 1 - (( (( 3/( -fS, 5S, 6R)-5- methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate sodium salt (139 mg) as a white solid.
  • Example 14 Preparation of sodium 5-(2-(l-((((3/?, £,5»£, ⁇ i/?)-5-inethoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)isoxazol-3-olate
  • Step 1 Preparation of 3-(l-(terf-butoxycarbonyl)azetidin-3-yl)propanoic acid
  • Step 2 Preparation of tert- butyl 3-(5-ethoxy-3,5-dioxopentyl)azetidine-l-carboxylate
  • Step 3 Preparation of tert-butyl 3-(2-(3-hydroxyisoxazol-5-yl)ethyl)azetidine-l- carboxylate
  • Step 5 Preparation of sodium 5-(2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
  • Lyophilization give 5-(2-( l -((((3//,-/ri',5A6//)-5-methoxy-4-((2//,3//)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)isoxazol-3-olate sodium salt (130 mg) as a colorless foam.
  • Example 15 Preparation of sodium 3-((l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoate
  • Step 1 Preparation of tert- butyl 3-(3-(/er/-butoxy)-3-oxopropoxy)azetidine- l-carboxylate
  • Step 3 Preparation of 3-((l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)propanoic acid
  • DIPEA (0.35mL, 2.0 mmol) was added dropwise to a solution of 3-(azetidin-3- yloxy)propanoic acid (290 mg, 2.0 mmol) in acetonitrile (5 mL) at 0°C under N 2. The mixture was stirred at 0°C for 10 min and Intermediate 1 (150 mg, 0.33 mmol) and DMAP (5 mg) were added. After addition, the reaction was stirred at room temperature overnight under N 2 atmosphere. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was acidified by ammonium acetate buffer and 5% sodium bicarbonate solution (20 mL), then extratcted with dichloromethane.
  • Example 16 Preparation of sodium 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate
  • Step 1 Preparation of (S)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate
  • Step 3 Preparation of 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid
  • Step 4 Preparation of sodium 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin- 3-yl)oxy)acetate
  • Example 17 Preparation of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl3-(2- (((isopropoxycarbonyl)oxy)methoxy)-2-oxoethoxy) azetidine-l-carboxylate
  • Example 19 Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)amino)acetate
  • Step 1 Preparation of tert-butyl 3-((2-(tert-butoxy)-2-oxoethyl)amino)azetidine-l- carboxylate
  • Step 3 Preparation of 2-((l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)amino)acetic acid
  • Step 4 Preparation of sodium 2-(( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)amino)acetate
  • Example 20 Preparation of sodium 2-(((/?)-l-(((( /?, S,5A,6/?)-5-methoxy-4-((.2/?, /?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate
  • Step 1 Preparation of ( ?)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate
  • Step 3 Preparation of 2-((( ?)-l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid
  • Step 4 Preparation of sodium 2-(((R)-l-((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-
  • Example 21 Preparation of sodium 2-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)acetate
  • Step 1 Preparation of ethyl 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetate
  • dichloromethane (10 mL) was added tert-butyl 2-oxoacetate (1.04 g, 4.0 mmol, 50% in toluene) dropwise at 0°C.
  • the mixture was stirred at 0°C for 10 min, and sodium triacetoxyborohydride (848 mg, 4.0 mmol) was added at 0°C.
  • the reaction was stirred at room temperature overnight under N 2 atmosphere. The reaction mixture was then washed with water.
  • the dichloromethane solution was dried over Na 2 S0 4 , filtered and concentrated to give crude product.
  • Step 4 Preparation of 2-(3-((((((.?/?, S,5»S,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)acetic acid
  • Step 5 Preparation of sodium 2-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
  • Example 22 Preparation of Preparation of sodium 4-(((((((( /?, S,5»S,6/?)-5-methoxy-4- ((2»V,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
  • Step 2 Preparation of allyl 4-(((tert-butox carbonyl)amino)methyl)benzoate
  • Step 4 Preparation of allyl 4-((((((((3/?, V,5,V,6/?)-5-methoxy-4-((2V,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
  • Step 5 Preparation of 4-(((((((((3/?, V,5,V,6/?)-5-methoxy-4-((2V,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoic acid
  • Step 6 Preparation of sodium 4-(((((((((J/?, V,5,V,6/?)-5-methoxy-4-((2V,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
  • Step 1 Preparation of 4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoic acid
  • Step 2 Preparation of sodium 4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoate
  • Example 24 Preparation of sodium 6-((((((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate
  • Step 3 Preparation of allyl 6-(((tert-butox carbonyl)amino)methyl)nicotinate
  • Step 5 Preparation of allyl 6-((((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate
  • Step 6 Preparation of 6-(((((((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinic acid
  • Step 7 Preparation of sodium 6-((((((((((3/?, S,5A,6/?)-4-((2/?,3/?)-3-ethyl-2-methyloxiran-2- yl)-5-methoxy-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinate
  • Example 25 Preparation of sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3- carboxylate
  • Step 1 l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3-carboxylic acid
  • Step 2 Preparation of sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3- carboxylate
  • Example 26 Preparation of sodium 2-((R)-3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
  • Step 1 Preparation of tert-butyl (R)-2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate
  • Step 2 Preparation of(R)-2-(3-aminopyrrolidin-l-yl)acetic acid [0215] To a solution of tert-butyl (R)-2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate (600mg, 2.00 mmol) in DCM (2 mL) was added TFA (1 mL). The reaction mixture was then stirred at room temperature for 3 hours. Concentration gave crude (R)-2-(3- aminopyrrolidin-l-yl)acetic acid TFA salt (560 mg) as a brown oil. LC-MS (ESI) found:306 [M+l] + .
  • Step 3 Preparation of 2-((R)-3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetic acid
  • Step 4 Preparation of sodium 2-((R)-3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
  • Example 27 Preparation of sodium 2-((S)-3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
  • Step 1 Preparation of (S)-tert-butyl 2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate
  • Step 3 Preparation of sodium 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
  • Example 28 Preparation of sodium 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate
  • Step 1 Preparation of ethyl 2-(4-((tert-butoxycarbonyl)amino)piperidin-l-yl)acetate
  • Step 3 Preparation of ethyl 2-(4-(((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate
  • Step 4 Preparation of 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetic acid
  • Step 5 Preparation of sodium 2-(4-(((((3/?, ,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
  • Example 29 Preparation of sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)phosphonate
  • Step 1 Preparation of l-benzhydrylazetidin-3-yl methanesulfonate
  • Step 3 Preparation of ethyl hydrogen (l-benzhydrylazetidin-3-yl)phosphonate
  • Step 4 Preparation of ethyl hydrogen azetidin-3-ylphosphonate
  • Step 5 Preparation of ammonium ethyl (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-
  • Step 6 Preparation of sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
  • Example 30 Preparation of sodium 3-(3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)propanoate
  • Step 1 Preparation of methyl 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoate
  • Step 2 Preparation of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoic acid
  • Step 3 Preparation of 3-(3-aminoazetidin-l-yl)propanoic acid trifluoroacetic acid salt
  • Step 4 Preparation of 2-(3-((((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)acetic acid
  • Step 5 Preparation of sodium 2-(3-(((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
  • Example 31 Preparation of sodium 2-(4-((((J ?, V,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate
  • Step 1 Preparation of tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)piperazine-l-carboxylate
  • Step 3 Preparation of 2-(4-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetic acid
  • Step 4 Preparation of sodium 2-(4-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetate
  • Example 32 Preparation of sodium 2-(4-(((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
  • Step 1 Preparation of ethyl 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol- l-yl)acetate
  • Step 2 Preparation of 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l- yl)acetic acid
  • Example 33 Preparation of sodium 2-(4-(((((((3 R,4S,5S,6R)-5-metho ⁇ y-4-((2R,3 R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
  • Step 1 Preparation of ethyl 2-(4-((methylamino)methyl)-lH-l,2,3-triazol-l-yl)acetate
  • Step 3 Preparation of sodium 2-(4-(((((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-
  • Example 34 Preparation of sodium (J/?,5/?)-3,5-dihydroxy-7-(((((J/?, V,5.V,6/?)-5- methoxy-4-((2 ?,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
  • Step 1 Preparation of (.?/?, 5/?)-7-amino-3,5-dihydroxyheptanoic acid
  • Step 3 Preparation of (3/?,5/?)-3,5-dihydroxy-7-(((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)heptanoic acid
  • Step 4 Preparation of sodium (3/?,5/?)-3,5-dih droxy-7-(((((3/?,4S,5A,6/?)-5-methoxy-4-
  • Example 35 Preparation of sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoate
  • Step 2 Preparation of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)-2,2- dimethylpropanoic acid
  • Step 4 Preparation of 3-(3-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)-2,2-dimethylpropanoic acid
  • Step 5 Preparation of sodium 3-(3-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoic acid
  • Example 36 Preparation of sodium ( 3R,5R)-3,5-dihydroxy-7-((l-((((3R,4S,5S,6R)-5 - methoxy-4-((2 ?,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
  • Example 37 Preparation of sodium 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro [3.3] heptan-2-yl)acetate
  • Example 38 Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- oxopiperazin-l-yl)acetate
  • Example 39 Preparation of sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptan-6-yl)oxy)acetate equivalent
  • the solvent was lyophilized to give sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro[3.3]heptan-6-yl)oxy)acetate (100 mg) as a white solid.
  • Example 40 Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3- methylazetidin-3-yl)oxy)acetate , ,
  • Example 41 Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate

Abstract

The disclosure provides, at least in part, liver, intestine and/or kidney-targeting compounds and their use in treating liver, intestine and/or kidney disorders, such as non-alcoholic steatohepatitis, alcoholic steatohepatitis, hepatocellular carcinoma, liver cirrhosis, and hepatitis B; and/or chronic kidney disease, glomerular disease such as IGA nephropathy, lupus nephritis, or polycystic kidney disease. The compounds are contemplated to have activity against methionyl aminopeptidase 2.

Description

TARGETING COMPOUNDS
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of, and priority to, U.S. provisional application serial numbers 62/654,710, filed April 9, 2018; and 62/597,598, filed December 12, 2017; the contents of each of which are hereby incorporated by reference herein in their entirety.
BACKGROUND
[0002] Membrane transporters are found throughout the body and play an important role in drug disposition. The tissue distribution of a drug can have a significant impact on both the agents’ safety and efficacy. Of the over 400 human transporters about 20 of them are important for drug transport (Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010; 9(3):2l5-36).
[0003] There are two types of drug transporters; the solute carriers (SLC) and the ATP- binding cassette (ABC) transporters. SLC transporters are expressed in many tissues such as brain, liver, small intestine, kidney and are involved in the uptake of drugs and nutrients into cells. SLC transporters are categorized into three categories (i) the first type transport by facilitated diffusion via a concentration gradient, (ii) the second type utilizes Na+ or H+ cotransport and (iii) the third type involves exchanging substrates to aid transport. The main SLC transporters include organic anion transporting polypeptides (OATP), organic anion transporters (OAT) and organic cation transporters (OCT).
[0004] The OATPs (SLCOs) represent a superfamily of membrane transport proteins that mediate the sodium-independent transport of a diverse range of amphiphilic organic
compounds. They are expressed in the liver and various other organs and are involved in the distribution of drugs from the blood to tissues. OATP1B1 (SLCOlBl), 1B3 (SLC01B3), and 2B1 (SLC02B1) are important isoforms for drug transport and are localized to the sinusoidal (basolateral) membrane of hepatocytes (Hagenbuch B., & Meier PJ, The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003; 1609(1); 1-18; Fenner KS, Jones HM, Ullah M, Kempshall S, Dickins M, Lai Y, Morgan P, Barton HA. The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: from obscure liver transporters to key determinants of hepatobiliary clearance. Xenobiotica. 2012 Jan; 42(l):28- 45. Epub 2011 Nov 11). [0005] Within the liver, OATP1B1 is expressed uniformly throughout the lobules. The OATP1B1 transporters exhibit broad substrate selectivity that includes anionic (e.g. statins such as pravastatin, pitavastatin, and rosuvastatin), zwitterionic (e.g. rifampicin) and neutral lipophilic (e.g. paclitaxel) drugs. OATP1B1 also transports endogenous substances such as bile acids, thyroid hormones, steroid sulfates, glucuronide conjugates and peptides.
[0006] OATP1B3 is in the sinusoidal membranes of hepatocytes located around the central vein. OATP1B3 has considerable substrate overlap with OATP1B1 but the gastrointestinal peptide Cholecystokinin is reported to be exclusively transported by OATP1B3. Other specific OATP1B3 substrates (amongst the OAT transporters) include docetaxel, digoxin, paclitaxel and the toxin amanitin (Letschert, K., et al., Molecular characterization and inhibition of amanitin uptake into human hepatocytes. Toxicol Sci, 2006. 91(1): p. 140-9). CCK-8 is a specific inhibitor of OATP1B3. Cyclosporine and CI-1034 are inhibitors of the three major OATP isoforms (Rizwan, AN. and Burckhardt, G, Organic anion transporters of the SLC22 family: biopharmaceutical, physiological, and pathological roles. Pharm Res, 2007. 24(3): p. 450-70; Sahi, T, et al., Metabolism and transporter-mediated drug-drug interactions of the endothelin-A receptor antagonist CI-1034. Chem Biol Interact, 2006. 159(2): p. 156-68).
[0007] As human OATP1B1 and OAT1B3 are liver specific they may be used for liver targeting to enhance liver concentration and minimize drug exposure to the peripheral tissues (e.g. brain) to reduce toxicity and/or side effects. Some medicinal chemistry design principles of orally active drugs for liver targeting through OATP transporters have been described (Tu M, Mathiowetz AM, Pfefferkom JA, Cameron KO, Dow RL, Litchfield J, Di L, Feng B, Liras S. Medicinal chemistry design principles for liver targeting through OATP transporters. Curr Top Med Chem. 2013; l3(7):857-66). These principles include, e.g., efficient hepatocyte uptake via OATP1B1 and/or OATP1B3 transporters which generally require an acidic function in the molecule, low passive permeability to minimize distribution into peripheral tissues that lack liver-specific OATP transporters, and sufficient bioavailability to support oral
administration.
[0008] Whilst OATP1B1 and 1B3 have been primary human liver-specific transporter targets, OATP2B1 and Sodium Tauracholate Transporting Polypeptide (NCTP1) represent additional human liver uptake transporters which could contribute to a liver targeted approach.
[0009] In addition to the sinusoidal membrane of the hepatocyte, OATP2B1 is widely expressed in tissues including apical membrane of enterocytes, the basal membrane of the syncytiotrophoblasts, the luminal membrane of brain capillary endothelial cells, small arteries and veins of the heart, and the lung. OATP2B1 has broad substrate specificity transporting mostly anionic organic endo- and xenobiotics. It exhibits some pH dependency in that it seems to transport some substrates more efficiently at lower pH. Its substrate specificity appears somewhat more restricted than OATP1B1 and 1B3, however it transports fexofenadine, statins, glibenclamide, glyburide, estrone-3 -sulfate (E3S), dehydroepiandrosterone-3-sulfate (DHEAS), prostaglandin E2, and taurocholate. OATP2B1 in the liver mediates the hepatic uptake of many xenobiotics, including bromosulphophthalein (BSP), benzylpenicillin, fexofenadine, glibenclamide, pravastatin, atorvastatin, rosuvastatin, and fluvastatin. The apparent pH dependency of transport may be important for the gastrointestinal transport of drug substrates such as fexofenadine, as pH varies substantially along the gastro-intestinal tract. OATP2B1 inhibitors include many organic anions, mono- and dicarboxylic acids, steroid hormones and their derivatives, drugs, such as rifamycin SV, pravastatin cyclosporine A and gemfibrozil as well as constituents of citrus juices and herbal extracts.
[0010] OATP family members are poorly conserved evolutionarily and orthologues for human OATPs may not exist in rodents. The significant species differences for the
OATP/Oatp transporters make it challenging to translate pharmacokinetic or pharmacodynamic data from preclinical species to humans.
[0011] Sodium Taurocholate Co-Transporting Polypeptide (NTCP, SLOC10A1) is another liver-specific transporter located on the basolateral membrane domain of hepatocytes in human, rat and other species (Stieger, B., et ah, In situ localization of the hepatocytic Na+/Taurocholate cotransporting polypeptide in rat liver. Gastroenterology, 1994. 107(6): p. 1781-7).
[0012] NTCP is primarily responsible for the uptake of bile acids from the sinusoids. NTCP is one of the key transporters in the enterohepatic circulation of bile acids, which also include the canalicular efflux transporter BSEP, and ASBT and OSTa/b in the gastrointestinal tract (Doring, B., et ah, The SLC10 carrier family: transport functions and molecular structure. Curr Top Membr, 2012. 70: p. 105-68). Other substrates include sulfated steroids (estrone-3 -sulfate, DHEAS) and thyroid hormones. In comparison to sodium independent organic anion transporters of the SLCO family (i.e. OATPs), NTCP interacts with a relatively narrow panel of drugs and other xenobiotics. A computational model of a substrate pharmacophore has been developed (Dong Z. et al. A Substrate Pharmacophore for the Human Sodium Taurocholate Co-transporting Polypeptide. Int J Pharm. 2015 Jan 15; 478(1): 88-95). NTCP is important in the hepatic uptake of statins, particularly rosuvastatin, and is therefore a risk factor for transporter-mediated DDI for this drug. [0013] Chronic liver diseases such as fatty liver disease and liver fibrosis can be risk factors that may dispose progression to hepatocellular carcinoma, and represent a common and difficult clinical challenge of worldwide importance. For example, liver fibrosis is the excessive accumulation of extracellular matrix proteins including collagen that occurs in most types of chronic liver diseases. Advanced liver fibrosis results in cirrhosis, liver failure, and portal hypertension and often requires liver transplantation. Non-alcoholic steatohepatitis (NASH) is a severe form of fatty liver disease that affects 2 to 5 percent of Americans. Many people with NASH have risk factors also associated with HCC, e.g., obesity, diabetes and/or elevated blood cholesterol levels. Alpha-l antitrypsin deficiency is an inherited disorder associated with an increased risk of liver and lung disease. The disorder is associated with the retention of the liver-produced protein alpha-l antitrypsin in the liver and low levels of alpha-l antitrypsin in the serum. Clinical features of alpha-l antitrypsin deficiency include chronic liver inflammation and liver fibrosis, which can progress in advanced forms to chronic hepatitis, cirrhosis and hepatocellular carcinoma. Chronic kidney disease may increase the risk for recurrence and progression of other cancers such as non-muscle invasive bladder cancer, and can also limit the ability to receive and withstand cancer therapies. Other kidney diseases such as lupus nephritis can lead to kidney failure and patients suffering from lupus nephritis are at a higher risk for cancers such as B-cell lymphoma, and for heart and blood vessel problems.
[0014] Methionine aminopeptidase 2 (MetAP2) encodes a protein that functions at least in part by enzymatically removing the amino terminal methionine residue from certain newly translated proteins such as glyceraldehyde-3 -phosphate dehydrogenase. Increased expression of the MetAP2 gene has been historically associated with various forms of cancer. Molecules inhibiting the enzymatic activity of MetAP2 have been identified and have been explored for their utility in the treatment of various tumor types.
SUMMARY
[0015] This disclosure is directed in part to, for example, carboxylic acid (and carboxylate salt) sidechains appended to the 6-position alcohol of fumagillol that are MetAP2 modulators selectively targeting the liver, intestine and/or kidney, their use as medicinal agents, processes for their preparation, and pharmaceutical compositions containing them as an active ingredient both alone or in combination with other agents, as well as provides for their use as
medicaments and/or in the manufacture of medicaments for the inhibition of MetAP2 activity, and for the treatment of a liver, intestinal and/or kidney disorder, in warm-blooded animals such as humans. Also provided are pharmaceutical compositions comprising at least one disclosed compound and a pharmaceutically acceptable carrier. [0016] In an embodiment, disclosed herein is a pharmaceutically acceptable base salt of a compound represented by:
Figure imgf000006_0001
wherein L1, L2 and T are as defined herein.
[0017] Also provided herein are methods of treating or mitigating a liver, intestine and/or kidney disorder in a patient in need thereof, comprising administering an effective amount of a compound described herein to the patient. In some embodiments, the liver disorder may be one or more of, for example, non-alcoholic steatohepatitis, alcoholic steatohepatitis, carcinoma (e.g., hepatocellular carcinoma), liver cirrhosis, and hepatitis B. In other embodiments, the kidney disorder may be one or more of chronic kidney disease, glomerular disease such as iGA nephropathy, lupus nephritis, polycystic kidney disease.
[0018] For example, disclosed herein are methods of treating, or preventing the development of, hepatic preneoplastic lesions in patient in need thereof, comprising administering an effective amount of a compound disclosed herein. In some embodiments, for example, the patient may be at risk for but not yet suffering from NASH. In some embodiments, the patient may have at least one of, for example: minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation, and/or balloon degeneration. In some embodiments, the patient may suffer from type 2 diabetes and/or is obese. In some embodiments the patient may suffer from alpha- 1 antitrypsin deficiency. In some embodiments, provided herein are methods of treating hepatocellular carcinoma in a patient in need thereof, comprising administering to the patient an effective amount of a disclosed compound.
[0019] Contemplated methods of treating may also include administering a disclosed compound in combination with one or more other agents (and/or in combination with weight loss surgery and/or weight loss therapy). For example, contemplated methods herein can include administering one or more additional agents, e.g. as disclosed herein, for example, an anti diabetic medication (metformin), a modulator of glucagon -like peptide -1 receptor such as exenatide and liraglutide, obeticholic acid, a PPAR agonist, and/or a thiazolidinedione such as pioglitazone. DETAILED DESCRIPTION
[0020] The features and other details of the disclosure will now be more particularly described. Before further description of the present invention, certain terms employed in the specification, examples and appended claims are collected here. These definitions should be read in light of the remainder of the disclosure and as understood by a person of skill in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art.
Definitions
[0021] “Treating” includes any effect, e.g., lessening, reducing, modulating, or eliminating, that results in the improvement of the condition, disease, disorder and the like.
[0022] The term“alkenyl” as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond. Exemplary alkenyl groups include, but are not limited to, a straight or branched group of 2-6 or 3-4 carbon atoms, referred to herein as C2-6alkenyl, and C3-4alkenyl, respectively. Exemplary alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, etc.
[0023] The term“alkoxy” as used herein refers to a straight or branched alkyl group attached to oxygen (alkyl-O-). Exemplary alkoxy groups include, but are not limited to, alkoxy groups of 1-6 or 2-6 carbon atoms, referred to herein as Ci-6alkoxy, and C2-6alkoxy, respectively. Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, isopropoxy, etc.
[0024] The term“alkoxyalkyl” as used herein refers to a straight or branched alkyl group attached to oxygen, attached to a second straight or branched alkyl group (alkyl -O-alkyl-). Exemplary alkoxyalkyl groups include, but are not limited to, alkoxyalkyl groups in which each of the alkyl groups independently contains 1-6 carbon atoms, referred to herein as Ci. 6alkoxy-Ci-6alkyl. Exemplary alkoxyalkyl groups include, but are not limited to
methoxym ethyl, 2-methoxyethyl, 1 -methoxy ethyl, 2-methoxypropyl, ethoxym ethyl, 2- isopropoxy ethyl etc.
[0025] The term“alkyoxycarbonyl” as used herein refers to a straight or branched alkyl group attached to oxygen, attached to a carbonyl group (alkyl -O-C(O)-). Exemplary
alkoxycarbonyl groups include, but are not limited to, alkoxycarbonyl groups of 1-6 carbon atoms, referred to herein as Ci-6alkoxycarbonyl. Exemplary alkoxycarbonyl groups include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, t-butoxycarbonyl, etc.
[0026] The term“alkenyloxy” used herein refers to a straight or branched alkenyl group attached to oxygen (alkenyl-O-). Exemplary alkenyloxy groups include, but are not limited to, groups with an alkenyl group of 3-6 carbon atoms, referred to herein as C3-6alkenyloxy.
Exemplary“alkenyloxy” groups include, but are not limited to allyloxy, butenyloxy, etc.
[0027] The term“alkynyloxy” used herein refers to a straight or branched alkynyl group attached to oxygen (alkynyl-O). Exemplary alkynyloxy groups include, but are not limited to, groups with an alkynyl group of 3-6 carbon atoms, referred to herein as C3-6alkynyloxy.
Exemplary alkynyloxy groups include, but are not limited to, propynyloxy, butynyloxy, etc.
[0028] The term“alkyl” as used herein refers to a saturated straight or branched hydrocarbon. Exemplary alkyl groups include, but are not limited to, straight or branched hydrocarbons of 1-6, 1-4, or 1-3 carbon atoms, referred to herein as Ci-6alkyl, Ci-4alkyl, and Ci-3alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l-butyl, 3 -methyl -2 -butyl, 2-methyl- 1 -pentyl, 3-methyl-l-pentyl, 4- methyl-l -pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2 -pentyl, 2,2-dimethyl- 1- butyl, 3, 3 -dimethyl -1 -butyl, 2-ethyl- 1 -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, etc.
[0029] The term“alkylcarbonyl” as used herein refers to a straight or branched alkyl group attached to a carbonyl group (alkyl-C(O)-). Exemplary alkylcarbonyl groups include, but are not limited to, alkylcarbonyl groups of 1-6 atoms, referred to herein as Ci-6alkylcarbonyl groups. Exemplary alkylcarbonyl groups include, but are not limited to, acetyl, propanoyl, isopropanoyl, butanoyl, etc.
[0030] The term“alkynyl” as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond. Exemplary alkynyl groups include, but are not limited to, straight or branched groups of 2-6, or 3-6 carbon atoms, referred to herein as C2-6alkynyl, and C3-6alkynyl, respectively. Exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, etc.
[0031] The term“carbonyl” as used herein refers to the radical -C(O)-.
[0032] The term“cyano” as used herein refers to the radical -CN.
[0033] The term“cycloalkoxy” as used herein refers to a cycloalkyl group attached to oxygen (cycloalkyl-O-). Exemplary cycloalkoxy groups include, but are not limited to, cycloalkoxy groups of 3-6 carbon atoms, referred to herein as C3-6cycloalkoxy groups. Exemplary cycloalkoxy groups include, but are not limited to, cyclopropoxy, cyclobutoxy, cyclohexyl oxy, etc
[0034] The terms“cycloalkyl” or a“carbocyclic group” as used herein refers to a saturated or partially unsaturated hydrocarbon group of, for example, 3-6, or 4-6 carbons, referred to herein as C3-6cycloalkyl or C4-6cycloalkyl, respectively. Exemplary cycloalkyl groups include, but are not limited to, cyclohexyl, cyclopentyl, cyclopentenyl, cyclobutyl or cyclopropyl.
[0035] The terms“halo” or“halogen” as used herein refer to F, Cl, Br, or I.
[0036] The terms“heteroaryl” or“heteroaromatic group” as used herein refers to a monocyclic aromatic 5-6 membered ring system containing one or more heteroatoms, for example one to three heteroatoms, such as nitrogen, oxygen, and sulfur. Where possible, said heteroaryl ring may be linked to the adjacent radical though carbon or nitrogen. Examples of heteroaryl rings include but are not limited to furan, thiophene, pyrrole, thiazole, oxazole, isothiazole, isoxazole, imidazole, pyrazole, triazole, pyridine or pyrimidine etc.
[0037] The terms“heterocyclyl” or“heterocyclic group” are art-recognized and refer to saturated or partially unsaturated, 4-10 membered ring structures, including spirocyclic, bridged or fused rings, and whose ring structures include one to three heteroatoms, such as nitrogen, oxygen, and sulfur. Where possible, heterocyclyl rings may be linked to the adjacent radical through carbon or nitrogen. Examples of heterocyclyl groups include, but are not limited to, pyrrolidine, piperidine, morpholine, thiomorpholine, piperazine, oxetane, azetidine, tetrahydrofuran or dihydrofuran etc.
[0038] The term“heterocyclyloxy” as used herein refers to a heterocyclyl group attached to oxygen (heterocyclyl -0-).
[0039] The term“heteroaryloxy” as used herein refers to a heteroaryl group attached to oxygen (heteroaryl-O-).
[0040] The terms“hydroxy” and“hydroxyl” as used herein refers to the radical -OH.
[0041] The term“oxo” as used herein refers to the radical =0.
[0042] “Pharmaceutically or pharmacologically acceptable” include molecular entities and compositions that should not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate. For human administration, preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologies standards.
[0043] The term“pharmaceutically acceptable carrier” or“pharmaceutically acceptable excipient” as used herein refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical
administration. The use of such media and agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
[0044] The term“pharmaceutical composition” as used herein refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
[0045] “ Individual,”“patient,” or“subject” are used interchangeably and include any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans. The compounds of the disclosure can be administered to a mammal, such as a human, but can also be administered to other mammals such as an animal in need of veterinary treatment, e.g., domestic animals (e.g, dogs, cats, and the like), farm animals (e.g, cows, sheep, pigs, horses, and the like) and laboratory animals (e.g, rats, mice, guinea pigs, and the like). “Modulation” includes antagonism (e.g, inhibition), agonism, partial antagonism and/or partial agonism.
[0046] In the present specification, the term“therapeutically effective amount” means the amount of the subject compound that will elicit the biological or medical response of a tissue, system or animal, (e.g. mammal or human) that is being sought by the researcher, veterinarian, medical doctor or other clinician. The compounds of the disclosure are administered in therapeutically effective amounts to treat a disease. Alternatively, a therapeutically effective amount of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect, such as an amount that results in substantially mitigating a liver or kidney disease or disorder.
[0047] The term "pharmaceutically acceptable salt(s)" as used herein refers to salts of acidic or basic groups that may be present in compounds used in the compositions. Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including, but not limited to, malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, / oluenesulfonate and pamoate (i.e., 1 , 1 '-methylene-/v.s-(2- hydroxy-3-naphthoate)) salts. Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts, particularly calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts. Compounds included in the present compositions that include a basic or acidic moiety may also form pharmaceutically acceptable salts with various amino acids. The compounds of the disclosure may contain both acidic and basic groups; for example, one amino and one carboxylic acid group. In such a case, the compound can exist as an acid addition salt, a zwitterion, or a base salt.
[0048] The compounds of the disclosure may contain one or more chiral centers and, therefore, exist as stereoisomers. The term“stereoisomers” when used herein consist of all enantiomers or diastereomers. These compounds may be designated by the symbols“(+),”“(- ),”“R” or“S,” depending on the configuration of substituents around the stereogenic carbon atom, but the skilled artisan will recognize that a structure may denote a chiral center implicitly. The present invention encompasses various stereoisomers of these compounds and mixtures thereof. Mixtures of enantiomers or diastereomers may be designated“(±)” in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
[0049] The compounds of the disclosure may contain one or more double bonds and, therefore, exist as geometric isomers resulting from the arrangement of substituents around a carbon-carbon double bond. The symbol . denotes a bond that may be a single, double or triple bond as described herein. Substituents around a carbon-carbon double bond are designated as being in the“Z’ or“£” configuration wherein the terms“Z’ and“£” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the“ E” and“Z” isomers. Substituents around a carbon-carbon double bond alternatively can be referred to as“cis” or“trans,” where“cis” represents substituents on the same side of the double bond and“trans” represents substituents on opposite sides of the double bond.
[0050] Compounds of the disclosure may contain a carbocyclic or heterocyclic ring and therefore, exist as geometric isomers resulting from the arrangement of substituents around the ring. The arrangement of substituents around a carbocyclic or heterocyclic ring are designated as being in the“Z” or“E” configuration wherein the terms“Z” and“E” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting carbocyclic or heterocyclic rings encompass both“Z” and“E” isomers. Substituents around a carbocyclic or heterocyclic rings may also be referred to as“cis” or“trans”, where the term“cis” represents substituents on the same side of the plane of the ring and the term“trans” represents substituents on opposite sides of the plane of the ring. Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated“cis/trans.”
[0051] Individual enantiomers and diasteriomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, (3) direct separation of the mixture of optical enantiomers on chiral liquid chromatographic columns or (4) kinetic resolution using stereoselective chemical or enzymatic reagents. Racemic mixtures can also be resolved into their component enantiomers by well known methods, such as chiral-phase liquid chromatography or crystallizing the compound in a chiral solvent. Stereoselective syntheses, a chemical or enzymatic reaction in which a single reactant forms an unequal mixture of stereoisomers during the creation of a new stereocenter or during the transformation of a pre-existing one, are well known in the art. Stereoselective syntheses encompass both enantio- and diastereoselective transformations, and may involve the use of chiral auxiliaries. For examples, see Carreira and Kvaemo, Classics in Stereoselective Synthesis , Wiley-VCH: Weinheim, 2009
[0052] The compounds disclosed herein can exist in solvated as well as unsolvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. In one embodiment, the compound is amorphous. In one embodiment, the compound is a single polymorph. In another embodiment, the compound is a mixture of polymorphs. In another embodiment, the compound is in a crystalline form.
[0053] The invention also embraces isotopically labeled compounds of the invention which are identical to those recited herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as ¾ ¾ 13C, 14C, 15N, 180, 170, 31P, 32P, 35 S, 18F, and 36Cl, respectively.
For example, a compound of the invention may have one or more H atom replaced with deuterium. [0054] Certain isotopically-labeled disclosed compounds ( e.g ., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-l4 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labeled compounds of the invention can generally be prepared by following procedures analogous to those disclosed in the examples herein by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
[0055] Also contemplated herein are prodrugs of disclosed compounds. The term“prodrug” refers to compounds that are transformed in vivo to yield a disclosed compound or a
pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (such as by esterase, amidase, phosphatase, oxidative and or reductive metabolism) in various locations (such as in the intestinal lumen or upon transit of the intestine, blood or liver). Prodrugs are well known in the art (for example, see Rautio,
Kumpulainen, et al, Nature Reviews Drug Discovery 2008, 7, 255). For example, if a compound of the invention or a pharmaceutically acceptable salt, hydrate or solvate of the compound contains a carboxylic acid functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as (Ci-8)alkyl, (C2-i2)alkylcarbonyloxymethyl, 1 -(alkyl carbonyl oxy)ethyl having from 4 to 9 carbon atoms, 1- methyl-l-(alkylcarbonyloxy)-ethyl having from 5 to 10 carbon atoms,
alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, l-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1 -methyl- l-(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms,
l-(N-(alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl,
4-crotonolactonyl, gamma-butyrol acton-4 -yl, di-N,N-(Ci-2)alkylamino(C2.3)alkyl (such as b- dimethylaminoethyl), carbamoyl-(Ci-2)alkyl, N,N-di(Ci-2)alkylcarbamoyl-(Ci-2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-3)alkyl.
[0056] Similarly, if a compound of the invention contains an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (Ci-6)alkylcarbonyloxymethyl, l-((Ci-6)alkylcarbonyloxy)ethyl, 1 -methyl- l-((Ci. 6)alkylcarbonyloxy)ethyl (Cl-6)alkoxycarbonyloxym ethyl, N-(Ci.
6)alkoxycarbonylaminomethyl, succinoyl, (Ci-6)alkylcarbonyl, a-amino(Ci-4)alkylcarbonyl, arylalkylcarbonyl and a-aminoalkylcarbonyl, or a-aminoalkylcarbonyl-a-aminoalkylcarbonyl, where each a-aminoalkylcarbonyl group is independently selected from the naturally occurring L-amino acids, P(0)(0H)2, -P(0)(0(Ci-6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).
[0057] If a compound of the invention incorporates an amine functional group, a prodrug can be formed, for example, by creation of an amide or carbamate, an N-alkylcarbonyloxyalkyl derivative, an (oxodioxolenyl)methyl derivative, an N-Mannich base, imine or enamine. In addition, a secondary amine can be metabolically cleaved to generate a bioactive primary amine, or a tertiary amine can metabolically cleaved to generate a bioactive primary or secondary amine. For examples, see Simplicio, et al, Molecules 2008, 13, 519 and references therein.
I. Compounds
[0058] In certain embodiments, the present disclosure provides a pharmaceutically acceptable base salt of a compound represented by:
Figure imgf000014_0001
wherein:
L1 is -NRXR2;
R1 is H or Ci-3alkyl, and R2 is selected from the group consisting of: a 4-9 membered monocyclic heterocyclic ring, heteroaryl, -Ci-2alkylene-heteroaryl, phenyl, -Ci-2alkylene- phenyl and -Ci-6alkylene; wherein -Ci-6alkylene may be straight or branched and may optionally be substituted with one or more substituents each independently selected from Rp; or
R1 and R2 taken together with the nitrogen to which they are attached form a 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring; wherein the 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring is optionally substituted with one or more substituents each independently selected from Rh;
L2 is selected from the group consisting of: Ci-4alkylene, -0-Ci-6alkylene, -NRa-Ci. 4alkylene, -S-Ci-4alkylene, - S(0)2-Ci-4alkylene, -S(0)-Ci-4alkylene, =CRa-, -Ci-2alkylene- heteroaryl, a 4-9 membered monocyclic heterocyclic ring, and a bond; wherein Ci-4alkylene and -0-Ci-6alkylene may be straight or branched and may optionally be substituted with one or more substituents each independently selected from Rp; T is selected from the group consisting of: -C(0)OH, tetrazole, -SO3H, isoxazol-3-ol, isothiazol-3-ol, l-alkyl-lH-pyrazol-3-ol, l,2,4-oxadiazol-5-ol, l,2,4-thiadiazol-5-ol, 1H- imidazole-2,5-dione, oxazolidine-2,4-dione-5-yl, thiazolidine-2,4-dione-5-yl, 1,2,4- oxadiazolidine-3,5-dione-2-yl, 4-hydroxy-l,5-dihydro-2H-pyrrol-2-one-l-yl, 6-hydroxy-4H- l,3-dioxin-4-one-2-yl, -C(0)NHS(0)2-Ci-3alkyl, -NH-S(0)2-Ci-3alkyl, -S(0)2-NH-Ci-3alkyl, and -P(0)(OH)(ORb); wherein -C(0)NHS(0)2-Ci-3alkyl, -NH-S(0)2-Ci-3alkyl and -S(0)2-NH- Cl-3alkyl may optionally be substituted with one or more substituents each independently selected from Rp;
Rp is independently selected, for each occurrence, from the group consisting of fluorine, hydroxyl, cyano, and -C(0)NRaRb; or two Rp are taken together to form =0;
Rh is independently selected, for each occurrence, from the group consisting of hydrogen, Ci-3alkyl, fluoro and hydroxyl; or two Rh are taken together to form =0; and
Ra and Rb are independently selected, for each occurrence, from the group consisting of hydrogen and C 1.3 alkyl.
[0059] In some embodiments, L1 is selected from the group consisting of:
Figure imgf000015_0001
wherein independently for each occurrence p is 1 or 2, q is 1 or 2, r is 0, 1 or 2, Z is CH2 or O, and Y is N or CH.
[0060] In some embodiments, L1 is selected from the group consisting of:
Figure imgf000015_0002
[0061] In some embodiments, L1 is selected from the group consisting of:
Figure imgf000015_0003
wherein independently for each occurrence p is 1 or 2, and q is 1 or 2. [0062] In some embodiments, L1 is selected from the group consisting of:
Figure imgf000016_0001
[0063] In some embodiments, L2 is selected from the group consisting of: -CH2-, -CH2-CH2-, -0-CH2-, -0-CH2-CH2-, -NRa-CH2-, -CH2-C(CH3)(CH3)-, -0-CH2-C(CH3)(CH3)-,-0-
C(CH3)(CH3)-, -0-CH2-CH2-CHRP-CH2-CHRP-CH2-, -CH2C(Rp)2-, -CH2-CHRp-, -S-CH2-, -
Figure imgf000016_0002
Figure imgf000016_0003
, and a bond; wherein independently for each occurrence Xa is CH or N. [0064] In some embodiments, the base salt is an alkali metal salt. For example, in some embodiments, the alkali metal is selected from the group consisting of, e.g., sodium, potassium, and lithium.
[0065] In some embodiments, T is selected from the group consisting of:
Figure imgf000016_0004
[0066] In some embodiments, T may be selected from a base salt (e.g., a lithium, sodium or potassium salt) of a moiety selected from the group consisting of:
Figure imgf000017_0001
[0067] Also provided herein are compounds that may be selected from the group consisting of: sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)-3- methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide; sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetate; sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperidine-4-carboxylate; sodium 5-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)tetrazol-2-ide; sodium 2-((l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)acetate; sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoate; sodium 5-(2-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)tetrazol-2-ide; sodium (2-(l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetyl)(methylsulfonyl)amide; sodium ethyl(2-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate; sodium 2-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)-2-methylpropanoate; sodium 3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2,2-dimethylpropanoate; sodium (3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoyl)(methylsulfonyl)amide; sodium 5-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate; sodium 5- (2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)ethyl)isoxazol-3-olate; sodium 3-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoate; sodium 2-(((S)-l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate; sodium 2-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)amino)acetate; sodium 2-(((R)-l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate; sodium 2-(3- (((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)acetate; sodium 4- ((((((3R,4S,5S,6R)-5-methoxy-4-((2S,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)benzoate; sodium 4-(((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)benzoate; sodium 6-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate; sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)- 2 -methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidine-3-carboxylate; sodium 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate; sodium 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate; sodium 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate; sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)phosphonate; sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)propanoate; sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate; sodium 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate; sodium 2-(4-((((((3R,4S,5S,6R)-
5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate; sodium (3R,5R)-3,5- dihydroxy-7-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)heptanoate; sodium 3-(3- (((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoate; sodium (3R,5R)-3,5-dihydroxy-7-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)heptanoate; sodium 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro[3.3]heptan-2-yl)acetate; sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- oxopiperazin-l-yl)acetate; sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro[3.3]heptan-6-yl)oxy)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3- methylazetidin-3-yl)oxy)acetate; sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)- 2 -methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-4-carboxylate; sodium 5-((((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-3-carboxylate; sodium 6-(l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate; sodium 5-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinate; sodium 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate; sodium 3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidine-3-carboxylate; sodium 6-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate; sodium 5-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azeti din-3 -yl) picolinate; sodium l-[l-({[(3R,4S,5S,6R)-5-methoxy-4- [(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)azetidin-3-yl]-lH-imidazole-4-carboxylate; sodium 5-(3-fluoro-l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)picolinate; sodium 2-(l-((((3R,4S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-ylidene)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)thio)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfonyl)acetate; sodium 2-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)-2-azaspiro[3.3]heptane-6-carboxylate; sodium 2,2-difluoro-3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate; sodium 2-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)sulfmyl)acetate; sodium l-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2-oxo-l,2- dihydropyridine-4-carboxylate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)-2-oxo-l,2-dihydropyridine-3-carboxylate; sodium 4-[l- ({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-l,3-thiazole-2-carboxylate; sodium 3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2-oxopropanoate; sodium l-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3-carboxylate; sodium 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiiO[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)-2H-l, 2, 3-tri azole-4- carboxylate; sodium 6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridine-3 -carboxylate; sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,3-dihydro-lH- pyrrolo[3,4-c]pyridine-6-carboxylate; sodium l-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylate; sodium 3-fluoro-l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidine-3-carboxylate; sodium 2-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)-5-oxa-2-azaspiro[3.4]octane-6-carboxylate ; and sodium 2-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)ethane-l-sulfonate; and a pharmaceutically acceptable salt or stereoisomer thereof.
II. Methods
[0068] In some embodiments, the disclosure provides a method of treating or mitigating a liver, intestine and/or kidney disorder in a patient in need thereof, comprising administering an effective amount of a disclosed compound to the patient. In some embodiments, for example, the liver disorder is one or more of, e.g.: non-alcoholic steatohepatitis, alcoholic
steatohepatitis, hepatocellular carcinoma, liver cirrhosis, and hepatitis B. In other
embodiments, the kidney disorder may be one or more of chronic kidney disease, glomerular disease such as iGA nephropathy, lupus nephritis, polycystic kidney disease
[0069] Contemplated patients include not only humans, but other animals such as companion animals (e.g., dogs, cats).
[0070] Methods of treating hepatocellular carcinoma in a patient in need thereof are also contemplated herein, comprising adminsetering an effective amount of a disclosed compound to the patient. In some embodiments, the patient suffers from type 2 diabetes, hepatitis B, cirrhosis, alpha-l antitrypsin deficiency, and/or is obese.
[0071] For example, provided herein are methods of treating, or preventing the development of, hepatic preneoplastic lesions in patient in need thereof, comprising administering an effective amount of a disclosed compound to the patient. In some embodiments, the patient is at risk for but not yet suffering from NASH. In some embodiments, the patient has at least one of, for example: minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation, and/or balloon degeneration. For example, in some embodiments, the patient is diabetic (e.g. type 2 diabetes) or in pre-diabetic condition. The patient may or may not also suffer from obesity. In some embodiments, the patient has a risk of developing NASH (and/or alcoholic steatohepatitis or cirrhosis), but is not suffering from NASH and/or alcoholic steatohepatitis or cirrhosis, or in other embodiments, the patient has developed NASH, alcoholic steatohepatitis or cirrhosis, but has not suffered from hepacellular carcinoma. For example, a patient may have minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation and/or balloon degeneration. Compounds may be administered in an amount insufficient to modulate or suppress angiogenesis in said patient. In some embodiments, the patient suffers from type 2 diabetes and/or is obese. Contemplated patients may optionally be currently administering or have been administered amiodarone, antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
[0072] A patient suscepted of suffering from NASH or one or more of NASH symptoms may be scored using a NASH scoring. A NASH scored is the combined score of fibrosis (0-4), steatosis (0-3), lobular inflammation (0-3), and ballooning degeneration (0-2) as illustrated in table A (see Design and validation of a histological scoring system for nonalcoholic fatty liver disease, Kleiner et ak, Hepatology 41; 2005).
Table A
Figure imgf000022_0001
Figure imgf000023_0002
Figure imgf000023_0001
[0073] Without being bound to any particular theory, contemplated compounds disclosed herein may beneficially affect hepatic glucose uptake, insulin action, steatosis, inflammation and/or cell dysplasia.
[0074] Disclosed compounds may be administered to patients (animals and/or humans) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. It will be appreciated that the dose required for use in any particular application will vary from patient to patient, not only with the particular compound or composition selected, but also with the route of administration, the nature of the condition being treated, the age and condition of the patient, concurrent medication or special diets then being followed by the patient, and other factors which those skilled in the art will recognize, with the appropriate dosage ultimately being at the discretion of the attendant physician. For treating clinical conditions and diseases noted above, a compound of this invention may be administered orally, subcutaneously, topically, parenterally, by inhalation spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. Parenteral
administration may include subcutaneous injections, intravenous or intramuscular injections or infusion techniques.
[0075] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
[0076] In general, a suitable daily dose of a disclosed compound will be that amount of the compound which is the lowest dose effective to safely produce the maximal therapeutic effect. Such an effective dose will generally depend upon the factors described above. For example, a disclosed compound can be administered at about 0.01 mg/kg to about 200 mg/kg, at about 0.1 mg/kg to about 100 mg/kg, or at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents), the effective amount may be less than when the agent is used alone. Contemplated methods may include administration of a disclosed compounds, for example, hourly, twice hourly, every three to four hours, daily, twice daily, 1, 2, 3 or 4 times a week, every three to four days, every week, or once every two weeks depending on half-life and clearance rate of the particular compound.
[0077] Treatment can be continued for as long or as short a period as desired. The compositions may be administered on a regimen of, for example, one to four or more times per day. A suitable treatment period can be, for example, at least about one week, at least about two weeks, at least about one month, at least about six months, at least about 1 year, or indefinitely. A treatment period can terminate when a desired result disclosed herein, e.g., treating or mitigating a liver disorder disclosed herein, is achieved. A treatment regimen can include a corrective phase, during which a dose sufficient to treat or mitigate a liver disorder disclosed herein, and can be followed by a maintenance phase, during which a e.g. a lower dose sufficient to prevent recurrence of the liver disorder is administered. A suitable maintenance dose is likely to be found in the lower parts of the dose ranges provided herein, but corrective and maintenance doses can readily be established for individual subjects by those of skill in the art without undue experimentation, based on the disclosure herein.
[0078] Contemplated methods may also include administering a disclosed compound in combination with one or more other agents (and/or in combination with weight loss surgery and/or weight loss therapy). In some embodiments, other agents may be selected from an antidiabetic medication (e.g., metformin, a modulator of glucagon-like peptide -1 receptor such as exenatide, liraglutide, lixisenatide, albiglutide and dulaglutide; and/or a thiazolidinedione such as rosiglitazone, pioglitazone, troglitazone, and/or netoglitazone). In some embodiments, other agents may include a C-C chemokine receptor type 5/type 2 (CCR5/CCR2) modulator, such as but not limited to cenicriviroc. In some embodiments, the other agent may be fibroblast growth factor 21 (FGF21) modulator, such as but not limited to BMS-986036, an
investigational pegylated analogue of FGF21. In some embodiments, the other agent is a peroxisome proliferation-activated receptor (PPAR) agonist, and/or may include agents such as elafibranor, clofibrate, gemfibrozil, ciprofibrate, bezafibrate, and/or fenofibrate,
thiazolidinediones, NSAIDs, indoles, aleglitazar, muraglitazar and tesaglitazar. In some embodiments, the other agent is a obeticholic acid, aramchol, emricasan, simtuzumab, GS-4997 (Gilead), GR-MD-02 (Galectin Therapeutics), Tipelukast, ARI-3037MO (Arisaph
Pharmaceuticals), and/or volixibat. In some embodiments, a contemplated other agent is selected from an antioxidant (e.g., vitamin E, selenium or betaine), a blood pressure lowering medication (e.g., isinopril, losartan, metoprolol, amlodipine, aliskiren), and a cholesterol lowering medication (e.g., atorvastatin, rosuvastatin, or nicotinic acid). In other embodiments, a contemplated other agent is selected from amiodarone, antiviral drugs such as nucleoside analogues, aspirin, corticosteroids, methotrexate, tamoxifen, and tetracycline.
[0079] For example, a patient with NASH may also have other conditions, such as diabetes, high blood pressure or high cholesterol levels. Thus, contemplated herein are disclosed compounds in combination with at least one other agent that may be used to treat one or more of these conditions. For example, the second active agent can be, for example, an antidiabetic medication (e.g., metformin, and/or a thiazolidinedione such as rosiglitazone, pioglitazone, troglitazone, and/or netoglitazone), an antioxidant (e.g., vitamin E, selenium or betaine), a blood pressure lowering medication (e.g., lisinopril, losartan, metoprolol, amlodipine, aliskiren), or a cholesterol lowering medication (e.g., atorvastatin, rosuvastatin, or nicotinic acid), or another agent such as vitamin E. In other embodiments, contemplated methods further include administering to the patient an active agent such as one or more of: amiodarone, antiviral drugs such as nucleoside analogues, aspirin, NSAIDS, corticosteroids, methotrexate, tamoxifen, or tetracycline.
III. Pharmaceutical Compositions and Kits
[0080] The disclosure also provides pharmaceutical compositions comprising compounds as disclosed herein formulated together with a pharmaceutically acceptable carrier. In particular, the present disclosure provides pharmaceutical compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers. These formulations include those suitable for oral, rectal, topical, buccal, parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) rectal, vaginal, or aerosol
administration, although the most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used. For example, disclosed compositions may be formulated as a unit dose, and/or may be formulated for oral or subcutaneous administration.
[0081] Exemplary pharmaceutical compositions may be used in the form of a pharmaceutical preparation, for example, in solid, semisolid or liquid form, which contains one or more of disclosed compounds, as an active ingredient, in admixture with an organic or inorganic carrier or excipient suitable for external, enteral or parenteral applications. The active ingredient may be compounded, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use. The active object compound is included in the pharmaceutical composition in an amount sufficient to produce the desired effect upon the process or condition of the disease. [0082] For preparing solid compositions such as tablets, the principal active ingredient may be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g, water, to form a solid preformulation
composition containing a homogeneous mixture of a disclosed compound, or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
[0083] In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the subject composition is mixed with one or more
pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4)
disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
[0084] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the subject composition moistened with an inert liquid diluent. Tablets, and other solid dosage forms, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. [0085] Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the subject composition, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, cyclodextrins and mixtures thereof.
[0086] Suspensions, in addition to the subject composition, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
[0087] Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing a subject composition with one or more suitable non irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the body cavity and release the active agent.
[0088] Dosage forms for transdermal administration of a subject composition include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
[0089] The ointments, pastes, creams and gels may contain, in addition to a subject composition, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
[0090] Powders and sprays may contain, in addition to a subject composition, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays may additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. [0091] Disclosed compositions and compounds may alternatively be administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A non-aqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers may be used because they minimize exposing the agent to shear, which may result in degradation of the compounds contained in the subject compositions. Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of a subject composition together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular subject composition, but typically include non-ionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.
[0092] Pharmaceutical compositions contemplated herein may be suitable for parenteral administration comprise a subject composition in combination with one or more
pharmaceutically-acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
[0093] Examples of suitable aqueous and non-aqueous carriers which may be employed in contemplated pharmaceutical compositions include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate and cyclodextrins. Proper fluidity may be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
EXAMPLES
[0094] The compounds described herein can be prepared in a number of ways based on the teachings contained herein and synthetic procedures known in the art. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated. The starting materials for the examples are either commercially available or are readily prepared by standard methods from known materials.
[0095] At least some of the compounds identified as“Intermediates” herein are contemplated as compounds of the invention.
[0096] All reagents were purchased from commercial suppliers (Sigma-Aldrich, AlfaAesar, Acros Organics, etc.) and used without further purification unless otherwise stated. THF was continuously refluxed and freshly distilled from sodium and benzophenone under nitrogen, dichloromethane was continuously refluxed and freshly distilled from CaH2 under nitrogen.
[0097] Reactions were monitored by LCMS and/or TLC on silica gel 60 HSGF254 percolated plates (0.15-0.2 mm Si02) and visualized using UV light (254 nm or 365 nm) and/or staining with phosphomolybdic acid ethanol solution (10 g in 100 mL ethanol) and subsequent heating.
[0098] Analytical LCMS were typically performed on SHIMADZU LCMS-2010EV
(ChromolithSpeedROD, RP-l8e, 50x4.6 mm, mobile phase: Solvent A: CH3CN/H20
/HCOOH=l 0/90/0.05, Solvent B: CH3CN/H20 /HCOOH=90/l0/0.05, 0.8min@ 10% B, 2.7min gradient (10-95% B), then 0.8min@95%B, Flow rate: 3mL/min, temperature: 40°C).
[0099] Preparative HPLC were typically performed either by Method A: SHIMADZU LC- 8A (Column: YMC Pack ODS-A (l50*30mm, lOpm)) or Method B: LC-6AD (Column:
Shim=Pack PREP-ODS-H (250*20mm, lOpm)) with UV detection. Both methodswere controlled by LC solution Chemstation software using H20 (containing 0.1% HCOOH) and MeOH (or MeCN) as mobile phase at the indicated flow rate.
[0100] Analytical HPLC were typically performed on a SHIMADZU LC-2010A
(ChromolithSpeedROD, RP-l8e, 50x4.6 mm, mobile phase: Solvent A: CH3CN/H20
/HCOOH=l 0/90/0.05, Solvent B: CH3CN/H20 /HCOOH=90/l0/0.05, 0.8min@ 10% B, 2.7min gradient (10-95% B), then 0.8min@95%B, Flow rate: 3mL/min, temperature: 40°C).
[0101] Chiral HPLC were performed on a SHIMADZU LC-2010A (Chiral column, mobile phase: Solvent A: hexane (optionally containing 0.1% diethylamine), Solvent B: Ethanol or Isopropanol; Flow rate: 0.8 mL/min, temperature: 30°C).
[0102] 1H spectra were recorded on BrukerAvance II 400MHz NMR. Chemical shifts (d) were reported in ppm relative to tetramethylsilane (d = 0.000 ppm), and the spectra were calibrated to the residual solvent signal of chloroform (d = 7.26), Dimethyl sulfoxide (d = 2.50), or methanol (d = 3.30). Data for 1H NMR spectra were reported as follows: chemical shift (multiplicity, number of hydrogens). Abbreviations were described as follows: s (singlet), d (doublet), t (triplet), q (quartet), quint (quintet), m (multiple), br (broad).
Abbreviations
CDI = l,l '-carbonyl diimidazole
DBU = l,8-diazabicyclo(5.4.0)undec-7-ene
DCM = dichloromethane
DIPEA = diisopropylethylamine
DMAP = (4-dimethylamino)pyridine
DMSO = dimethyl sulfoxide
DMF = dimethyl formamide
EA = ethyl acetate
FA = formic acid
LDA = lithium diisopropylamine
PE = petroleum ether
r.t. = room temperature
TBTU = 2-(lH -Benzoin azole- 1 -yl)- 1,1, 3 ,3 -tetram ethyl aminium tetrailuoroborate
TEA = trithylamine
TFA = trifluoroacetic acid
THF = tetrahydrofuran
Example 1: Preparation of sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)-3-methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide
Figure imgf000031_0001
Step 1. Preparation of (S)-tert-butyl (l-amino-4-methyl-l-oxopentan-3-yl)carbamate
1 . BOC20, Py
dioxane
Figure imgf000031_0002
[0103] To a mixture of (S)-3-((tert-butoxycarbonyl)amino)-4-methylpentanoic acid (2.02 g, 8.73 mmol) in l,4-dioxane (20 mL) was added di-tert-butyl dicarbonate (5.03 g, 23.04 mmol), followed by dropwise addition of pyridine (0.43 mL, 5.38 mmol) at 0°C. The mixture was stirred at 0°C for 10 min and ammonium bicarbonate (898 mg, 11.35 mmol) was added. After addition of the ammonium bicarbonate, the resulting mixture was stirred at room temperature overnight. The mixture was then diluted with ethyl acetate and washed with water and brine, dried over Na2S04 and concentrated to give (S)-tert-butyl ( l-amino-4-m ethyl- 1 -ox opentan-3- yl)carbamate (2.02 g) as white solid. LC-MS (ESI) found: 175 [M+l-56]+.
Step 2. Preparation of (S)-tert-butyl (l-cyano-3-methylbutan-2-yl)carbamate
Figure imgf000031_0003
[0104] To a solution of (S)-tert-butyl (l-amino-4-methyl-l-oxopentan-3-yl)carbamate (1.72 g, 7.47 mmol) in DMF (15 mL) was added 2,4,6-trichloro-l,3,5-triazine (2.34 g, 12.68 mmol) at 0°C under N2 atmosphere. The resulting mixture was stirred at room temperature for 3 hr. The mixture was then diluted with ethyl acetate and washed with aqueous lithium chloride solution, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (silica gel, 2 to 5 % ethyl acetate in petroleum ether) to give (S)-tert- butyl (l-cyano-3-methylbutan-2-yl)carbamate (1.02 g) as white solid. LC-MS (ESI) found: 157 [M+l-56]+. 1HNMR (400 MHz, DMSO-i¾) S 7.04 (m, 1H), 3.57 - 3.42 (m, 1H), 2.66 (m, 1H), 2.55 - 2.51 (m, 2H), 1.69 (m, 1H), 1.40 (s, 9H), 0.83 (m, 6H).
Step 3. Preparation of (S)-tert-butyl (l-(hydroxyamino)-l-imino-4-methylpentan-3- yl)carbamate
Figure imgf000032_0001
[0105] To a mixture of (S)-tert-butyl (l-cyano-3-methylbutan-2-yl)carbamate (1.02 g, 4.80 mmol) in ethanol (6mL) was added aqueous hydroxylamine solution (1.5 mL, 24.02 mmol, 50% wt). The resulting mixture was stirred at 50°C overnight. The mixture was then
concentrated, diluted with ethyl acetate, and washed with water and brine. The organic layer was dried over Na2S04 and concentrated to give a residue, which was purified by flash chromatography (silica gel, 1 to 2% methanol in dichloromethane) to give (S)-tert-butyl (1- (hydroxyamino)-l-imino-4-methylpentan-3-yl)carbamate (1.0 g) as white solid. LC-MS (ESI) found: 190 [M+l-56]+.
Step 4. Preparation of (S)-tert-butyl (3-methyl-l-(5-oxo-2,5-dihydro-l,2,4-oxadiazol-3- yl)butan-2-yl)carbamate
Figure imgf000032_0002
[0106] To a solution of (S)-tert-butyl (l-(hydroxyamino)-l-imino-4-methylpentan-3- yl)carbamate (434 mg, 1.77 mmol) in dioxane (8 mL) was added DBU (315 mg, 1.95 mmol) and CDI (296 mg, 1.94 mmol), the mixture was heated to 80°C for 2hrs. The mixture was diluted with water and extracted with EA, and the aqueous water was acidified to pH2 with 1 N
HC1 and extracted with EA. The organic layers were washed with brine and dried over Na2S04, filtered and concentrated to provide the title compound (434 mg) as white solid. LC-MS (ESI) found: 216 [M+l-56]+.
Step 5. Preparation of (S)-3-(2-amino-3-methylbutyl)-l,2,4-oxadiazol-5(2H)-one trifluoroacetic acid salt
Figure imgf000033_0001
[0107] To a mixture of (S)-tert-butyl (3-methyl-l-(5-oxo-2,5-dihydro-l,2,4-oxadiazol-3- yl)butan-2-yl)carbamate (434 mg, 1.60 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (2.5 mL) dropwise at 0°C. The reaction was stirred at room temperature for 1 hr and concentrated under reduced pressure to give (S)-3-(2-amino-3-methylbutyl)- 1,2,4- oxadiazol-5(2H)-onetrifluoroacetic acid salt (425 mg) as yellow syrup, which was directly used in the next step without purification. LC-MS (ESI) found: 172 [M+l]+.
Step 6. Preparation of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl ((S)-3-methyl-l-(5-oxo-4,5-dihydro-l,2,4- oxadiazol-3-yl)butan-2-yl)carbamate
General Procedure for Coupling:
Figure imgf000033_0002
Intermediate 1
[0108] To a solution of (S)-3-(2-amino-3-methylbutyl)-l,2,4-oxadiazol-5(2H)-one (268 mg, 1.57 mmol) in acetonitrile (8 mL) was added N,N-Diisopropylethylamine (0.94 mL, 5.24 mmol) dropwise at 0°C. The mixture was stirred at 0°C for 10 min, and (3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl (4-nitrophenyl) carbonate (Intermediate 1, 585 mg, 1.31 mmol) was added. After addition, the reaction was stirred at room temperature overnight under N2 atmosphere. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL x 2) and 5% sodium bicarbonate solution (20 mL). The organic layer was dried over Na2S04, filtered and concentrated to give crude product. The crude product was purified by flash chromatography (silica gel, 1 to 2% MeOH in DCM) and preparative HPLC (Method A, H20 (0.1% FA)/ C¾CN) to give (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl((S)-3 -methyl- 1 -(5 -oxo-4, 5 - dihydro-l,2,4-oxadiazol-3-yl)butan-2-yl)carbamate (160 mg) as white solid. LC-MS (ESI) found: 480 [M+l]+. 1H NMR (400 MHz, CDCI3) d 10.15 (m, 1H), 5.45 (m, 1H), 5.14 (m, 1H), 4.64 (m, 1H), 3.63 (s, 2H), 3.37 (s, 3H), 2.91 (s, 1H), 2.77 - 2.49 (m, 4H), 2.08 (m, 1H), 1.99 - 1.72 (m, 6H), 1.69 (s, 3H), 1.60 (s, 3H), 1.17 (d, J= 8.0 Hz, 3H), 1.03 (s, 1H), 0.91 (m, 6H).
Step 7. Preparation of sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)-3- methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide
Figure imgf000034_0001
[0109] To a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (90 mg, 0.18 mmol) in MeCN was added dropwised 0.1 N NaOH (1.6 mL,0.16 mmol) solution at -60 °C with stirring. After
lypholization, 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)-3-methylbutyl)-5-oxo- l,2,4-oxadiazol-4-ide sodium salt (90 mg) was obtained as white solid. LC-MS (ESI) found: 480 [M+l]+. 1H NMR (400 MHz, DMSO- d) d 6.84 (d, J= 9.3 Hz, 1H), 5.18 (dd, J= 20.7, 13.3 Hz, 2H), 3.62 - 3.45 (m, 2H), 3.24 (s, 3H), 2.80 (d, J= 4.3 Hz, 1H), 2.60 - 2.50 (m, 2H), 2.24 (dd, J = 14.7, 6.2 Hz, 2H), 2.20 - 2.08 (m, 2H), 1.98 - 1.73 (m, 3H), 1.68 (s, 3H), 1.58 (s,
3H), 1.07 (s, 3H), 0.79 (t, J= 7.0 Hz, 6H).
Example 2: Preparation of sodium 2-(l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetate
Figure imgf000034_0002
Step 1 : Preparation of 2-(azetidin-3-yl)acetic acid(TFA salt)
Figure imgf000035_0001
[0110] To a solution of 2-(l-(tert-butoxycarbonyl)azetidin-3-yl)acetic acid (1.00 g, 4.64 mmol) in DCM (5 mL) was added TFA (2 mL) at 0°C. The reaction was then stirred at room temperature overnight. The reaction mixture was concentrated to give 2-(azeti din-3 -yl)acetic acid ( 1.35 g, TFA salt) as a yellow oil, which was directly used in the next step without further purification. LC -MS (ESI) found: 116 [M+l]+.
Step 2 : Preparation of 2-(l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetic acid
Figure imgf000035_0002
[0111] 2-(Azeti din-3 -yl)acetic acid (TFA salt, l.3g crude, 4.64 mmol) in CH3CN (10 mL) was coupled with Intermediate 1 (1.63 g, 3.71 mmol) by following the General Procedure for Coupling. The solvent was removed under vacuum while maintaining the temperature below 40°C, then diluted with DCM (60 mL) and washed with ammonium acetate buffer (pH~4, 15 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqueous layer was extracted with DCM (10 mL x 2). The combined organic layers were dried over Na2S04 and concentrated to give a yellow oil, which was purified by flash chromatography (silica gel, DCM:MeOH= 100:0 to 60: 1) to give the pure 2-(\-((((3Ii,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methy\-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)acetic acid (1.1 g) as light yellow syrup with HPLC purity 96%.LC-MS (ESI) found: 424 [M+l]+. 1H-NMR (400 MHz, DMSO- d6 ) d 12.25 (br, 1 H), 5.29 (s, lH),5. l9(t, J= 7.6 Hz, 1H), 3.96- 4.00(m,2H), 3.32-3.58 (m, 3 H), 3.28 (s, 3H), 2.78-2.86 (m, 2 H), 2.50-2.59 (m, 4 H), 2.15-2.19 (m, 2 H), 1.91-1.98 (m, 1 H), 1.75-1.80 (m, 6 H), 1.62 (s, 3H), 1.13 (s, 3H), 0.99 (d, J= 13.6 Hz, 1H).
Step 3 : Preparation of sodium 2-(l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetate
Figure imgf000036_0001
[0112] To a solution of 2-(l-((((3f?, S',5,S,, d/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)acetic acid (100 mg, 0.236 mmol) in MeCN (5 mL) was added dropwise 0.1 N NaOH solution (2.1 mL, 0.9 eq) at -60°C with stirring. After lypholization, 120 mg of the title compound was obtained as white solid. LC-MS (ESI) found: 446 [M+l]+. 1H-NMR (400 MHz, DMSO-i¾) d 5.28 (s, 1H), 5.20(t, J= 7.6 Hz, 1H), 3.89-3.93(m,2H),3.45-3.53 (m, 3H), 3.28 (s, 3H), 2.84 (d, J= 4.4Hz, 1H), 2.67-2.73 (m, 1H), 2.54-2.58 (m, 2H), 2.11-2.19 (m, 4H), 1.91-1.96 (m, 1H), 1.75-1.80 (m, 6H), 1.62 (s, 3H), 1.14 (s, 3H), 0.98 (d, J= 13.6 Hz, 1H).
Example 3: Preparation of sodium l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4- carboxylate
Figure imgf000036_0002
Step 1. Preparation of l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3-methylbut-
2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4-carboxylic acid
Figure imgf000037_0001
[0113] Piperidine-4-carboxylic acid (200 mg, 1.55 mmol) was coupled with Intermediate 1
(830 mg, 1.85 mmol) by following the General Procedure for Coupling. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C, then diluted with DCM (60 mL) and washed with ammonium acetate buffer (pH~4, 15 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqueous layer was extracted with DCM (10 mL x 2), dried over Na2S04 and concentrated to give a yellow oil, the resided was purified by column chromatography (silica gel, 1—10 % MeOH in DCM) and prep. HPLC (C18, 0 to 90% acetonitrile in H20 with 0.1 % formic acid) to give l-((((3f?,4ri,,5ri', df?)-5-methoxy-4-((2f?,3f?)- 2-m ethyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperidine-4-carboxylic acid (260 mg) as white solid. LC-MS (ESI) m/z (M+l): 438. 1H NMR (400 MHz, DMSO-76) d 12.28 (s, 1H), 5.36 (s, 1H), 5.19 (s, 1H), 3.87
(d, J= 13.0 Hz, 2H), 3.54 (dd, 7= 11.1, 2.6 Hz, 1H), 3.32 (s, 3H), 2.86 (d, J= 4.4 Hz, 3H),
2.58 (s, 2H), 2.42 (d, J= 10.8 Hz, 1H), 2.21 - 2.13 (m, 2H), 1.93 (dd, J= 13.5, 4.3 Hz, 1H), 1.79 (s, 4H), 1.71 (s, 4H), 1.61 (s, 3H), 1.41 (s, 2H), 1.08 (s, 3H), 1.02 (d, J= 13.5 Hz, 1H).
Step 2: Preparation of sodium l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4- carboxylate
Figure imgf000038_0001
[0114] To a solution of l-((((3f?, ,S,,5S,, 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperidine-4-carboxylic acid (122 mg, 0.27 mmol) in MeCN (10 mL) was added dropwise a 0.1 N NaOH solution (2.5 mL, 0.24 mmol) at -6°C with stirring, followed by lyophilization to give sodium 1 -((((3R,-fS,5S, 6R)- 5-methoxy-4-((2ri, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperidine-4-carboxylate (106 mg) as white solid. LC-MS (ESI) m/z (M+l): 438. 1H MR (400 MHz, DMSO-76) d 5.34 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 3.77 (s, 2H), 3.55 - 3.50 (m, 1H), 3.32 (s, 3H), 2.85 (d, J= 4.4 Hz, 3H), 2.58 (d, J= 10.5, 5.2 Hz, 2H), 2.21 - 2.11 (m, 2H), 1.92 (d, 7= 6.3 Hz, 2H), 1.78 (d, 7= 11.0 Hz, 2H), 1.66 (d, 7= 38.7 Hz, 9H),
1.36 (s, 2H), 1.08 (s, 3H), 1.02 (d, J= 13.6 Hz, 1H).
Example 4: Preparation of sodium 5-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)tetrazol-2-ide
Figure imgf000039_0001
Step 1: Preparation of tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate
B i oc -OEt B
O i oc
Figure imgf000039_0002
[0115] To a solution of NaH (0.8 g, 20 mmol) in THF (90 mL) at 0°C was added diethyl (cyanomethyl)phosphonate (3.45 g, 19.5 mmol). The reaction was stirred at 0°C for 45 min. Then asolution of tert-butyl 3-oxoazetidine-l-carboxylate (.9 g, 16.9 mmol) in THF (20 mL) was added at rt. After addition, the reaction was stirred at r.t. overnight. The reaction was then slowly poured into ice-water and extracted with EA twice. The organic layer was dried over Na2S04 and concentrated to give a residue, which was purified by flash chromatography (silica gel, 10 to -20% EA in PE) to give tert-butyl 3-(cyanomethylene)azetidine-l-carboxylate as a white solid (2 g). LC-MS (ESI) found: 139 [M+l-56]+.
Step 2: Preparation of tert-butyl 3-(cyanomethyl)azetidine-l-carboxylate
Boc Boc
Figure imgf000039_0003
[0116] To a solution of /cvV-butyl 3-(cyanomethylene)azetidine-l-carboxylate (1.9 g, 9.79 mmol) in methanol (30 mL) was added Pd/C (0.2 g). The reaction mixture was de-gassed 3 times with N2 then stirred under H2 atmosphere at r.t. overnight. The reaction mixture was filtered, and the filtrate was concentrated to give /cvv-butyl 3-(cyanomethyl)azetidine-l- carboxylate as a white solid (1.5 g), which was directly used in the next step without purification. LC-MS (ESI) found: 141 [M+l-56]+.
Step 3: Preparation of tert-butyl 3-((2H-tetrazol-5-yl)methyl)azetidine-l-carboxylate
Boc
i
Figure imgf000040_0001
[0117] To a solution of tert- butyl 3 -(cyanomethyl)azetidine-l -carboxylate (1.2 g, 6.12 mmol ) in DMF (15 mL) was added NaN3 (0.47 g, 7.34 mmol). The reaction mixture was stirred at l20°C overnight. The reaction was diluted by water and extracted with DCM for 3 times. The dichloromethane solution was dried over Na2S04, filtered and concentrated to give crude product, which was purified by prep-HPLC (Method B: 0.l%NH4OH) to give tert-butyl 3- ((2H-tetrazol-5-yl)methyl)azetidine-l -carboxylate as a yellow solid (280 mg). LC-MS (ESI) found: 184 [M+l-56]+.
Step 4: Preparation of 5-(azetidin-3-ylmethyl)-2H-tetrazole
Figure imgf000040_0002
[0118] To a solution of tert- butyl 3-((2H-tetrazol-5-yl)methyl)azetidine-l-carboxylate (280 mg, 1.1 mmol) in DCM (3 mL) was added TFA (400 mg, 3.51 mmol). The reaction was stirred at r.t. overnight. The reaction was concentrated to give 5-(azetidin-3-ylmethyl)-2H-tetrazole as a yellow oil (430 mg, TFA salt), which was directly used in the next step without any purification.
Step 5: Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl lH-imidazole-l-carboxylate
Figure imgf000041_0001
Intermediate 2
[0119] To a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-ol (2 g, 7.08 mmol) in DCM (30 mL) was added CDI (3.4 g, 20.97 mmol) at 0 °C. After addition, the reaction mixture was stirred at 0°C for 2 to 3 h. The reaction was monitored by HPLC. Once the starting material was completely consumed, the mixture reaction was diluted with DCM (20 mL) and washed with water (30 mL x 3). The organic layers were combined and dried over Na2S04. After filtration, the filtrate was concentrated under reduced pressure to afford the title compound (2.3 g) as oil, which was confirmed by 1HNMR. 1HNMR (400 MHz, CDCl3) ό 8.12 (s, 1 H), 7.38 (s, 1 H), 7.05 (s, 1 H), 5.81 (s, 1 H), 5.18 (t, J = 6 Hz,l H), 3.74 (d, J = 16 Hz, 1 H), 3.49 (s, 3 H), 3.01 (d, J = 4 Hz, 1
H), 2.58-2.60 (m, 2 H), 2.26-2.35 (m, 1 H), 2.07-2.13 (m, 5 H), 1.90 (d, J = 12 Hz, 1 H), 1.73
(s, 3 H), 1.64 (s, 3 H), 1.10-1.21 (m, 4 H).
Step 6: Preparation of (3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((2H-tetrazol-5-yl)methyl)azetidine-l- carboxylate
Figure imgf000041_0002
[0120] To a solution of 5-(azetidin-3-ylmethyl)-2H-tetrazole (430 mg, TFA salt) in DCM (5 mL) was added DIPEA (750 mg, 5.85 mmol) at 0°C. A solution of (3R, -iS,5S, ri/i)-5-methoxy-4- ((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 1H- imidazole-l -carboxylate (350 mg, 0.936 mmol) in DCM (3 mL) was added to the reaction mixture. The reaction was stirred at r.t. overnight. The reaction mixture was washed with ammonium acetate buffer (pH 4.0, 5 mL x2). The dichloromethane layer was separated and dried over Na2S04. After filtration, the filtrate was concentrated to give crude product, which was purified by prep. HPLC (Method B: 0. l%NH4OH) to give (3R, -fS, 5S, ri/i)-5-methoxy-4- ((2f?,3f?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((2H- tetrazol-5-yl)methyl)azetidine-l-carboxylate as a yellow oil (45 mg). LC-MS (ESI) found: 448 [M+l]+.
Step 7: Preparation of Sodium 5-((l-(((( /?, S,5A,6/?)-5-methoxy-4-((2/?, /?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)tetrazol-2-ide
Figure imgf000042_0001
[0121] To a stirring solution of (3f?,4ri,,5ri,, 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl 3-((2H-tetrazol-5- yl)methyl)azetidine-l-carboxylate (45 mg, 0.10 mmol) in MeCN (0.5 mL) was added 0.1M NaOH (0.9 mL, 0.09 mmol) at -78°C. The reaction was lyophilized to give sodium 5-((l- ((((3R, 4S, 5S, 6R)-5-methoxy-4-((2R, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)tetrazol-2-ide as yellow oil (45 mg). LC-MS (ESI) found: 448 [M-23+l]+. 1H NMR (400 MHz, DMSO-i¾) d 5.26 (s, 1 H), 5.17 (s, 1 H), 3.90 (s, 2 H), 3.59 (s, 1 H), 3.49 (d, J= 9.6 Hz, 2 H), 3.26 (s, 3 H), 3.21 (s, 1 H), 3.10 (s, 1 H), 2.99 - 2.89 (m, 1 H), 2.88 - 2.76 (m, 2 H), 2.57 - 2.52 (m, 1 H), 2.40 (dd, J = 14.1, 7.0 Hz, 1 H), 2.11 (dd, = 14.9, 7.2 Hz, 2 H), 2.01 - 1.86 (m, 1 H), 1.78 - 1.66 (m, 4 H), 1.65 - 1.53 (m, 4 H), 1.35 - 1.15 (m, 5 H), 1.02 (d, J= 22.2 Hz, 2 H), 0.91 (dd, = 11.5, 5.6 Hz, 5 H), 0.82 (d, J= 6.8 Hz, 1 H).
Example 5: Preparation of sodium 2-((l-(((( /?, S,5A,6/?)-5-methoxy-4-((2/?, /?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)acetate N
Figure imgf000043_0003
Step 1: Preparation of 2-(azetidin-3-yloxy)acetic acid
Figure imgf000043_0001
[0122] To a solution of 2-((l-(/er/-butoxycarbonyl)azeti din-3 -yl)oxy)acetic acid (500 mg, 2.16 mmol) in DCM (5 mL) was added TFA (2 mL). The mixture was stirred at r.t. for 2hr. The reaction mixture was concentrated and the residue was used in the next step without further purification.
Step 2: Preparation of 2-((l-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetic acid
Figure imgf000043_0002
[0123] 2-(Azeti din-3 -yloxy)acetic acid (200 mg, 0.87 mmol) was coupled with Intermediate 1 (469 mg, 1.05 mmol) by following the General Procedure for Coupling. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. Then diluted with DCM (60 mL) and washed with ammonium acetate buffer (pH~4, 15 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqueous layer was extracted with DCM (10 mL x 2). The combined organic layers were washed with aq. NaHC03 solution (15 mL x 2, 5% w/w), dried over Na2S04 and concentrated to give yellow oil. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep. HPLC (Cl 8, 0 to ~90 % acetonitrile in H20 with 0.1 % formic acid) to give 2-((l-((((3i?,47,57, d ?)-5-methoxy-4- ((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)acetic acid (180 mg) as white solid. LC-MS (ESI) m/z (M+l): 440.1H NMR (400 MHz, DMSO) d 5.30 (s, 1H), 5.19 (s, 1H), 4.36 (d, J= 4.0 Hz, 1H), 4.05 (d, J= 8.9 Hz, 2H), 4.00 (s, 2H), 3.81 - 3.70 (m, 2H), 3.52 (dd, 7= 11.0, 2.4 Hz, 1H), 2.85 (d, 7= 4.3 Hz, 1H), 2.59 - 2.52 (m, 2H), 2. l7 (s, 2H), 1.92 (dd, 7 = 13.2, 4.4 Hz, 1H), 1.75 (d, J = 11.0 Hz, 3H), 1.71 (s, 4H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, 7= 13.6 Hz, 1H).
Step 3: Preparation of sodium 2-((l-((((3/?,7V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetate
Figure imgf000044_0001
[0124] To a solution of 2-(( l -{{{{3R,4S,5S,6R)- -mei\\oxy- -{{2R, 3//)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetic acid (120 mg, 0.27 mmol) in MeCN (10 ml) was added dropwise 0.1 N NaOH solution (2.5 ml, 0.24 mmol) at -60°C with stirring. Lyophilization gave 2-(( 1 -((((3R, -fS,5S, 6R)- 5-methoxy-4-((2i?,5i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)oxy)acetate sodium salt (94 mg) as white solid. LC-MS (ESI) m/z (M+l): 440. 1H NMR (400 MHz, DMSO-76) d 5.29 (s, 1H), 5.19 (s, 1H), 4.36 (s, 1H),
4.03 - 3.92 (m, 2H), 3.74 (s, 2H), 3.54 - 3.43 (m, 3H), 2.85 (d, J = 4.3 Hz, 1H), 2.56 (d, J = 4.1 Hz, 2H), 2.17 (d, J = 7.3 Hz, 2H), 1.93 (s, 1H), 1.75 (d, J = 10.9 Hz, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, J = 13.3 Hz, 1H).
Example 6: Preparation of sodium 3-(l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoate
Figure imgf000045_0002
Step 1: Preparation of 3-(l-(terf-butoxycarbonyl)azetidin-3-yl)propanoic acid
Boc
Figure imgf000045_0001
[0125] To a solution of /cvV-butyl 3-(2-cyanoethyl)azetidine-l-carboxylate (1.0 g, 4.75 mmol) in THF (10 mL) was added aqueous lithium hydroxide solution (7.1 mL, 7.1 mmol) at 0°C. After addition, the reaction was stirred at room temperature for 16 hr. The mixture was washed with ether and the aqueous layer was acidified to pH = 3~4 by adding 1M hydrochloride acid. The mixture was extracted with chloroform and isopropyl alcohol (3 / 1) three times. The combined organic layers were dried over Na2S04, filtered and concentrated to give 3-(l-(tert- butoxycarbonyl)azetidin-3-yl)propanoic acid (0.9 g) as white solid.
Step 2 : Preparation of 3-(azetidin-3-yl)propanoic acid (TFA salt)
Figure imgf000045_0003
[0126] To a solution of 3-(l-(/er/-butoxycarbonyl)azetidin-3-yl)propanoic acid (0.9 g, 3.92 mmol) in DCM (5 mL) was added TFA (2 mL) at 0°C. The reaction was then stirred at room temperature overnight. The reaction mixture was concentrated to give 3-(azetidin-3- yl)propanoic acid (1.3 g, TFA salt) as yellow oil, which was used for the next step reaction directly without any purification. Step 3: Preparation of 3-(l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoic acid
Figure imgf000046_0001
[0127] 3-(Azetidin-3-yl)propanoic acid (TFA salt, 260 mg crude, 0.78mmol) was coupled with Intermediate 1 (0.28 g, 0.62 mmol) by following the General Procedure for Coupling. The solvent was removed under vacuum below 40°C, then diluted with DCM (10 mL) and washed with ammonium acetate buffer (pH~4, 5 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqueous layers were extracted with DCM (10 mL x 2). The combined organic layers was dried over Na2S04 and concentrated to give a yellow oil, which was purified by flash chromatography (silica gel, 0 to -1.5% MeOH in DCM) to give the pure 3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoic acid (l30mg) as light yellow syrup. Step 4: Preparation of sodium 3-(l-((((J ?, A,5A,6/?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoate
Figure imgf000046_0002
[0128] To a solution of 3-(l-((((3i?,4S,,5<S,,d/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoic acid (60 mg, 0.137 mmol) in MeCN (3 mL) was added dropwise of the 0.1 N NaOH solution (1.2 mL, 0.9 eq) at -60°C with stirring. After lypholization, 62 mg of the title compound was obtained as yellowish solid. LC-MS (ESI) found: 438,460,461 [M+l]+. 1H- NMR (400 MHz, DMSO- d6) d 5.28 (s, 1H), 5.20(t, J= 7.6 Hz, 1H), 3.89-3.93(m,2H), 3.45- 3.53 (m, 3H), 3.28 (s, 3H), 2.84 (d, J= 4.4Hz, 1H), 2.67-2.73 (m, 1H), 2.54-2.58 (m, 2H), 2.11- 2.19 (m, 4H), 1.91-1.96 (m, 1H), 1.75-1.80 (m, 6H), 1.62 (s, 3H), 1.14 (s, 3H), 0.98 (d, J= 13.6 Hz, 1H).
Example 7: Preparation of sodium 5-(2-(l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,.?/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)tetrazol-2-ide
Figure imgf000047_0001
Step 1: Preparation of terf-butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate Boc
Figure imgf000047_0002
[0129] To a solution of 2-(l-(tert-butoxycarbonyl)azetidin-3-yl)acetic acid (2.5 g, l l .63mmol ) in THF (anhydrous, 20 mL) was added a solution of BH3-THF (31.3 mL, 3 l .3mmol, 1M in THF) dropwise at -20°C for 30 min. The resulting mixture was stirred at r.t. overnight. The reaction mixture was cooled to 0-5°C. MeOH (lOmL) was added dropwise to the mixture while controlling the temperature below l0°C. The mixture was stirred at r.t. for 20 min and then concentrated under vacuum to give a residue, which was purified by flash chromatography (silica gel, 0 to ~2% MeOH in DCM) to give tert- butyl 3 -(2-hydroxy ethyl)azeti dine- 1- carboxylate (2.15 g) as colorless oil. LC-MS: m/z=l46 (M+H-56)+.
Step 2: Preparation of tert- butyl 3-(2-(tosyloxy)ethyl)azetidine-l-carboxylate Boc
Figure imgf000048_0001
0 °C to r.t.
[0130] To a solution of /cvV-butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate (1.8 g, 8.95 mmol) in DCM (15 mL) was added triethylamine (3.71 mL, 26.8 mmol) at 0°C, followed by dropwise addition of a solution of 4-methylbenzene-l-sulfonyl chloride (2.2 g, 11.61 mmol) in DCM (10 mL) over 40 min. The resulting mixture was stirred at 25°C overnight. The mixture was washed with water (10 mL), brine (10 mL), dried over anhydrous Na2S04 and
concentrated. The residue was purified by flash chromatography (silica gel, 0 to -1.5% MeOH in DCM) to provide /cvV-butyl 3-(2-(tosyloxy)ethyl)azetidine-l-carboxylate (1.7 g) as pale yellow oil. LC-MS:m/z=300 (M+H-56)+.
Step 3: Preparation of tert- butyl 3-(2-cyanoethyl)azetidine-l-carboxylate Boc
Figure imgf000048_0002
[0131] To a solution of /cvV-butyl 3-(2-(tosyloxyethyl)azetidine-l-carboxylate (1.7 g, 4.78 mmol) in DMSO (10 mL) was added NaCN (0.65 g, 7. l7mmol). The resulting mixture was stirred at l00°C overnight under N2 atmosphere. The mixture was diluted with water (20 mL) and extracted with ethyl acetate (30 mL x 2). The organic layers were combined, dried over anhydrous Na2S04 and concentrated to give crude product, which was purified by flash chromatography (silica gel, 0 to ~2% MeOH in DCM) to give tert- butyl 3-(2- cyanoethyl)azetidine-l-carboxylate (0.85 g) as yellow oil.LC-MS: m/z=2l l (M+H)+.
Step 4: Preparation of tert- butyl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate
Figure imgf000048_0003
[0132] To a solution of /cvV-butyl 3-(2-cyanoethyl)azetidine-l-carboxylate (0.5 g, 2.38mmol) in DMF (5 mL) was added tri -//-butylti n azide (2.37 g, 7.l4mmol). The resulting mixture was stirred at l00°C overnight under N2 atmosphere. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL x 2). The organic layers were combined, dried over anhydrous Na2S04 and concentrated to give crude product, which was purified by prep. HPLC to give /ert-butyl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate (200 mg) as oil. LC-MS: m/z=254 (M+H)+.
Step 5: Preparation of 5-(2-(azetidin-3-yl)ethyl)-2H-tetrazole (TFA salt)
Figure imgf000049_0001
[0133] To a solution of /cvV-butyl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate (200 mg, 0.79 mmol) in DCM (2 mL) was added TFA (1 mL) at 0°C. The reaction was then stirred at room temperature overnight. The reaction mixture was concentrated to give 5-(2-(azetidin-3- yl)ethyl)-2H-tetrazole (0.32 g, TFA salt) as yellow oil, which was directly used in the next step without any purification. LC-MS (ESI) found: 154 [M+l]+. Step 6: (J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(2H-tetrazol-5-yl)ethyl)azetidine-l-carboxylate
Figure imgf000049_0002
[0134] 5-(2-(Azetidin-3-yl)ethyl)-2H-tetrazole (TFA salt, 0.32g crude, 0.79mmol) was coupled with Intermediate 1 (0.28 g, 0.63 mmol) by following the General Procedure for Coupling. The solvent was removed under vacuum while keeping the temperature below 40°C, then diluted with DCM (10 mL) and washed with ammonium acetate buffer (pH~4, 5 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqueous layers were extracted with DCM (10 mL x 2). The combined organic layers were washed with aq. NaHC03 solution (15 mL x 2, 5% w/w), dried over Na2S04 and concentrated to give a yellow oil, which was purified by prep. HPLC (Method B: 0.l%NH4OH) to give (3R, -fS,5S, 6R)-5-methoxy-4-((2R, 3/f)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl 3-(2-(2H-tetrazol-5- yl)ethyl)azetidine-l-carboxylate (50 mg) as light yellow syrup. LC-MS (ESI) found: 462
[M+l]+. Step 7: Preparation of sodium 5-(2-(l-((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)tetrazol-2-ide
Figure imgf000050_0001
[0135] To a stirring solution of (3f?, ri',5ri', 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl 3-(2-(2H-tetrazol-5- yl)ethyl)azetidine-l-carboxylate (50 mg, 0.1 lmmol) in MeCN (2mL) was added dropwise a solution of 0.1 M NaOH (1 mL, 0.9 eq) at -70°C. After lypholization, 55 mg of the title compound was obtained as yellowish solid. LC-MS (ESI) found: 484 [M+Na],462 [M+l]+. 1H- NMR (400 MHz, DMSO-i¾) d 5.47 (s, 1H), 5. l2(m, 1H), 3.97(m,2H), 3.60-3.62 (m, 3 H), 3.37 (s, 3 H), 2.91-2.95 (m,3H), 2.51-2.55 (m, 3H), 2.11-2.14 (m, 1H), 1.78-1.88 (m, 7 H), 1.67 (s, 3H), 1.59 (s, 3H), 1.18 (s, 3H), 1.00 (d, 7= 12.4Hz, 1H).
Example 8: Preparation of sodium ( 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2 - methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetyl)(methylsulfonyl)amide
Figure imgf000051_0001
Step 1: Preparation of tert- butyl 3-formylazetidine-l-carboxylate
Figure imgf000051_0003
[0136] To a solution of 2-(l-(/er/-butoxycarbonyl)azetidin-3-yl)acetic acid (2.0 g, 9.3 mmol) in DCM (20 mL) was added methanesulfonamide (1.06 g, 11.1 mmol), TBTU (4.47 g, 13.95 mmol) and DIPEA (2.4 g, 18.6 mmol). The mixture reaction was stirred at r.t. overnight. The mixture was extracted with 0.1N HC1, then the acidic water was extracted with DCM and the dichloromethane solution was dried over Na2S04, filtered and concentratedto give crude product, which was purified by flash chromatography (silica gel, 0 to ~2% MeOH in DCM) to give /c/7-butyl 3-formylazetidine-l-carboxylate as yellow oil (1.3 g). LC-MS (ESI) found: 237 [M+l-56]+.
Step 2: Preparation of 2-(azetidin-3-yl)-N-(methylsulfonyl)acetamide
Figure imgf000051_0002
[0137] To a solution of /c/7-butyl 3-formylazetidine-l-carboxylate (1.3 g, 4.5 mmol) in DCM
(10 mL) was added TFA (2.53 g, 22.2 mmol). The reaction was stirred at r.t. overnight. The reaction was concentrated to give 2-(azetidin-3-yl)-N-(methylsulfonyl)acetamide (2.1 g, TFA salt), which was directly used in the next step without any purification.
Step 3: Preparation of (J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(methylsulfonamido)-2- oxoethyl)azetidine-l-carboxylate
Figure imgf000052_0001
[0138] To a solution of 2-(azeti din-3 -yl)-N-(methylsulfonyl)acetamide (2.1 g, TFA salt ) in DCM (20 mL) was added DIPEA (4.6 g, 36 mmol) at 0°C. A solution of (3R,4S,5S,6R)-5- methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl lH-imidazole-l-carboxylate (1.3 g, 3.6 mmol) in DCM (5 mL) was added dropwise to the above reaction. The reaction mixture was stirred at r.t. overnight. The reaction mixture was washed with ammonium acetate buffer (pH 4.0, 5 mL x 2). The dichloromethane layer was separated, dried over Na2S04, filtered and concentrated to give crude product, which was purified by prep. HPLC (Method B: 0.l%NH4OH) to give (3R, 4S, 5S, ri/i)-5-methoxy-4- ((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-
(methylsulfonamido)-2-oxoethyl)azetidine-l-carboxylate as yellow oil (310 mg). LC-MS (ESI) found: 501 [M+l]+.
Step 4: Preparation of sodium(2-(l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)acetyl)(methylsulfonyl)amide
Figure imgf000052_0002
[0139] To a solution of (3R,-(S,5S, 6R)-5-methoxy-4-((2R, J/i)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(methylsulfonamido)-2-oxoethyl)azetidine-l- carboxylate (310 mg, 0.62 mmol) in MeCN (5 mL) was added 0.1M NaOH (5.5 mL, 0.55 mmol) at -78°C. The reaction was lyophilized to give sodium ( 2-(l-((((3R4S,5S,6R)-5 - methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetyl)(methylsulfonyl)amide (300 mg). LC-MS (ESI) found: 501 [M-23+2]+. 1H NMR (400 MHz, DMSO-76) d 5.26 (s, 1 H), 5.17 (t, J= 7.4 Hz, 1 H), 3.90 (t, j= 8.4 Hz, 2 H), 3.49 (dd, j= 10.8, 2.4 Hz, 3 H), 3.26 (s, 3H), 2.82 (d, j= 4.2 Hz, 1 H), 2.78 - 2.68 (m, 1 H), 2.66 (s, 3 H), 2.55 (dd, J= 12.6, 5.4 Hz, 2 H), 2.23 (d, J= 7.8 Hz, 2 H), 2.20 - 2.05 (m, 2 H), 1.91 (td, J= 13.2, 4.2 Hz, 1 H), 1.74 (t, 7= 9.6 Hz, 2 H), 1.68 (s, 4 H),
1.59 (s, 3 H), 1.05 (s, 3 H), 0.98 (d, J= 13.4 Hz, 1 H).
Example 9: Preparation of sodium ethyl(2-(l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate
Figure imgf000054_0001
Step 1: Preparation of tert-butyl 3-formylazetidine-l-carboxylate
Boc
Figure imgf000054_0002
[0140] To a solution of (COCl)2 (4.0 g, 31.5 mmol) in DCM (30 mL) was added DMSO (2.5 g, 31.4 mmol) at -78°C. The mixture was stirred at -78°C for 30min. A solution of tert- butyl 3- (hydroxymethyl)azetidine-l-carboxylate (3.0 g, 15.8 mmol) in DCM (30 mL) was added dropwise to the reaction at -78°C over 10 min. The reaction was stirred at -78°C for lh. TEA (8.1 g, 31.4 mmol) was added dropwise to the mixture at -78°C slowly. After addition, the reaction was stirred at -78°C for 30min then warmed to 0°C. The reaction was quenched by sat.NELCl (100 mL) and then extracted with DCM for 3 times. The dichloromethane layer was separated and dried over Na2S04. After filtration, the filtrate was concentrated to give tert- butyl 3-formylazetidine-l-carboxylate (4.2 g crude), which was directly used in the next step without any purification.
Step 2: Preparation of (E)-tert-but \ 3-(2-(diethoxyphosphoryl)vinyl)azetidine-l- carboxylate
Figure imgf000055_0001
[0141] To a solution of NaH (0.63 g, 17.38 mmol, 60%) in THF (20 mL) was added tetraethyl methylenebis(phosphonate) (4.72 g, 17.38 mmol) at 0°C. The reaction was stirred at 0°C for lh. A solution of /c/T-butyl 3 -for yl azeti di ne- 1 -carboxyl ate (4.2 g crude) in THF (20 mL) was added dropwise to the mixture at 0°C. After addition, the reaction was stirred at r.t. overnight. The reaction was quenched with sat.MLCl (100 mL) and then extracted with DCM for 3 times. The dichloromethane layer was separated and dried over Na2S04. After filtration, the filtrate was concentratedto give a residue, which was purified by flash chromatography (silica gel, 0 to ~2% MeOH in DCM) to give (E)-tert- butyl 3-(2-
(diethoxyphosphoryl)vinyl)azetidine-l-carboxylate as a yellow oil (1.2 g). LC-MS (ESI) found: 264 [M+l-56]+.
Step 3: Preparation of tert- butyl 3-(2-(ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l- carboxylate
Figure imgf000055_0002
[0142] To a solution of (E)-tert- butyl 3-(2-(diethoxyphosphoryl)vinyl)azetidine-l- carboxylate (1.2 g, 3.76 mmol) in ethanol (30 mL) was added Pd/C (0.12 g). The hydrogenation was performed with 1 atm and r.t. overnight. The reaction mixture was filtered and the filtrate was concentrated to give /c/V-butyl 3-(2-(ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l- carboxylate as a yellow oil (1.2 g), which was used directly in the next reaction without purification. LC-MS (ESI) found: 266 [M+l-56]+. Step 4: Preparation of ethyl hydrogen (2-(azetidin-3-yl)ethyl)phosphonate
Figure imgf000056_0001
[0143] To a solution of /cvV-butyl 3-(2-(ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l- carboxylate (1.2 g, 3.74 mmol) in DCM (10 mL) was added TMSBr (1.14 g, 7.47 mmol). The mixture was stirred under N2 atmosphere at r.t. overnight. The reaction solvent was removed under vacuum to give crude residue, which was purified by prep. HPLC (Method B: 0.1%
NH4OH) to give ethyl hydrogen (2-(azetidin-3-yl)ethyl)phosphonateas a yellow oil (108 mg). LC-MS (ESI) found: 194 [M+l]+.
Step 5: Preparation of (3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2- (ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l-carboxylate
Figure imgf000056_0002
[0144] To a solution of ethyl hydrogen (2-(azetidin-3-yl)ethyl)phosphonate (108 mg, 0.56 mmol) in DCM (2 mL) was added DIPEA (72 mg, 0.56 mmol) at 0°C. A solution of
( 3R , 4S, 5S, 6/?)-5-methoxy-4-((2A, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl lH-imidazole-l-carboxylate (186 mg, 0.488 mmol) in DCM (2 mL) was added to the above mixture. The reaction was stirred at r.t. overnight. The reaction mixture was monitored by TLC and then washed with ammonium acetate buffer (pH 4.0, 5 mL x 2). The dichloromethane layer was separated, dried over Na2S04, filtered and concentratedto give crude product, which was purified by prep. HPLC (Method B: 0.l%NH4OH) to give
( 3R , 4S, 5S, 6/?)-5-methoxy-4-((2A, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl 3-(2-(ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l-carboxylate as a yellow oil (53 mg). LC-MS (ESI) found: 502 [M+l]+.
Step 6: Preparation of sodium ethyl(2-(l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate
Figure imgf000057_0001
[0145] To a stirring solution of (3f?, ri',5ri', 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2- (ethoxy(hydroxy)phosphoryl)ethyl)azetidine-l-carboxylate (53 mg, 0.1 mmol) in MeCN (0.5 mL) was added 0.1M NaOH (0.9 mL, 0.09 mmol) at -78°C. The reaction mixture was lyophilized to give sodium ethyl(2-(l-((((3i?,¥ri',5ri', 6/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)phosphonate as a solid (57 mg). LC-MS (ESI) found: 502 [M-23+l]+. 1HNMR (400 MHz, DMSO-£¾) d 5.28 (s, 1 H), 5.19 (t, J= 7.4 Hz, 1 H), 3.90 (t, J= 7.9 Hz, 2 H), 3.66 - 3.56 (m, 2 H), 3.51 (dd, j= 11.0, 2.5 Hz, 1 H), 3.43 (s, 2 H), 3.28 (s, 3 H), 2.84 (d, j= 4.3 Hz, 1 H), 2.55 (t, J= 5.4 Hz, 3 H), 2.26 - 2.08 (m, 2 H), 1.97 (dt, J= 26.2, 8.2 Hz, 1 H), 1.80 - 1.74 (m, 2 H), 1.70 (s, 4 H), 1.63 (d, = 14.9 Hz, 5 H), 1.23 (s, 2 H), 1.16 - 1.05 (m, 7 H), 1.01 (d, 7 = 13.7 Hz, 1 H).
Example 10: Preparation of sodium 2-((l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)-2-methylpropanoate
Figure imgf000058_0001
Step 1: Preparation of ferf-Z>Hiy/ 3-((l-methoxy-l-oxopropan-2-yl)oxy)azetidine-l- carboxylate
Figure imgf000058_0002
[0146] To a solution of tert-butyl 3-hydroxyazetidine-l-carboxylate (2 g, 11.5 mmol) in THF
(20 mL) was added NaH (692 mg, 17.3 mmol) while cooling in an ice bath. After 30min, methyl 2-bromopropanoate (2.3 g, 13.8 mmol) was added. The resulting mixture was stirred at r.t. for 1 hr. The mixture was diluted with water and extracted with EtOAc (100 mL x2). The organic layer was washed with H20 (100 ml), dried over Na2S04, filtered and concentrated. The reside was purified by column chromatography (silica gel, 1 to -10 % ethyl acetate in petroleum ether) to give tert- butyl 3-((l-methoxy-l-oxopropan-2-yl)oxy)azetidine-l- carboxylate (2.3 g) as yellow oil. LC-MS (ESI) m/z (M+23): 282. Step 2: Preparation of tert-butyl 3-((l-methoxy-2-methyl-l-oxopropan-2- yl)oxy)azetidine-l-carboxylate
Figure imgf000059_0001
[0147] To a solution of tert-butyl 3-((l-methoxy-l-oxopropan-2-yl)oxy)azetidine-l- carboxylate (2.3 g, 8.8 mmol) in THF (20 mL) was added LDA (6.6 mL, 13.3 mmol) at -78°C. After 30min, CH3I (1.5 g, 10.5 mmol) was added. The resulting mixture was stirred at r.t. for 2 hrs. The mixture was diluted with water and extracted with EtOAc (100 mL x2). The organic layer was washed with H20 (100 mL), dried over Na2S04, filtered and concentrated. The reside was purified by flash chromatography (silica gel, 5 to -10 % ethyl acetate in petroleum ether) to give tert-butyl 3-((l-methoxy-2-methyl-l-oxopropan-2-yl)oxy)azetidine-l-carboxylate (1.8 g) as yellow oil. LC-MS (ESI) m/z (M+l): 274.
Step 3: Preparation of 2-((l-(tert-butox carbonyl)azetidin-3-yl)oxy)-2-methylpropanoic acid
Boc
Figure imgf000059_0003
[0148] To a solution of tert-butyl 3-((l-methoxy-2-methyl-l-oxopropan-2-yl)oxy)azetidine- l-carboxylate (1.8 g, 6.5 mmol) in THF/H20 (15 ml/5 ml) was added LiOH (2.7 g, 65 mmol). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated and diluted with H20, the pH adjusted to~3 with cone. HC1, and extracted with EtOAc (30 mL x2). the ethyl acetate extracts were washed with water and brine, dried over Na2S04 and concentrated to give 2-((l-(/er/-butoxycarbonyl)azeti din-3 -yl)oxy)-2-methylpropanoic acid (1.3 g) as a colorless oil. LC-MS (ESI) m/z (M+l): 260.
Step 4: Preparation of 2-(azetidin-3-yloxy)-2-methylpropanoic acid
Figure imgf000059_0002
[0149] To a solution of 2-((l -(tert-butoxycarbonyl)azeti din-3 -yl)oxy)-2-methylpropanoic acid (300 mg, 1.15 mmol) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at r.t. for 1 h. The reaction mixture was concentrated and the residue was used in next step without further purification. LC-MS (ESI) m/z (M+l): 160. Step 5: Preparation of 2-((l-((((J ?, A,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)-2-methylpropanoic acid
Figure imgf000060_0001
[0150] 2-(Azetidin-3-yloxy)-2-methylpropanoic acid (200 mg, 0.78 mmol) in CH3CN (20 ml) was coupled with Intermediate 1 (418 mg, 0.93 mmol) by following the General Procedure for Coupling. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The reaction mixture was washed with ammonium acetate buffer (pH 4.0, 5 mL x 2) and 5% sodium bicarbonate solution (20 mL). The mixture was extracted with DCM. The DCM layer was dried over Na2S04, concentrated to give crude product for prep. HPLC (C18, 0 to ~90 % acetonitrile in H20 with 0.1 % formic acid) to give 2-((l- ((((3A, 4S, 5S, 6R)-5-methoxy-4-((2R, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)-2-methylpropanoic acid (82mg) as a white solid. LC -MS (ESI) m/z (M+l): 468.
Step 6: Preparation of sodium 2-(( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)-2-methylpropanoate
Figure imgf000060_0002
[0151] To a solution of 2-((l-((((3f?, S,,5ri,,6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)-2- methylpropanoic acid (83 mg, 0.17 mmol) in MeCN (5 mL) was added dropwise 0.1 N NaOH solution (1.6 mL, 0.15 mmol) at -60°C. After lyophilization, 2-(( l -((((3R,-fS,5S,6R)-5- ethoxy- 4 -{{2R, 3i?)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azeti din-3 -yl)oxy)-2-methylpropanoatesodium salt (75 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 468. 1H NMR (400 MHz, DMSO- d) d 5.29 (d, J= 2.3 Hz, 1H), 5.19 (t, J= 7.4 Hz, 1H), 4.52 - 4.45 (m, 1H), 4.05 - 3.95 (m, 2H), 3.66 (s, 2H), 3.51 (dd, j= 11.0, 2.6 Hz, 1H), 3.28 (s, 3H), 2.84 (d, j= 4.3 Hz, 1H), 2.55 (t, J= 5.3 Hz, 2H), 2.22 - 2.12 (m, 2H), 1.93 (td, 7= 13.4, 4.5 Hz, 1H), 1.75 (d, = 11.0 Hz, 2H), 1.70 (s, 3H), 1.61 (s, 3H), 1.15 (s, 6H), 1.07 (s, 3H), 1.00 (d, 7= 13.2 Hz, 1H), 0.94 (d, J= 6.6 Hz, 1H).
Example 11: Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-2,2-dimethylpropanoate
Figure imgf000061_0001
Step 1: Preparation of (E)-tert-butyl 3-(3-ethoxy-3-oxoprop-l-en-l-yl)azetidine-l- carboxylate
Figure imgf000061_0002
[0152] To a solution of tert-butyl 3-formylazetidine-l-carboxylate (7 g, 0.037 mol) in anhydrous DCM (30 mL) was added ethyl 2-(triphenylphosphoranylidene)acetate (14.4 g, 0.042 mol) in portions at 0°C. The reaction mixture was stirred at room temperature overnight. The resulting mixture was concentrated under vacuum. The residue was purified by flash chromatography (silica gel, 1 to -20% ethyl acetate in petroleum ether) to give (E)-tert-butyl 3- (3 -ethoxy-3 -oxoprop-l-en-l-yl)azeti dine- l-carboxylate (7.0 g) as a pale-yellow oil.
Step 2: Preparation of tert-butyl 3-(3-ethoxy-3-oxopropyl)azetidine-l-carboxylate
Figure imgf000062_0001
[0153] A solution of (E)-tert-butyl 3-(3-ethoxy-3-oxoprop-l-en-l-yl)azetidine-l-carboxylate (5 g, 0.019 mol) in MeOH (80 mL) was degassed under N2 atmosphere three times, then Pd/C (1.5 g, 10% wt) was added in one portion. The mixture was stirred under an H2 balloon at room temperature overnight. The resulting mixture was filtered through a pad of celite and the filtrate was concentrated under vacuum. The residue was purified by flash chromatography (silica gel,
1 to -3% MeOH in DCM) to give tert-butyl 3-(3-ethoxy-3-oxopropyl)azetidine-l-carboxylate (4.5 g) as a colorless oil.
Step 3: Preparation of tert-butyl 3-(3-ethoxy-2,2-dimethyl-3-oxopropyl)azetidine-l- carboxylate
Figure imgf000062_0002
[0154] To a solution of tert-butyl 3 -(3 -ethoxy-3 -oxopropyl)azeti dine- l-carboxylate (1 g, 3.87 mmol ) in anhydrous THF (20 mL) was added a solution of LDA (11.6 mL, 11.6 mmol, 1M in THF) dropwise at -78°C over 30 min. After the addition, the mixture was stirred at -78°C for 1 hr. Then Mel (2.2 g, 15.5 mmol) was added dropwise. The resulting mixture was quenched by sat.NH4Cl at 0 °C and extracted with ethyl acetate (20 mL x 2). The combined organic layers were dried over Na2S04, filter and concentrated to give crude product. The crude product was purified by flash chromatography (silica gel, 1 to -20% ethyl acetate in petroleum ether) to give tert-butyl 3-(3-ethoxy-2,2-dimethyl-3-oxopropyl)azetidine-l-carboxylate (380 mg).
Step 4: Preparation of 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2,2-dimethylpropanoic acid
Figure imgf000062_0003
[0155] To a mixture of tert-butyl 3-(3-ethoxy-2,2-dimethyl-3-oxopropyl)azetidine-l- carboxylate (380 mg, 1.32 mmol) in THF (4 mL) was added 4M aq.NaOH solution (3.3 mL, 13.2 mmol). The mixture was stirred at 50°C for 4 hrs. The volatiles were removed under reduced pressure, and the residue was diluted with water (7 mL) and washed with EA (10 mL x 2). The aqueous layer was adjusted pH to 3 with 1N hydrochloric acid and extracted with EA (10 mL x 2). The combined organic layers were washed with brine, dried over Na2S04 and concentrated under reduced pressure to give 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2,2- dimethylpropanoic acid (320 mg) as a pale-yellow oil.
Step 5: Preparation of 3-(azetidin-3-yl)-2,2-dimethylpropanoic acid
CF3C02H
Figure imgf000063_0001
[0156] To a solution of 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2,2-dimethylpropanoic acid (320 mg, 1.24 mmol) in DCM (2 mL) was added TFA (1 mL, l2.4mmol) dropwise at 0°C. The mixture was stirred at room temperature for lhr. The resulting mixture was concentrated under vacuum to give a residue, which was co-evaporated with toluene (5 mL x 2) to give 3-(azetidin- 3-yl)-2,2-dimethylpropanoic acid (295 mg, contained TFA) as brown oil, which was directly used in the next step without purification. LC-MS: m/z 158 (M+H)+
Step 6: Preparation of 3-(3-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)cyclobutyl)-2,2- dimethylpropanoic acid
Figure imgf000063_0002
[0157] 3-(Azetidin-3-yl)-2,2-dimethylpropanoic acid (295 mg, -1.24 mmol) was coupled to Intermediate 1 (500 mg, 1.12 mmol) by following the General Procedure for Coupling. The solvent was removed under vacuum below 40°C, then diluted with DCM (20 mL) and washed with ammonium acetate buffer (pH~4, 5 mL x 2) and 5% sodium bicarbonate solution (20 mL). The aqeous layers were extracted with DCM (5 mL x 2). The combined organic layers were dried over Na2S04 and concentrated to give a yellow oil, which was purified by flash chromatography (silica gel, 1 to -3% MeOH in DCM) to give pure 3-(3-((((3R,4S,5S,6R)-5- methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)cyclobutyl)-2,2-dimethylpropanoic acid (l60mg)as a light yellow syrup.
Step 7: Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-
2,2-dimethylpropanoate
Figure imgf000064_0001
[0158] To a solution of 3-(3-((((3f?, ,S,,5S,,6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)cyclobutyl)-2,2- dimethylpropanoic acid (160 mg, 0.34 mmol) in CH3CN (0.5 mL) was added O. lM NaOH (3 mL, 0.31 mmol) dropwise at 0~5°C. The mixture was freeze dried to give the 3-(3-
((((37?, 4S, 5S, 6/ )- 5 -m ethoxy-4 -((2/?, 37?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)cyclobutyl)-2,2-dimethylpropanoate as a white solid (170 mg). LC-MS (ESI) found: 465 [M+l]+.1H NMR (400 MHz, DMSO) d 5.28 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 3.89 (t, J= 8.2 Hz, 2H), 3.51 (dd, J= 10.9, 2.5 Hz, 3H), 3.27 (s, 3H), 2.84 (d, J= 4.3 Hz, 1H), 2.69 - 2.61 (m, 1H), 2.56 (dd, J= 8.2, 4.2 Hz, 2H), 2.17 (dd, J= 12.5, 6.3
Hz, 2H), 1.90 (dd, J= 13.2, 4.5 Hz, 1H), 1.74 - 1.59 (m, 11H), 1.05 (d, 7= 19.9 Hz, 4H), 0.92 (d, J= 10.7 Hz, 6H).
Example 12: Preparation of sodium (3-(l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoyl)(methylsulfonyl)amide
Figure imgf000064_0002
Step 1: Preparation of terf-butyl3-(3-(methylsulfonamido)-3-oxopropyl)azetidine-l- carboxylate Boc
Figure imgf000065_0003
DCM
[0159] To a solution of 3-(l-(/er/-butoxycarbonyl)azetidin-3-yl)propanoic acid (2.0 g, 8.7 mmol) in DCM (20 mL) was added methanesulfonamide (0.99 g, 10.4 mmol), TBTU (4.18 g, 13.05 mmol) and DIPEA (2.20 g, 17.4 mmol). The mixture reaction was stirred at r.t.
overnight. The reaction was washed with water 3 times and the dichloromethane solution was dried over Na2S04, filtered and concentrated to give crude product, which was purified by flash chromatography (silica gel, 0 to ~3% MeOH in DCM) to give ter/-butyl3-(3- (methylsulfonamido)-3-oxopropyl)azetidine-l-carboxylate as a yellow oil (0.80 g). LC-MS (ESI) found: 251 [M+l-56]+.
Step 2: Preparation of 3-(azetidin-3-yl)-N-(methylsulfonyl)propenamide
Figure imgf000065_0001
[0160] To a solution of /c/V-butyl 3-(3-(methylsulfonamido)-3-oxopropyl)azetidine-l- carboxylate (0.8 g, 2.6 mmol) in DCM (8 mL) was added TFA (1.5 g, 13.1 mmol). The reaction was stirred at r.t. overnight. The reaction was concentrated to give 3-(azetidin-3-yl)-N- (methylsulfonyl)propanamide (1.4 g, TFA salt), which was directly used in the next step without any purification.
Step 3: Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(3-(methylsulfonamido)-3- oxopropyl)azetidine-l-carboxylate
Figure imgf000065_0002
[0161] To a solution of 3-(azetidin-3-yl)-N-(methylsulfonyl)propanamide (1.4 g, TFA salt) in DCM (20 mL) was added DIPEA (3.46 g, 24 mmol) at 0°C. Then a solution of ( 3R,4S,5S,6R)- 5-methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl lH-imidazole-l-carboxylate (0.97 g, 2.4 mmol) in DCM (5 mL) was added to the above reaction. The reaction mixture was stirred at r.t. overnight. The reaction mixture was extracted with ammonium acetate buffer (pH 4.0, 5 mL x 2) and stirred for 15 min. The dichloromethane solution was separated, dried over Na2S04, filtered and concentrated to give crude product, which was purified by Prep. HPLC (Method B: 0.1% NH4OH) to give ( 3R,4S,5S, 6R)-5 - methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(3-(methylsulfonamido)-3-oxopropyl)azetidine-l-carboxylate as a yellow oil (220 mg). LC- MS (ESI) found: 515 [M+l]+.
Step 4: Preparation of Sodium (3-( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoyl)(methylsulfonyl)amide
Figure imgf000066_0001
[0162] To a solution of (3R,-(S,5S, 6R)-5-methoxy-4-((2R, J/i)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl3-(3-(methylsulfonamido)-3-oxopropyl)azetidine-l- carboxylate (220 mg, 0.43 mmol) in MeCN (5 mL) was added 0.1M NaOH (3.8 mL, 0.38 mmol) at -78°C. The mixture was lyophilized to give sodium ( 3-(l-((((3R,4S,5S, 6R)-5 - methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoyl)(methylsulfonyl)amide as white solid. (200 mg). LC- MS (ESI) found:5 l5 [M-23+2]+. 1H NMR (400 MHz, CDCl3) d 5.52 (s, 1 H), 5.19 (d, J= 7.2 Hz, 1 H), 4.04 (dd, J= 17.1, 8.8 Hz, 2 H), 3.82 - 3.56 (m, 3 H), 3.46 (d, J= 8.8 Hz, 3 H), 3.09 (s, 3 H), 2.95 (d, j= 3.9 Hz, 1 H), 2.75 - 2.49 (m, 4 H), 2.28 (s, 2 H), 2.16 (dd, j= 8.9, 6.4 Hz, 2 H), 2.06 (dd, j= 14.7, 10.3 Hz, 2 H), 1.93 (s, 1 H), 1.91 (d, j= 13.2 Hz, 2 H), 1.85 (d, j= 5.7 Hz, 1 H), 1.74 (s, 3 H), 1.66 (s, 3 H), 1.25 (s, 1 H), 1.21 (s, 3 H), 1.05 (d, j= 12.2 Hz, 1 H). Example 13: Preparation of sodium 5-((l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate
1 ,1 '-carbonyl- diimidazole,
Figure imgf000067_0001
Step 1: Preparation of tert-butyl 3-(4-ethoxy-2,4-dioxobutyl)azetidine-l-carboxylate
1 ,1'-carbonyl- diimidazole,
Figure imgf000067_0002
[0163] To a stirred solution of 2-(l -(tert-butoxycarbonyl)azeti din-3 -yl)acetic acid (2.0 g, 9.3 mmol) in THF (20 mL) was added 1,1’ -carbonyl diimidazole (2.2 g, 13.9 mmol). The mixture was stirred at r.t. for lh, then magnesium chloride (883 mg, 9.3 mmol) and ethyl potassium malonate (2.44 g, 9.3mmol) were added successively. The mixture was then heated to 50°C for 15 hr. The mixture was filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (silica gel, 5 % MeOH in DCM) to afford tert-butyl 3-(4- ethoxy-2,4-dioxobutyl)azetidine-l-carboxylate (1.5 g) as a yellow oil. LC-MS (ESI) m/z (M+l): 286.
Step 2: Preparation of tert-butyl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l- carboxylate
Figure imgf000067_0003
[0164] To a solution of tert-butyl 3-(4-ethoxy-2,4-dioxobutyl)azetidine-l-carboxylate (500 mg, 1.75 mmol) in H20 (10 ml) was added NH2OH/HCl (945 mg, l3.5mmol) at 0°C. After 30 min, MeOH/THF (5 mL/5 mL) was added. The resulting mixture was stirred at 0°C for 20 min and r.t. for 2 hr. The reaction mixture was concentrated and diluted withH20, and the pH was adjusted to ~4 with 2NHC1. The acidic water mixture was extracted with EtOAc (30 mL x2), dried over Na2S04 and concentrated. The residue was purified by flash chromatography (silica gel, 5 % MeOH in DCM) to give tert-butyl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l- carboxylate (300 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 255.
Step 3: Preparation of 5-(azetidin-3-ylmethyl)isoxazol-3-ol
Boc
Figure imgf000068_0001
[0165] To a solution of tert-butyl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l-carboxylate (300 mg, 1.18 mmol) in DCM(3 mL) was added TFA (1 mL). The mixture was stirred at r.t. for
1 h and concentrated under vacuum. The crude product was used in next step without further purification.
Step 4: Preparation of (3T?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((3-hydroxyisoxazol-5- yl)methyl)azetidine-l-carboxylate
Figure imgf000068_0002
[0166] 5-(Azetidin-3-ylmethyl)isoxazol-3-ol (260 mg, 1.03 mmol) was coupled with
Intermediate 1 (505 mg, 1.13 mmol) by following the General Procedure for Coupling. The reaction mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by column chromatography (silica gel, 1 to~l0 % MeOH in DCM) and prep. HPLC (C18, 0 ~90 % acetonitrile in H20 with 0.1 % formic acid) to give (3i?,4ri',5ri',6/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l- carboxylate (160 mg) as a white solid.LC-MS (ESI) m/z (M+l): 463.
Step 5: Preparation of sodium 5-((l-((((3/?, ,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)isoxazol-3-olate
Figure imgf000069_0001
[0167] To a solution of (3R,-(S,5S, 6R)-5-methoxy-4-((2R, J/i)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-((3-hydroxyisoxazol-5-yl)methyl)azetidine-l- carboxylate (160 mg, 0.34 mmol) in MeCN (5 mL) was added dropwise 0.1 N NaOH solution (3.1 mL, 0.31 mmol) at -60°C with stirring. Lyophilization provided 5 -( ( 1 - (( (( 3/( -fS, 5S, 6R)-5- methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate sodium salt (139 mg) as a white solid. LC-MS (ESI) m/z (M+l): 463. 1H NMR (400 MHz, DMSO-76) d 5.28 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 3.91 (t, J= 8.3 Hz, 2H), 3.66 (s, 1H), 3.59 - 3.46 (m, 3H), 3.28 (s, 3H), 2.84 (d, J = 4.3 Hz, 1H), 2.77 - 2.66 (m, 1H), 2.56 (dd, 7= 11.5, 5.3 Hz, 2H), 2.42 (d, J= 7.3 Hz, 2H), 2.24
- 2.07 (m, 2H), 1.92 (tt, 7= 19.9, 9.9 Hz, 1H), 1.76 (t, J= 8.8 Hz, 2H), 1.70 (s, 4H), 1.61 (s, 3H), 1.07 (s, 3H), 1.00 (d, J= 13.5 Hz, 1H).
Example 14: Preparation of sodium 5-(2-(l-((((3/?, £,5»£,<i/?)-5-inethoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethyl)isoxazol-3-olate
Figure imgf000070_0001
Step 1: Preparation of 3-(l-(terf-butoxycarbonyl)azetidin-3-yl)propanoic acid
Figure imgf000070_0002
[0168] To a solution of /cvV-butyl 3 -(3 -ethoxy-3 -oxopropyl)azeti dine- l-carboxylate (2.0 g, 7.78 mmol) in MeOH (30 mL) was added aqueous LiOH (10 mL, 23.35 mmol,l M) at 0°C. The reaction was stirred at 25°C for 6 hrs. The mixture was diluted with THF and extracted with diethyl ether (20 mL x 2). The aqueous layer was adjusted to ~pH3-4 withHCl (1M) and extracted with dichloromethane (30 mL x 3). The dichloromethane layers were dried over anhydrous Na2S04 and concentrated under reduced pressure to give 3-(l -{tert- butoxycarbonyl)azetidin-3-yl)propanoic acid (1.78 g) as a white solid. LC-MS (ESI) found:
230 [M+H] +
Step 2: Preparation of tert- butyl 3-(5-ethoxy-3,5-dioxopentyl)azetidine-l-carboxylate
Figure imgf000070_0003
[0169] To a suspension of MgCl2 (998 mg, 10.48 mmol) and TEA (1.88 mL, 13.54 mmol) in MeCN (90 mL) was added potassium 3 -ethoxy-3 -oxopropanoate (1.56 g, 9.17 mmol) at 25°C. The reaction was stirred at 25°C for 2hrs, to prepare mixture 1. 3-( 1 -(tert- butoxycarbonyl)azetidin-3-yl)propanoic acid (1.0 g, 4.36 mmol) was added to DMF (30 mL), followed by adding CDI (778 mg, 4.80 mmol), the resulting mixture was stirred at 25°C for 2 hrs, to prepare mixture 2. The mixture 1 was added into the mixture 2, and then the resulting reaction mixture was stirred overnight at room temperature, the reaction mixture was extratcted with EA and the EA layer was washed with water several times. The organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure to give /er/-butyl 3-(5- ethoxy-3,5-dioxopentyl)azetidine-l-carboxylate (1.05 g) as a colorless oil. LC-MS (ESI) found: 300 [M+H] +
Step 3: Preparation of tert-butyl 3-(2-(3-hydroxyisoxazol-5-yl)ethyl)azetidine-l- carboxylate
Figure imgf000071_0001
[0170] To a cold slurry of /er/-butyl 3-(5-ethoxy-3,5-dioxopentyl)azetidine-l-carboxylate (220 mg, 0.76 mmol) in water (3 mL) was added hydroxylamine hydrochloride (330 mg, 4.78 mmol) at 0°C, followed by NaOH (380 mg, 9.5 mmol). After 5min, THF (1.5 mL) and MeOH (1.5 mL) were added, the resulting cloudy light yellow solution was stirred at 0°C for 20 min, then warmed up to room temperature for 6hr. The reaction was acidified with cone. HC1, diluted by water and extracted with diethyl ether (10 mL x 4). The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give /cvV-butyl 3-(2-(3-hydroxyisoxazol-5-yl)ethyl)azetidine-l-carboxylate (l80mg) as a light yellow oil. LC-MS (ESI) found: 269 [M+H] +
Step 4: Preparation of 5-(2-(azetidin-3-yl)ethyl)isoxazol-3-ol
Figure imgf000071_0002
[0171] To a mixture of /er/-butyl 3-(2-(3-hydroxyisoxazol-5-yl)ethyl)azetidine-l-carboxylate (180 mg, 0.67 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (2.5 mL) dropwise at 0°C. The reaction was stirred at room temperaturefor 1 hr. The mixture was concentrated under reduced pressure to give 5-(2-(azetidin-3-yl)ethyl)isoxazol-3-ol trifluoroacetic acid salt (200 mg) as yellow resin, which was directly used in the next step without purification. LC-MS (ESI) found: 169 [M+l] +.
Step 5: Preparation of sodium 5-(2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)isoxazol-3-olate
Figure imgf000072_0001
[0172] Under N2 atmosphere, a mixture of 5-(2-(azetidin-3-yl)ethyl)isoxazol-3- oltrifluoroacetic acid salt (200 mg, 0.839 mmol) in acetonitrile (5 mL) was added DIPEA (0.3 mL, 1.69 mmol) dropwise at 0°C. The mixture was stirred at 0°C for 10 min, then
Intermediate 1 (140 mg, 0.335 mmol) and 4-dimethylaminopyridine (5 mg) were added. After addition, the reaction was stirred at room temperature overnight under N2 atmosphere. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed twice with ammonium acetate buffer (pH 4.0, 5 mL x 2). The dichloromethane solution was dried over Na2S04, filtered and concentrated. The crude product was purified by preparative HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give (3/(-/A5k, 6//)-5-rnethoxy-4-((2/(3//)-2-rnethyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl3-(2-(3-hydroxyisoxazol-5- yl)ethyl)azetidine-l-carboxylate (130 mg) as a colorless oil. LC-MS (ESI) found:477 [M+l] +.
[0173] To a solution of (3R, S,5S, 6R)-5-methoxy-4-((2R, 3//)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-(2-(3-hydroxyisoxazol-5-yl)ethyl)azetidine-l- carboxylate (130 mg, 0.273 mmol) in acetonitrile/H20 (0.5 mL:2 mL) was added NaOH (2 mL, 0.1 N in water). Lyophilization give 5-(2-( l -((((3//,-/ri',5A6//)-5-methoxy-4-((2//,3//)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)isoxazol-3-olate sodium salt (130 mg) as a colorless foam. LC-MS (ESI) found:477 [M+l] +. 1H MR (400 MHz, DMSO-i/d) d 5.28 (s, 1H), 5.19 (t, J= 7.6 Hz, 1H), 3.99 - 3.82 (m, 2H), 3.67 (s, 1H), 3.51 (dd, j= 10.9, 2.6 Hz, 3H), 3.28 (s, 3H), 2.84 (d, j= 4.4 Hz, 1H), 2.61 - 2.51 (m, 4H), 2.17 (h, J= 7.7 Hz, 2H), 2.08 (t, J= 7.5 Hz, 2H), 1.99 - 1.88 (m, 1H),
1.77 - l .72(m, 2H),l .72- 1.68 (m, 5H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, J= 13.7 Hz, 1H). Example 15: Preparation of sodium 3-((l-((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoate
Boc H
Figure imgf000073_0001
Step 1: Preparation of tert- butyl 3-(3-(/er/-butoxy)-3-oxopropoxy)azetidine- l-carboxylate
Figure imgf000073_0002
[0174] /er/-Butyl 3-hydroxyazetidine-l-carboxylate (500 mg 2.89 mmol) was combined with acrylic acid tert. -butyl ester (1.80 g, 14.45 mmol) and tetra-n- butylammoniumhydrogensulphate (197 mg, 0.578 mmol) in dichloromethane (10 mL) at 0 °C. Aqueous 50% sodium hydroxide (2.67 mL, 3.44 mmol) was added and the mixture was stirred vigorously at 0° C for 1 h. The reaction was warmed up to RT, and the stirring was continued overnight at RT. The reaction was diluted with dichloromethane and water, acidifed with 10% citric acid solution and the phases were separated. The aqueous phase was extracted once again with dichloromethane. The organic layers were combined, washed with aqueous sodium chloride, dried over magnesium sulphate and concentrated by evaporation under vacuum. Flash chromatography gave /c/T-butyl 3-(3-(/c/V-butoxy)-3-oxopropoxy)azetidine- l -carboxyl ate (600 mg) as a colorless oil. LC-MS (ESI) found: 302 [M+H] + Step 2: Preparation of 3-(azetidin-3-yloxy)propanoic acid
Figure imgf000074_0001
[0175] To a mixture of /er/-butyl 3-(3-(tert-butoxy)-3-oxopropoxy)azetidine-l-carboxylate (840 mg, 2.79 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (2.5 mL) dropwise at 0°C. The reaction was stirred at room temperature for 16 hr. The mixture was concentrated under reduced pressure to give 3-(azetidin-3-yloxy)propanoic acid (1.0 g) as yellow syrup, which was directly used in the next step without purification. LC-MS (ESI) found: 146 [M+l]+.
Step 3: Preparation of 3-((l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)propanoic acid
Figure imgf000074_0002
[0176] DIPEA (0.35mL, 2.0 mmol) was added dropwise to a solution of 3-(azetidin-3- yloxy)propanoic acid (290 mg, 2.0 mmol) in acetonitrile (5 mL) at 0°C under N2. The mixture was stirred at 0°C for 10 min and Intermediate 1 (150 mg, 0.33 mmol) and DMAP (5 mg) were added. After addition, the reaction was stirred at room temperature overnight under N2 atmosphere. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was acidified by ammonium acetate buffer and 5% sodium bicarbonate solution (20 mL), then extratcted with dichloromethane. The dichloromethane solution was dried over Na2S04, filtered and concentrated. The crude product was purified by preparative HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 3-((l- ((((3R, 4S, 5S, 6// )- 5 -m ethoxy-4 -((2//, 3i?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoic acid (140 mg) as a colorless oil. LC-MS (ESI) found:454 [M+l] +. Step 4: Preparation of sodium 3-(( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)propanoate
Figure imgf000075_0001
[0177] To a solution of S-^l-^^df^^S'.J^d/^-S-methoxy-d-^i.R.df^^Z-inethyl-S-p- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)propanoic acid (140 mg, 0.3 lmmol) in acetonitrile/H20 (0.5 mL/2 mL) was added NaOH (3 mL, 0.1 N in water). Lyophilization gave 3 -(( 1 -((((3/( -(S, 5S, 6/i)-5-methoxy-4- ((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoate sodium salt (142 mg) as a colorless foam. LC- MS (ESI) found: 454 [M+l]+. 1H MR (400 MHz, DMSO- d) d 5.34 - 5.26 (m, 1H), 5.19 (ddt, J= 7.4, 5.9, 1.5 Hz, 1H), 4.23 (tt, J= 6.4, 4.2 Hz, 1H), 4.09(m, 2H), 3.77- 3.61 (m, 2H), 3.54 - 3.44 (m, 3H), 3.28 (s, 3H), 2.85 (d, J= 4.4 Hz, 1H), 2.59 - 2.53 (m, 2H), 2.16 (p, J = 7.6, 7.1 Hz, 2H), 2.08 (t, J= 7.0 Hz, 2H), 1.93 (dt, J= 13.3, 6.7 Hz, 1H), 1.81 - 1.64 (m, 6H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (dt, j= 13.6, 3.7 Hz, 1H).
Example 16: Preparation of sodium 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate
Figure imgf000076_0001
Figure imgf000076_0004
Step 1: Preparation of (S)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate
Figure imgf000076_0002
[0178] A solution of (S)-tert-butyl 3-hydroxypyrrolidine-l-carboxylate (500 mg, 2.70 mmol) in anhydrous THF (5.0 mL) was added dropwise to a slurry of sodium hydride (60% dispersion in oil, 128 mg, 3.21 mmol) in THF (10.0 mL) at 0°C under a nitrogen atmosphere, and the mixture stirred for 15 mins at 0 °C. A solution of tert-butyl 2-bromoacetate (576 mg, 3.0mmol) in THF (5.0 mL) was added and the mixture warmed to room temperature and stirred for 18 hours. Water (20.0 mL) was added, and the mixture was extracted with ethyl acetate (50.0 mL). The organic extract was washed with brine, dried over anhydrous Na2S04, and evaporated in vacuo. The residue was purified by flash chromatography (silica gel, 1 to ~50 % ethyl acetate in petroleum ether) to give (S)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate (620 mg) as a white solid. LC-MS (ESI) found: 302 [M+l]+.
Step 2: Preparation of (S)-2-(pyrrolidin-3-yloxy)acetic acid trifluoroacetic acid salt
Figure imgf000076_0003
[0179] To a mixture of (S)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate (620 mg, 2.1 mmol) in dichloromethane (10 mL) was added trifluoroacetic acid (4 mL) dropwise at 0°C. The reaction was stirred at room temperature for 1 hr. The mixture was concentrated under reduced pressure to give (S)-2-(pyrrolidin-3-yloxy)acetic acid
trifluoroacetic acid salt (480 mg) as yellow syrup, which was directly used in the next reaction without purification. LC-MS (ESI) found: 146[M+1]+.
Step 3: Preparation of 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid
Figure imgf000077_0001
[0180] (S)-2-(Pyrrolidin-3-yloxy)acetic acid trifluoroacetic acid salt (480 mg, 2.0 mmol) was coupled with Intermediate 1 (550 mg, 1.2 mmol) by following the General procedure for Coupling. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL x 2) followed by 5% sodium
bicarbonate solution (20 mL). The dichloromethane solution was dried over Na2S04, filtered and concentrated to give crude product. The crude product was purified by silica gel chromatography (eluting with 1 to~2 % MeOH in DCM) and preparative HPLC (Method A, ¾0 (0.1% FA)/ CHsCN) to give 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid (375 mg) as a white solid. LC-MS (ESI) found: 454 [M+l]+.
Step 4: Preparation of sodium 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin- 3-yl)oxy)acetate
Figure imgf000077_0002
[0181] To a solution of 2-(((S)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid (135 mg, 0.3 mmol) in acetonitrile (2 mL) was added aq.NaOH (2.7 mL,
0.27 mmol) dropwise at -20°C. The mixture was freeze drying to give sodium 2-(((S)-l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate (150 mg, 0.29 mmol) as a white solid. LC-MS (ESI) found: 454 [M+l]+. 1H NMR (400 MHz, DMSO- d) d 5.36 (d, J = 5.5 Hz, 1H), 5.19 (s, 1H), 4.20 (d, J= 22.1 Hz, 1H), 3.62 - 3.41 (m, 3H), 3.29 (d, J= 5.0 Hz, 8H), 2.91 - 2.80 (m, 1H), 2.72 - 2.53 (m, 2H), 2.30 - 2.08 (m, 2H), 1.98 (dd, = 15.9, 11.1 Hz, 2H), 1.81 (dd, 11.1, 2.9 Hz, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.08 (d, J= 3.5 Hz, 3H), 1.01 (d, 7= 13.5 Hz, 1H).
Example 17: Preparation of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl3-(2- (((isopropoxycarbonyl)oxy)methoxy)-2-oxoethoxy) azetidine-l-carboxylate
Figure imgf000078_0001
[0182] To a solution of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetic acid (235 mg, 0.54 mmol) in dry DMF (20 mL) was added K2C03 (221 mg, 1.60 mmol) at r.t. The mixture was cooled to 0°C, and chloromethyl isopropyl carbonate (160 mg, 1.07 mmol) and Nal (79.70 mg, 0.54 mmol) were added to the reaction solution at 0°C under N2 atmosphere. After the completion of addition, the reaction mixture stirred at r.t. overnight. The mixture was diluted with EA and washed with water. The organic layer was then washed with brine, dried over Na2S04, and concentrated. The residue was purified by prep-HPLC (Method A, H20 (0.1% FA)/ C¾CN) to give (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl3 - (2(isopropoxycarbonyl)oxy)methoxy)-2-oxoethoxy)azetidine-l-carboxylate (68.6 mg) as a yellow solid. LC-MS (ESI) found: 556 [M+l]+. 1H NMR (400 MHz, CDCl3) d 5.80 (s, 2H), 5.51 (s, 1H), 5.21 (t, 7= 7.5 Hz, 1H), 4.92 (dt, 7= 9.9, 6.3 Hz, 1H), 4.37 (s, 1H), 4.16 - 4.09 (m, 3H), 4.01 - 3.90 (m, 2H), 3.61 (dd, 7= 11.1, 2.7 Hz, 1H), 3.45 (d, 7= 2.4 Hz, 3H), 2.99 (t, 7= 8.5 Hz, 1H), 2.65 - 2.52 (m, 2H), 2.39 - 1.83 (m, 7H), 1.74 (s, 3H), 1.64 (d, 7= 8.6 Hz, 3H), 1.33 (s, 3H), 1.32 (s, 3H), 1.19 (d, 7= 6.3 Hz, 3H), 1.07 (d, 7= 12.7 Hz, 1H)
Example 18: Preparation of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl3-(2-oxo-2-
((pivaloyloxy)methoxy)ethoxy)azetidine-l-carboxylate
Figure imgf000079_0001
[0183] To a solution of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)oxy)acetic acid (235 mg, 0.54 mmol) in dry DMF (20 mL) was added K2C03 (221 mg, 1.60 mmol) at r.t. The mixture was cooled to 0°C, chloromethylpivalate (163 mg, l .08mmol) and Nal (79.70 mg, 0.54 mmol) was added to the reaction solution at 0°C under N2 atmosphere. After the completion of addition, the reaction mixture was stirred at r.t. overnight. The mixture was diluted with EA and washed with water. The combined organic layers were washed with brine and dried over Na2S04, concentrated. The residue was purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl3-(2-oxo-2- ((pivaloyloxy)methoxy)ethoxy)azetidine-l-carboxylate (98.3 mg) as a yellow solid. LC-MS (ESI) found: 554 [M+l]+. 1H NMR (400 MHz, CDCl3) d 5.80 (s, 2H), 5.51 (s, 1H), 5.20 (t, 7 = 7.5 Hz, 1H), 4.36 (s, 1H), 4.11 (dd, 7= 11.8, 8.5 Hz, 3H), 4.02 - 3.90 (m, 2H), 3.61 (dd, 7 = 11.1, 2.7 Hz, 1H), 3.44 (d, 7= 2.5 Hz, 3H), 2.99 (t, 7= 8.8 Hz, 1H), 2.65 - 2.49 (m, 2H), 2.44 - 2.30 (m, 1H), 2.21 - 1.77 (m, 6H), 1.74 (s, 3H), 1.65 (s, 3H), 1.22 (s, 9H), 1.19 (d, 7= 6.3 Hz, 3H), 1.07 (d, 7= 10.3 Hz, 1H). Example 19: Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)amino)acetate
OF 3C02
ate 1
Figure imgf000080_0003
Step 1: Preparation of tert-butyl 3-((2-(tert-butoxy)-2-oxoethyl)amino)azetidine-l- carboxylate
Figure imgf000080_0001
[0184] To a solution of tert-butyl 3-aminoazetidine-l-carboxylate (500 mg, 2.90mmol) in THF (20 ml) was added K2C03 (802 mg, 5.8mmol) and tert-butyl 2-bromoacetate (678 mg, 3.48mmol). The resulting mixture was stirred at r.t. overnight. The mixture was diluted with water and extracted with EtOAc (100 mL x 2). The organic layers were washed with H20 (100 mL), dried over Na2S04, filtered and concentrated. The residue was purified by flash chromatography (silica gel, 0 to ~2 % EtOAc in PE) to give tert-butyl 3-((2-(tert-butoxy)-2- oxoethyl)amino)azetidine-l-carboxylate (650 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 287.
Step 2: Preparation of 2-(azetidin-3-ylamino)acetic acid
Boc
N. TFA
DCM
HN
Figure imgf000080_0002
[0185] To a solution of tert-butyl 3-((2-(/er/-butoxy)-2-oxoethyl)amino)azetidine-l- carboxylate (650 mg, 2.27 mmol) in DCM (1 mL) was added TFA (3 mL). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated, and the crude product was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 131.
Step 3: Preparation of 2-((l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)amino)acetic acid
Figure imgf000081_0001
[0186] A solution of 2-(azetidin-3-ylamino)acetic acid (310 mg, l.36mmol) was coupled with Intermiate 1 (731 mg, 1.63 mmol) by following the General Procedure for Coupling. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by flash chromatography (silica gel, 1 % ~ 5 % MeOH in DCM) and prep. HPLC (Cl 8, 0 to ~90 % acetonitrile in H20 with 0.1 % ammonium hydroxide) to give, upon lyophilization, 2-(( l -((((3R,-(S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)amino)acetic acid (160 mg) as a white solid. LC-MS (ESI) m/z (M+l): 439.
Step 4: Preparation of sodium 2-(( l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)amino)acetate
Figure imgf000081_0002
[0187] To a solution of 2-(( l -{{{{3R,4S,5S,6R)- -mei\\oxy- -{{2R, 3//)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)amino)acetic acid (100 mg, 0.22 mmol) in MeCN (5 mL) was added dropwise 0.1 N NaOH solution (2 mL, 0.2 mmol) at -60 °C with stirring. After lyophilization, 2-(( l -((((3R,4S,5S,6R)- 5-methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)amino)acetate sodium salt (84mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 439. 1H NMR (400 MHz, DMSO) d 5.28 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 3.91 (t, J= 7.5 Hz, 2H), 3.61 - 3.48 (m, 4H), 3.28 (s, 3H), 2.84 (d, J= 4.3 Hz, 1H), 2.74 (s, 2H), 2.59 - 2.53 (m, 2H), 2.17 (dd, j= 14.0, 7.1 Hz, 2H), 1.93 (t, 7= 11.5 Hz, 1H), 1.76 (s, 2H), 1.70 (s, 3H), 1.65 (s, 1H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, 7= 13.5 Hz, 1H). Example 20: Preparation of sodium 2-(((/?)-l-(((( /?, S,5A,6/?)-5-methoxy-4-((.2/?, /?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate
Figure imgf000082_0003
Step 1: Preparation of ( ?)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate
Boc
Boc
i
,N,
Figure imgf000082_0001
OH
Figure imgf000082_0002
[0188] To a solution of (R)-tert-butyl 3-hydroxypyrrolidine-l-carboxylate (500 mg, 2.67 mmol) in THF (20 mL) was added NaH (160 mg, 4.0 mmol, 60% in mineral oil) while cooling with an ice-bath. Thirty mins later, tert-butyl 2-bromoacetate (624 mg, 3.20 mmol) was added. The resulting mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The mixture was diluted with water and extracted with EtOAc (100 ml x 2). The organic layer was washed with H20 (100 mL), dried over anhydrous Na2S04, filtered and concentrated. The residue was purified by silica gel column chromatography (silica gel, 10 % EtOAc in PE) to give (A)-tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)pyrrolidine-l-carboxylate (450 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 302.
Step 2: Preparation of (R)-2-(pyrrolidin-3-yloxy)acetic acid
Figure imgf000083_0001
[0189] To a solution of (R)-tert- butyl 3-(2-(/er/-butoxy)-2-oxoethoxy)pyrrolidine-l- carboxylate (450 mg, 1.49 mmol) in DCM (1 mL) was added TFA (3 mL). The mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The reaction mixture was concentrated, and the residue was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 146.
Step 3: Preparation of 2-((( ?)-l-((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid
Figure imgf000083_0002
[0190] To a solution of (f?)-2-(pyrrolidin-3-yloxy)acetic acid (280 mg, 1.15 mmol) in CH3CN (20 mL) was added DIPEA (0.8 mL, 4.6 mmol) drop-wise at 0 °C. The mixture was stirred at 0 °C for 10 min, and Intermediate 1 (620 mg, 1.38 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by silica gel column chromatography (silica gel, 1 % ~ 5 % MeOH in DCM) and prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % ammonium hydroxide) to give 2-(((R)- l -((((3//,-/N5A6//)-5-methoxy-4-((2//,3//)-2-methyl-3-(3-methylbut-
2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3 -yl)oxy)acetic acid (240 mg) as a white solid. LC-MS (ESI) m/z (M+l): 454.
Step 4: Preparation of sodium 2-(((R)-l-((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-
3-yl)oxy)acetate
Figure imgf000084_0001
[0191] To a solution of 2-(((R)-l-((((3i?, S,,5,S,, d ?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3- yl)oxy)acetic acid (130 mg, 0.28 mmol) in MeCN (5 mL) was added dropwise a 0.1 N NaOH solution (2.5 mL, 0.25 mmol) at -60 °C with stirring. After lyophilization, sodium 5-((l- ((((3R, 4S, 5S, 6// )- 5 -m ethoxy-4 -((2//, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate (104 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 454. 1H NMR (400 MHz, DMSO-d6 ) d 5.35 (s, 1H), 5.19 (t, J= 7.5 Hz, 1H), 4.20 (s, 1H), 3.53 (s, 3H), 3.32 - 3.25 (m, 7H), 2.89 - 2.81 (m, 1H), 2.63 - 2.52 (m, 2H), 2.17 (d, J= 5.3 Hz, 2H), 2.02 - 1.89 (m, 2H), 1.87 - 1.75 (m, 3H), 1.69 (d, j= 13.3 Hz, 4H), 1.61 (s, 3H), 1.08 (d, j= 2.2 Hz, 3H), 1.01 (d, j= 13.8 Hz, 1H). Example 21: Preparation of sodium 2-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)acetate
Figure imgf000085_0001
Step 1: Preparation of ethyl 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetate
Figure imgf000085_0002
[0192] To a solution of tert-butyl azeti din-3 -ylcarbamate (344 mg, 2.0mmol) in
dichloromethane (10 mL) was added tert-butyl 2-oxoacetate (1.04 g, 4.0 mmol, 50% in toluene) dropwise at 0°C. The mixture was stirred at 0°C for 10 min, and sodium triacetoxyborohydride (848 mg, 4.0 mmol) was added at 0°C. The reaction was stirred at room temperature overnight under N2 atmosphere. The reaction mixture was then washed with water. The dichloromethane solution was dried over Na2S04, filtered and concentrated to give crude product. The crude product was purified by chromatography (silica gel, 1 to ~50 % ethyl acetate in petroleum ether) to give ethyl 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetate (410 mg) as a white solid. LC-MS (ESI) found: 259 [M+l]+.
Step 2: Preparation of 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetic acid
Figure imgf000085_0003
[0193] A mixture of ethyl 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetate (400 mg, l.55mmol), lithium hydroxide (74 mg, 3.lmmol) in THF (5mL) and water (2 mL) was stirred at room temperature overnight. The mixture was concentrated, and the aq. layer was washed with ethyl acetate (50 mL X 2). The aq. layer was then acidified to ~pH4 with 1M HC1 and extracted with DCM. The DCM layer was dried over Na2S04, filtered and concentrated under reduced pressure to give 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetic acid (310 mg) as a colorless oil, which was directly used in the next reaction without purification. LC-MS (ESI) found: 231 [M+l]+. Step 3: Preparation of 2-(3-aminoazetidin-l-yl)acetic acid trifluoroacetic acid salt
Figure imgf000086_0001
[0194] To a mixture of 2-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)acetic acid (310 mg, 1.35 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (3 mL) dropwise at 0°C. The reaction was stirred at room temperature for 1 hr. The mixture was concentrated under reduced pressure to give 2-(3-aminoazetidin-l-yl)acetic acid trifluoroacetic acid salt (260 mg) as yellow syrup, which was directly used in the next step without purification. LC-MS (ESI) found: 13 l[M+l]+.
Step 4: Preparation of 2-(3-(((((.?/?, S,5»S,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)acetic acid
Figure imgf000086_0002
[0195] 2-(3-Aminoazetidin-l-yl)acetic acid trifluoroacetic acid salt (297mg, 1.2 mmol) was coupled with Intermediate 1 (440 mg, l.Ommol) by following the General Procedure for Coupling. The reaction was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL x 2) followed by 5% sodium bicarbonate solution (20 mL). The organic layer was dried with Na2S04, filtered and concentrated to give crude product. The residue was purified by prep-HPLC (Method B: 0.1% NH4OH) to give 2-(3- (((((3R, 4S, 5S, 6R)-5 -m ethoxy-4 -((2 A 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)acetic acid (115 mg) as a white solid. LC-MS (ESI) found: 439 [M+l]+.
Step 5: Preparation of sodium 2-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)acetate
Figure imgf000087_0001
[0196] To a solution of 2-{2-{{{{{3R,4S,5S,6R)-S-mei\\oxy-4-{{2R, J/i)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- 1 - yl)acetic acid (115 mg, 0.26 mmol) in acetonitrile (2 mL) was added aq. NaOH (2.4 mL, 0.24 mmol) dropwise at -20°C. The mixture was freeze dried to give sodium 2-(((S)-l- ((((37?, 4S, 5S, 6/ )- 5 -m ethoxy-4 -((2/?, 37?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate (110 mg) as a white solid. LC-MS (ESI) found: 439 [M+l]+. 1H MR (400 MHz, DMSO- d) d 7.59 (d, J= 7.8 Hz, 1H), 5.30 (m, 1H), 5.19 (t, J= 7.2 Hz, 1H), 4.09 (m, 1H), 3.61 - 3.42 (m, 5H), 3.25 (m, 3H), 2.86 (m, 5H), 2.57 (d, J= 4.3 Hz, 1H), 2.26 - 2.06 (m, 2H), 2.02 - 1.73 (m, 3H), 1.69 (s, 3H), 1.61 (s, 3H), 1.20 - 0.83 (m, 4H).
Example 22: Preparation of Preparation of sodium 4-(((((( /?, S,5»S,6/?)-5-methoxy-4- ((2»V,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
Figure imgf000087_0002
Step 1: Preparation of 4-(((tert-butoxycarbonyl)amino)methyl)benzoic acid
Figure imgf000088_0001
[0197] 4-(Aminomethyl)benzoic acid (2.0 g, 13.2 mmol) was dissolved in a mixture of dioxane (20 mL) and 1 M NaOH (20 mL). The resulting solution was cooled to 0 °C in an ice bath and di-tert-butyl dicarbonate (2.9 g, 13.6 mmol) was added. The reaction mixture was stirred at r.t. for lh. TLC showed the reaction was complete. The solvent was concentrated under reduced pressure, and a 1 M NaHS04 solution was added to adjust the pH value to 2. The mixture was filtered to give 4-(((/c/7-butoxycarbonyl)a ino) ethyl jbenzoic acid (2.3 g) as a white solid. LC-MS (ESI) m/z (M+l): 252.
Step 2: Preparation of allyl 4-(((tert-butox carbonyl)amino)methyl)benzoate
Boc Boc
Figure imgf000088_0002
[0198] To a mixture of 4-(((/c/7-butoxycarbonyl)a ino) ethyl jbenzoic acid (2.3 g, 9.1 mmol) in MeOH (20 mL) and H20 (10 mL) was added KOH (510 mg, 9.1 mmol). The mixture was stirred at r.t. for 30 min. The mixture was concentrated, the residue was dissolved in DMF (10 mL) and allyl bromide (1.2 g, 9.56 mmol) was added. After the completion of the addition of allyl bromide, the reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The mixture was concentrated, and the residue was purified by flash
chromatography (silica gel, 5 % ~ 10 % ethyl acetate in petroleum ether) to give allyl 4-(((tert- butoxycarbonyl)amino)methyl)benzoate (2.1 g) as a yellow oil. LC-MS (ESI) m/z (M+l): 292. Step 3: Preparation of allyl 4-(aminomethyl)benzoate
Figure imgf000088_0003
[0199] To a solution of allyl 4-(((tert-butoxycarbonyl)amino)methyl)benzoate (300 mg, 1.03 mmol) in DCM (3 mL) was added TFA (1 mL) at r.t. After the completion of the addition, the mixture was stirred at r.t. for additional 1 h. The reaction mixture was concentrated, and the residue was used in next step without further purification. LC-MS (ESI) m/z (M+l): 191. Step 4: Preparation of allyl 4-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2V,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
Figure imgf000089_0001
[0200] To a solution of allyl 4-(aminomethyl)benzoate (350 mg, 1.83 mmol) in CH3CN (20 mL) was added DIPEA (1.3 mL, 7.32 mmol) drop-wise at 0 °C. After the completion of addition, the mixture was stirred at 0°C for another 10 min and Intermediate 1 (818 mg, 1.83 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated under vacuum while keeping the temperature below 40°C. The residue was purified by flash chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give allyl 4-((((((3/(-/A5k,d//)-5-methoxy-4-((2A3//)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate (350 mg) as a yellow oil. LC-MS (ESI) m/z (M+l):
500.
Step 5: Preparation of 4-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2V,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoic acid
Figure imgf000090_0001
[0201] To a solution of allyl 4-((((((3A, 4S,5S, df?)- 5 -m ethoxy-4 -((2 , 3f?)-2-methyl-3-3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate (350 mg, 0.7 mmol) in CEE CN/ Acetone was added Pd(PPh3)4 (87 mg, 0.7 mmol) and tetrahydro pyrrole (198 mg, 2.8 mmol ). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated and the residue was purified by prep-ffPLC (Cl 8, 0 ~ 90 % acetoniltrile in H20 with 0.1 % NH3 H20) to give 4- ((((((.3R, 4S, 5S, d/^)-5-methoxy-4-((25', 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)benzoic acid (120 mg) as a yellow oil. LC-MS (ESI) m/z (M+l): 460.
Step 6: Preparation of sodium 4-((((((J/?, V,5,V,6/?)-5-methoxy-4-((2V,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate
Figure imgf000090_0002
[0202] To a solution of 4-((((((3f?, S',5,S,, df?)-5-methoxy-4-((2S,,3f?)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl) benzoic acid (120 mg, 0.26 mmol) in MeCN (3 mL) was added a 0.1 N NaOH solution (2.3 mL, 0.23 mmol) dropwise at -60°C. After lyophilization, sodium A-((((((3R,4S,5S, 6R)-5-me\\\oxy-A-((2S,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)benzoate (94 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 460. 1H NMR (400 MHz, DMSO-d6 ) d 7.81 (d, J= 7.5 Hz, 2H), 7.66 (s, 1H), 7.21 (d, J = 7.7 Hz, 2H), 5.32 (s, 1H), 5.19 (t, J= 7.3 Hz, 1H), 4.21 (d, J= 6.1 Hz, 1H), 3.57 - 3.53 (m, 1H), 3.29 (s, 3H), 2.84 (d, j= 4.3 Hz, 1H), 2.57 (d, j= 4.3 Hz, 1H), 2.18 (dd, j= 13.2, 6.6 Hz, 2H), 1.86 (m, 4H), 1.70 (s, 3H), 1.62 (s, 1H), 1.61 (s, 3H), 1.10 (s, 3H), 1.06 (s, 1H).
Example 23: Preparation of sodium 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)benzoate
Figure imgf000091_0001
Step 1: Preparation of 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoic acid
Figure imgf000091_0002
[0203] To a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl lH-imidazole-l-carboxylate (250 mg, 0.66 mmol) in dry THF (5 mL) was added 4-aminobenzoic acid (109 mg, 0.80 mmol) and TFA (0.07 mL, 0.95 mmol) at 0°C. The reaction was then stirred at room temperature overnight. TLC showed the reaction was complete. The resulting mixture was diluted with DCM and washed with water. The combined organic layers were dried over anhydrous Na2S04 and concentrated. The crude product was purified by flash chromatography (silica gel, l~ 3 % MeOH in DCM) and pre-HPLC (Cl 8, 0 ~ 90 % acetoniltrile in H20 with 0.1 % NH3 H20) to give 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoic acid (141.4 mg) as a white solid. LC-MS (ESI) found: 446 [M+l]+.
Step 2: Preparation of sodium 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoate
Figure imgf000092_0001
[0204] To a solution of 4-(3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoic acid (68 mg, 0.15 mmol) in MeCN (1.4 mL) was added a 0.1 M NaOH (1.4 mL, 0.14 mmol) solution dropwise at 0°C with stirring. After lyophilization, sodium 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)benzoate (60 mg) was obtained as a white solid. LC-MS (ESI) found: 446 [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 9.51 (s, 1H), 7.70 (d, J= 8.5 Hz, 2H), 7.30 (d, J = 8.2 Hz, 2H), 5.39 (s, 1H), 5.16 (m, J= 7.4 Hz, 1H), 3.55 (m, J= 10.8, 2.6 Hz, 1H), 3.28 (s, 3H), 2.82 (d, J= 4.4 Hz, 1H), 2.55 (d, 7= 4.4 Hz, 1H), 2.51 - 2.47 (m, 1H), 2.22 - 2.07 (m, 2H), 1.91 (m, 7= 29.4, 16.6, 5.2 Hz, 3H), 1.73 (m, 7= 28.0, 14.1 Hz, 1H), 1.67 (s, 3H), 1.57 (s,
3H), 1.06 (s, 3H), 1.02 (d, J= 4.3 Hz, 1H).
Example 24: Preparation of sodium 6-((((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate
Figure imgf000093_0001
Step 1: Preparation of 6-(aminomethyl)nicotinic acid
Figure imgf000093_0002
[0205] To a solution of 6-Cyanonicotinic acid (1 g, 6.7 mmol) in MeOH (20 mL) was added Pd(OH)2 (10% Pd in carbon, 0.1 g). The mixture was stirred at r.t. under a hydrogen atmosphere overnight. Then the mixture was diluted with H20 (10 mL), the catalyst was filtered, and the filtrate was concentrated in vacuo to give 6-(aminomethyl)nicotinic acid (750 mg), which was used in the next step without further purification.
Step 2: Preparation of 6-(((tert-butox carbonyl)amino)methyl)nicotinic acid
Figure imgf000093_0003
[0206] To a solution of 6-(aminomethyl)nicotinic acid (750 mg, 4.92 mmol) in THF (50 mL) was added 50 mL of 1N aq NaOH and Boc20 (1.2 g, 5.66 mmol) at 25 °C. After the completion of addition, the mixture was stirred overnight. Then the mixture was diluted with water (25 mL) and brine (25 mL), acidified slowly to pH = 3 by 1N aq HC1, and extracted with EtOAc (3 x 20 mL). The combined organic extracts were dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (silica gel, 5 % ~ 10 % ethyl acetate in petroleum ether) to give 6-(((tert- butoxycarbonyl)amino)methyl)nicotinic acid (720 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 253.
Step 3: Preparation of allyl 6-(((tert-butox carbonyl)amino)methyl)nicotinate
Figure imgf000094_0001
[0207] To a solution of 6-(((tert-butoxycarbonyl)amino)methyl)nicotinic acid (720 mg, 2.85 mmol) in DMF (8 mL) was added Cs2C03 (1.4 g, 4.28 mmol) at rt. After completion of addition, the reaction mixture was stirred for another 10 min, and then allylbromide (518 mg, 4.28 mmol) and a catalytic amount of KI were added. After completion of addition, the reaction mixture was heated to l00°C for 4h. Upon cooling of the reaction, water (50 mL) was added, and the aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic extracts were dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (silica gel, 5 % ~ 10 % ethyl acetate in petroleum ether) to give allyl 6-(((tert-butoxycarbonyl)amino)methyl)nicotinate (660 mg) as a white solid. LC-MS (ESI) m/z (M+l): 293.
Step 4: Preparation of allyl 6-(aminomethyl)nicotinate
Figure imgf000094_0002
[0208] To a solution of allyl 6-(((tert-butoxycarbonyl)amino)methyl)nicotinate (660 mg, 2.26 mmol) in DCM (4 mL) was added TFA (2 mL). The mixture was stirred at r.t. for 1 h. TLC showed the reaction was complete. The reaction mixture was concentrated and the residue was used in next step without further purification. LC-MS (ESI) m/z (M+l): 193.
Step 5: Preparation of allyl 6-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate
Figure imgf000095_0001
[0209] To a solution of allyl 6-(aminomethyl)nicotinate (660 mg, 3.43 mmol) in CH3CN (20 mL) was added DIPEA (2.4 mL, 13.6 mmol) drop-wise at 0°C. The mixture was stirred at 0 °C for another 10 min and Intermediate 1 (1.3 g, 3.1 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by flash chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give allyl 6- (((((( 3R , 4S, 5S, 6/^)-5-methoxy-4-((2k, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinate (650 mg) as a yellow oil. LC- MS (ESI) m/z (M+l): 501.
Step 6: Preparation of 6-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinic acid
Figure imgf000095_0002
[0210] To a solution of allyl 6-((((((3f?,4ri,,5ri,,6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinate (650 mg, 1.29 mmol) in CH3CN/acetone (10 mL) was added Pd(PPh3)4 (161 mg, 0.13 mmol) and tetrahydropyrrole (366 mg, 5.16 mmol). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated, and the residue was purified by prep-HPLC (C18, 0 ~ 90 % acetoniltrile in H20 with 0.1 % NH3TT20) to give 6-((((((3/(-/N5.5', 6//)-5-methoxy-4-((2A3//)-2-methyl-3-(3-methylbut-2-en- l -yl)oxiran-2-yl)- l - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinic acid (102 mg) as a white solid. LC-MS (ESI) m/z (M+l): 461.
Step 7: Preparation of sodium 6-((((((3/?, S,5A,6/?)-4-((2/?,3/?)-3-ethyl-2-methyloxiran-2- yl)-5-methoxy-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinate
Figure imgf000096_0001
[0211] To a solution of 6-((((((3f?, ,S,,5S,, 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)nicotinic acid (102 mg, 0.22 mmol) in MeCN (3 mL) was added 0.1 N NaOH solution (2.0 mL, 0.2 mmol) dropwise at -60°C. After lyophilization, sodium 6-((((((3A, 4S, 5S, 6R)-4-((2S, 3R)~ 3 -ethyl-2-methyloxiran-2-yl)-5-methoxy- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinate (73 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 461. 1H NMR (400 MHz, DMSO-d6 ) d 8.84 (s, 1H),
8.04 (dd, 7= 7.9, 1.9 Hz, 1H), 7.65 (t, J= 5.9 Hz, 1H), 7.17 (d, J= 7.9 Hz, 1H), 5.32 (s, 1H), 5.20 (t, J= 7.5 Hz, 1H), 4.27 (d, J= 6.0 Hz, 2H), 3.55 (dd, J= 10.6, 2.3 Hz, 1H), 3.30 (s, 3H),
2.85 (d, J= 4.3 Hz, 1H), 2.62 - 2.54 (m, 1H), 2.19 (m, 2H), 1.99 - 1.77 (m, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.10 (s, 3H), 1.05 (s, 1H).
Example 25: Preparation of sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3- carboxylate
Figure imgf000096_0002
Step 1 : l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3-carboxylic acid
Figure imgf000097_0001
[0212] To a mixture of azetidine-3 -carboxylic acid (202 mg, 2.0 mmol) in CH3CN (5 mL) was added DIPEA (650 mg, 5 mmol) drop-wise at 0~5°C. The mixture was stirred at 0~5°C for 10 min, then (J/i,-/V,5,.y, ri/i)-5-methoxy-4-((2/i, J/i)-2-methyl-3-(3-methylbut-2-en- 1 -yl)oxiran- 2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 900 mg, 2.0 mmol) was added to the mixture in portions at 0°C under N2 atmosphere. The reaction mixture was stirred at 25°C for another 16 hrs. TLC showed the reaction was complete. The solvent was removed under vacuum below 40°C, then diluted with DCM (60 mL) and washed with ammonium acetate buffer (pH~4, 15 mL x 2). The aqueous layer was extracted with DCM (10 mL x 2), dried over anhydrous Na2S04 and concentrated to give a yellow oil, which was purified by prep-HPLC (Method B: 0.1 % NH OH) to give (385 mg) l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidine-3-carboxylic acid as a white solid. LC-MS (ESI) found: 424 [M+l]+. Step 2: Preparation of sodium l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3- carboxylate
Figure imgf000097_0002
[0213] To a solution of l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3 -carboxylic acid (123 mg, 0.3 mmol) in MeCN (2 mL) was added a 0.1 N NaOH solution (2.7 mL, 0.9 eq) dropwise at -60°C with stirring. After lyophilization, (140 mg) of the title compound was obtained as a white solid. LC-MS (ESI) found: 410 [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 5.36 (d, J= 5.5 Hz, 1H), 5.19 (s, 1H), 4.20 (d, J= 22.1 Hz, 1H), 3.62 - 3.41 (m, 3H), 3.29 (d, J = 5.0 Hz, 4H), 2.91 - 2.80 (m, 1H), 2.72 - 2.53 (m, 2H), 2.30 - 2.08 (m, 2H), 1.98 (dd, J = 15.9, 11.1 Hz, 2H), 1.81 (dd, J= 11.1, 2.9 Hz, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.08 (d, J= 3.5 Hz, 3H), 1.01 (d, J= 13.5 Hz, 1H).
Example 26: Preparation of sodium 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
Figure imgf000098_0001
Step 1: Preparation of tert-butyl (R)-2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate
Figure imgf000098_0002
[0214] The mixture of tert-butyl (R)-pyrrolidin-3-ylcarbamate (400 mg, 2.15 mmol), tert- butyl 2-bromoacetate (430 mg, 2.26 mmol) and K2C03 (593 mg, 4.30 mmol) in DMF (10 mL) was stirred at 50°C for l6h. The slurry was diluted by EA, and washed by water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give tert-butyl (R)-2-(3-((tert- butoxycarbonyl)amino)pyrrolidin-l-yl)acetate (600 mg) as a white solid. LC-MS (ESI) found: 301 [M+H] +
Step 2: Preparation of(R)-2-(3-aminopyrrolidin-l-yl)acetic acid
Figure imgf000098_0003
[0215] To a solution of tert-butyl (R)-2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate (600mg, 2.00 mmol) in DCM (2 mL) was added TFA (1 mL). The reaction mixture was then stirred at room temperature for 3 hours. Concentration gave crude (R)-2-(3- aminopyrrolidin-l-yl)acetic acid TFA salt (560 mg) as a brown oil. LC-MS (ESI) found:306 [M+l] +.
Step 3: Preparation of 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetic acid
Figure imgf000099_0001
Intermediate 1 [0216] Under a N2 atmosphere, to a solution of (R)-2-(3-aminopyrrolidin-l-yl)acetic acid
2,2,2-trifluoroacetate salt (crude, 560 mg, 1.0 mmol) and diisopropylethylamine (259 mg, 2.10 mmol) in CH3CN (5 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 300 mg, 0.67 mmol) in CH3CN (lmL) drop-wise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 h. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)pyrrolidin- 1 -yl)acetic acid (50 mg) as a colorless oil. LC-MS (ESI) found: 453 [M+l] +.
Step 4: Preparation of sodium 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
Figure imgf000099_0002
[0217] To a solution of 2-((R)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)pyrrolidin- 1 - yl)acetic acid (50 mg, 0.11 mmol) in CH3CN (1 mL) was added a 0.1 N NaOH (1.6 mL, 0.099 mmol) solution at -60°C with stirring. After lypholization, sodium 2-((R)-3-(((((3R,4S,5S,6R)- 5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan- 6-yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate salt (51 mg) was obtained as white solid. 1HNMR (400MHz, DMSO-76) 57.86 (d, J= 7.3 Hz, 1H), 5.27 (s, 1H), 5.18 (d, J= 7.3 Hz,
1H), 3.91 (s, 1H), 3.52 (d, j= 10.4 Hz, 1H), 3.28 (s, 3H), 2.85 - 2.80 (m, 2H), 2.76 (s, 2H),
2.70 (d, J= 7.8 Hz, 1H), 2.57 - 2.53 (m, 2H), 2.46 (d, 7= 9.1 Hz, 2H), 2.20 - 2.14 (m, 2H), 1.97 - 1.74 (m, 5H), 1.71 (s, 3H), 1.61 (s, 3H), 1.57 - 1.51 (m, 1H), 1.09 (s, 3H), 1.04 (d, J =
14.1HZ, 1H).
Example 27: Preparation of sodium 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
Figure imgf000100_0001
Step 1: Preparation of (S)-tert-butyl 2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate
0
H 1 — ( I Br^ CoJ I
N <
HN
V-NH
Boc DIPEA, MeCN B RoncnrO ^J vU O<
[0218] To a solution of (S)-tert-butyl pyrrolidin-3-ylcarbamate (400 mg, 2.15 mmol) in
MeCN (8 mL) was added DIPEA (0.53 mL, 3.22 mmol) and a solution of tert-butyl 2- bromoacetate (0.31 mL, 2.15 mmol) in CH3CN (2mL) dropwise. The reaction mixture was stirred at r.t. overnight. The mixture was diluted with water and extracted with EtOAc (15 mL x2). The combined organic layers were dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (silica gel, 0 to 2 % MeOH in DCM) to give (S)-tert-butyl 2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l-yl)acetate (600 mg) as yellow oil. LC-MS (ESI) found: 454 [M+l]+: 301.
Step 2: Preparation of (S)-2-(3-aminopyrrolidin-l-yl)acetic acid
Figure imgf000101_0001
[0219] To a solution of (S)-tert-butyl 2-(3-((tert-butoxycarbonyl)amino)pyrrolidin-l- yl)acetate (600 mg, 2.0 mmol) in DCM (3 mL) was added TFA (1.5 mL) at 0°C. The mixture was stirred at r.t. for 1 h. The reaction mixture was concentrated and the residue was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 145.
Step 3: Preparation of sodium 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate
Figure imgf000101_0002
[0220] (S)-2-(3-aminopyrrolidin-l-yl)acetic acid (192 mg, 1.33 mmol) in CH3CN (5 mL) and water (lmL) was coupled with Intermediate 1 (500 mg, 1.11 mmol) by following the General Procedure for Coupling. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by prep-HPLC (Method B: 0.1% MEOH) to give 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)pyrrolidin- 1 - yl)acetic acid(60 mg, 10%). The acid was dissolved in MeCN (1 mL) and aq. NaOH (1.2 mL, 0.1M) was added at 0°C. After lyophilization, sodium 2-((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate (60 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 453. 1H NMR (400 MHz, DMSO- d) d 7.85 (d, J= 7.5 Hz, 1H), 5.27 (s,
1H), 5.20 (t, j= 7.4 Hz, 1H), 3.89 (s, 1H), 3.52 (dd, j= 10.7, 2.6 Hz, 1H), 3.29 (s, 3H), 2.88 - 2.64 (m, 5H), 2.55 (dd, J= 7.8, 5.4 Hz, 2H), 2.44 (dd, J= 13.3, 8.2 Hz, 2H), 2.25 - 2.11 (m, 2H), 2.04 - 1.65 (m, 8H), 1.64 - 1.47 (m, 4H), 1.14 - 0.97 (m, 4H). Example 28: Preparation of sodium 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate
Figure imgf000102_0001
Step 1: Preparation of ethyl 2-(4-((tert-butoxycarbonyl)amino)piperidin-l-yl)acetate
Figure imgf000102_0002
[0221] To a mixture of tert-butyl piperidin-4-yl carbamate (500 mg, 2.49 mmol) in DMF (20 mL) were added ethyl 2-bromoacetate (500 mg, 2.99 mmol) and K2C03 (687 mg, 4.98 mmol). The mixture was stirred at r.t. overnight. Water (50 mL) was added, and the mixture was extracted with ethyl acetate (50 mL). The organic extract was washed with brine, dried over anhydrous Na2S04, and evaporated under vacuum. The residue was purified by flash chromatography (silica gel, 1 % ~ 20 % ethyl acetate in petroleum ether) to give ethyl 2-(4- ((tert-butoxycarbonyl)amino)piperidin-l-yl)acetate (490 mg) as a white solid. LC-MS (ESI) found: 287 [M+l]+. Step 2: Preparation of ethyl 2-(4-aminopiperidin-l-yl)acetate
Bo
Figure imgf000103_0001
[0222] To a solution of ethyl 2-(4-((tert-butoxycarbonyl)amino)piperidin-l-yl)acetate (490 mg, 1.471 mmol) in DCM (2 mL) was added TFA (3 mL). The mixture was stirred at r.t. for 2 h. The reaction mixture was concentrated, and the crude product was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 187
Step 3: Preparation of ethyl 2-(4-(((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate
Figure imgf000103_0002
[0223] To a solution of ethyl 2-(4-aminopiperidin-l-yl)acetate (320 mg, 1.71 mmol) in CH3CN (20 mL) was added DIPEA (1.2 mL, 6.8 mmol) dropwise at 0°C. The mixture was stirred at 0°C for 10 min and Intermediate 1 (764 mg, 1.71 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. After the reaction complete, the mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by flash chromatography (silica gel, 1 % ~ 5 % MeOH in DCM) to give ethyl 2-(4-(((((3A, S,,5S,,di?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut-2- en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)piperidin- 1 -yl)acetate (510 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 495.
Step 4: Preparation of 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetic acid
Figure imgf000104_0001
[0224] To a solution of ethyl 2-(4-(((((3f?, ,S,,5S,, 6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)piperidin- 1 - yl)acetate (510 mg, 1.05 mmol) in THF/H20 (3 mL/l mL) was added LiOH (433 mg, 10.3 mmol). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated and the residue was purified by prep-HPLC (Method B: 0.1% NH4OH) to 2-(4-(((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetic acid (60 mg) as a white solid. LC-MS (ESI) m/z (M+l): 467.
Step 5: Preparation of sodium 2-(4-(((((3/?, ,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)piperidin-l-yl)acetate
Figure imgf000104_0002
[0225] To a solution of 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)piperidin- 1 - yl)acetic acid (60 mg, 0.12 mmol) in MeCN (1 mL) was added dropwise a 0.1 N NaOH solution (1.1 mL, 0.12 mmol) at -60°C with stirring. After lyophilization, sodium 2-(4- (((((3R, 4S, 5S, 6R)-5 -m ethoxy-4 -((2 A 3i?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)piperidin-l-yl)acetate (40 mg) was obtained as a white solid. LC-MS (ESI) m/z (M+l): 467. 1H NMR (400 MHz, DMSO- d) d 7.00 (s, 1H), 5.23 (d, J= 29.6 Hz, 2H), 3.52 (dd, J= 10.7, 2.7 Hz, 1H), 3.28 (s, 3H), 3.22 (d, J= 8.3 Hz, 2H), 2.83 (d, J= 4.1 Hz, 3H), 2.66 (s, 2H), 2.56 (d, J= 4.3 Hz, 1H), 2.18 (d, J= 7.0 Hz, 2H), 2.10 - 1.73 (m, 6H), 1.71 (s, 3H), 1.66 (s, 2H), 1.61 (s, 3H), 1.40 (s, 2H), 1.09 (s, 2H), 1.06 (s, 1H). Example 29: Preparation of sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)phosphonate
Figure imgf000105_0001
Step 1: Preparation of l-benzhydrylazetidin-3-yl methanesulfonate
Figure imgf000105_0002
[0226] To a mixture of l-benzhydrylazeti din-3 -ol (10.0 g, 41.79 mmol) in DCM (100 mL) was added TEA (12.7 g, 125.37 mmol) and MsCl (8.6 g, 75.22 mmol) at 0°C. Then the reaction mixture was warmed to r.t. and stirred for 3 hours. After the reaction was complete, the mixture was washed with water and the water layer was back-extracted with DCM. The combined organic layers were dried over Na2S04, and concentrated. The residue was purified by flash chromatography (silica gel, 2 % - 5 % MeOH in DCM) to give l-benzhydrylazetidin- 3-yl methanesulfonate (11.0 g) as a yellow solid. LC-MS (ESI) found: 318 [M+l]+.
Step 2: Preparation of diethyl (l-benzhydrylazetidin-3-yl)phosphonate
Figure imgf000106_0001
[0227] A solution of l-benzhydrylazeti din-3 -yl methanesulfonate (10 g, 31.54 mmol) in P(OEt)3 (52 g, 315.4 mmol) was heated to l50°C and stirred at this temperature for 8 hours. After the reaction complete, it was cooled to r.t. and purified by flash chromatography (silica gel, 2 % -5 % MeOH in DCM) to give diethyl (l-benzhydrylazetidin-3-yl)phosphonate (17.0 g) as a yellow oil. LC-MS (ESI) found: 360 [M+l]+.
Step 3: Preparation of ethyl hydrogen (l-benzhydrylazetidin-3-yl)phosphonate
Figure imgf000106_0002
[0228] To a mixture of diethyl (l-benzhydrylazeti din-3 -yl)phosphonate crude (10 g, 27.86 mmol) in EtOH (80 mL) was added 2.0M NaOH (80 mL, 167.13 mmol) at r.t. Then the reaction mixture was heated to 80°C and stirred for 8 hours. After the reaction was complete, the mixture was adjusted to pH = 2~3 using 2M HC1 and extracted with DCM 3 times. The DCM extracts were dried over Na2S04 and concentrated. The residue was purified by prep- HPLC (Method B: 0.1% NH4OH) to give ethyl hydrogen (l-benzhydrylazeti din-3 - yl)phosphonate (1.8 g) as a white solid. LC-MS (ESI) found: 332 [M+l]+.
Step 4: Preparation of ethyl hydrogen azetidin-3-ylphosphonate
Figure imgf000106_0003
[0229] To a mixture of ethyl hydrogen (l-benzhydrylazetidin-3-yl)phosphonate (800 mg, 2.41 mmol) in EtOH (16 mL) was added Pd/C (10%, 400 mg) at r.t. Then the reaction mixture was stirred at r.t. for 2 hours under H2. After the reaction complete, it was filtered and washed with EtOH. The filtrate was concentrated to give crude ethyl hydrogen azeti din-3 - ylphosphonate (700 mg) as an oil. LC-MS (ESI) found: 166 [M+l]+. Step 5: Preparation of ammonium ethyl (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3-
(ethyl(hydroxy)phosphonyl)azetidine-l-carboxylate
Figure imgf000107_0001
[0230] To a solution of crude ethyl hydrogen azeti din-3 -ylphosphonate (700 mg) in MeCN (6 mL) was added DIPEA (267 mg) at 0°C. Then (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (617 mg) was added at 0°C. The reaction mixture was then stirred for 2 hours at r.t. After the reaction complete, the mixture was concentrated under reduced pressure to remove MeCN and diluted by water. The water mixture was extracted with DCM. The combined organic layers was dried over Na2S04, concentrated to give the crude DIPEA salt (3R,4S,5S,6R)-5-methoxy-
4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3- (ethyl(hydroxy)phosphonyl)azetidine-l-carboxylate (1.3 g) as colorless oil. Then, 360 mg of the aforementioned crude material was purified by prep-HPLC (Method B: 0.1% NH4OH) ammonium ethyl (l-((((3i?,4ri',5ri', 6/?)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)phosphonate (80 mg) as an oil. LC-MS (ESI) found: 474 [M+l]+. 1H MR (400 MHz, DMSO) d 7.38 (s, 4H), 5.28 (s, 1H), 5.19 (s, 1H), 3.85 (s, 4H), 3.72 - 3.64 (m, 2H), 3.50 (s, 1H), 3.28 (s, 3H), 2.84 (d, J= 4.3 Hz, 1H), 2.57 (d, J= 4.3 Hz, 2H), 2.17 (d, J= 6.6 Hz, 2H), 1.99 - 1.88 (m, 1H), 1.81 - 1.66 (m, 6H), 1.61 (s, 3H), 1.12 - 1.04 (m, 6H), 1.04 - 0.98 (m, 1H).
Step 6: Preparation of sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)phosphonate
Figure imgf000107_0002
[0231] To a mixture of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl 3 -(ethyl (hydroxy)phosphonyl)azeti dine- 1- carboxylate (40 mg, 0.084 mmol) in MeCN (0.8 mL) was added 0.1M NaOH (0.76 mL, 0.076 mmol) at r.t. Then the mixture was freeze-dried to give sodium ethyl (l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)phosphonate (41 mg) as a white solid. LC-MS (ESI) found: 496 [M+l]+. 1H NMR (400 MHz, DMSO- d) d 5.31 - 5.23 (m, 1H), 5.22 - 5.16 (m, 1H), 3.95 - 3.72 (m, 4H), 3.71 - 3.58 (m, 2H), 3.54 - 3.46 (m, 1H), 3.27 (s, 3H), 2.87 - 2.81 (m, 1H), 2.56 (s, 2H), 2.46 - 2.38 (m, 1H), 2.24 - 2.10 (m, 2H), 1.99 - 1.90 (m, 1H), 1.70 (s, 6H), 1.61 (s, 3H), 1.10 - 1.03 (m, 6H), 1.03 - 0.97 (m, 1H).
Example 30: Preparation of sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)propanoate
Figure imgf000108_0001
Step 1: Preparation of methyl 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoate
Figure imgf000108_0002
[0232] To a solution of tert-butyl azeti din-3 -ylcarbamate (416 mg, 2.0 mmol) and K2C03 (560 mg, 4.0 mmol) in DMF (10 mL) was added methyl 3-bromopropanoate (330 mg, 2.0 mmol) dropwise at 0°C. The mixture was stirred for 30 min at 0°C. Then the reaction was stirred at room temperature overnight under a N2 atmosphere. After the reaction was complete water was added and the water later was extracted with DCM. The dichloromethane layer was dried over anhydrous Na2S04, filtered and concentrated to give crude product. The crude product was purified by chromatography (silica gel, 1 to -10 % MeOH in DCM) to give methyl 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoate (350 mg) as a white solid. LC-MS (ESI) found: 259 [M+H]+.
Step 2: Preparation of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoic acid
Figure imgf000109_0001
[0233] A mixture of methyl 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoate (350 mg, 1.35 mmol) and lithium hydroxide (74 mg, 3.1 mmol) in THF (5mL) and water (2 mL) was stirred at room temperature overnight. TLC showed the reaction was complete, and the mixture was concentrated to remove THF. Then the water solution was washed with ethyl acetate (50 mL x 2), acidified to -pH = 4 with 1M HC1 and extracted with DCM. The DCM layer was dried over anhydrous Na2S04, filtered and concentrated under reduced pressure to give 3-(3- ((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoic acid (280 mg) as a colorless oil, which was used directly in the next reaction without further purification. LC-MS (ESI) found: 245 [M+H]+.
Step 3: Preparation of 3-(3-aminoazetidin-l-yl)propanoic acid trifluoroacetic acid salt
Figure imgf000109_0002
[0234] To a mixture of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)propanoic acid (280 mg, 1.2 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (3 mL) dropwise at 0°C. The reaction was stirred at room temperature for 1 h. TLC showed the reaction was complete, and the mixture was concentrated under reduced pressure to give 3-(3-aminoazetidin- l-yl)propanoic acid trifluoroacetic acid salt (280 mg) as yellow syrup, which was used directly in the next step without further purification. LC-MS (ESI) found: 145[M+H]+.
Step 4: Preparation of 2-(3-(((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)acetic acid
Figure imgf000110_0001
[0235] 3-(3-aminoazetidin-l-yl)propanoic acid trifluoroacetic acid salt (280 mg, 1.0 mmol) was coupled with Intermediate 1 (440 mg, 1.0 mmol) by following the General Procedure for Coupling. The reaction was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL x 2) . The organic layer was dried with anhydrous Na2S04, filtered and concentrated to give crude product. The residue was purified by prep- HPLC (Method B: 0.1% NH4OH) to give 3-(3-(M3R,JS,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)propanoic acid (105 mg) as a white solid. LC-MS (ESI) found: 453 [M+H]+.
Step 5: Preparation of sodium 2-(3-(((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)acetate
Figure imgf000110_0002
[0236] To a solution of 3-(3-(((((3f?,4,S,,5S,,d/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- 1 - yl)propanoic acid (105 mg, 0.23 mmol) in acetonitrile (1 mL) was added aq. NaOH (2.4 mL, 0.24 mmol, 1M) dropwise at -20°C. After completion of the reaction, the mixture was freeze dried to give sodium 3-(3-(((((3f?,4ri,,5ri,,6/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)propanoic acid (115 mg) as a white solid. LC-MS (ESI) found: 453 [M+H]+. 1H NMR (400 MHz, DMSO- d) d 7.64 (d, J= 7.8 Hz, 1H), 5.37 (d, J= 26.7 Hz, 1H), 5.26 (t, J= 7.2 Hz, 1H), 4.08 (dd, J= 14.3, 7.0 Hz, 1H), 3.59 (dd, J= 10.8, 2.6 Hz, 2H), 3.47 (s, 3H), 3.34 (s, 3H), 2.91 (d, J = 4.2 Hz, 1H), 2.77 (dt, J= 13.6, 6.9 Hz, 2H), 2.64 (d, J= 4.3 Hz, 1H), 2.55 - 2.47 (m, 2H), 2.32 - 2.16 (m, 2H), 2.13 - 1.81 (m, 5H), 1.78 (s, 3H), 1.73 (d, J= 7.4 Hz, 1H), 1.68 (s, 3H), 1.31 - 0.99 (m, 4H).
Example 31: Preparation of sodium 2-(4-((((J ?, V,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate
Figure imgf000111_0001
Figure imgf000111_0003
Step 1: Preparation of tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)piperazine-l-carboxylate
Figure imgf000111_0002
[0237] To a solution of tert-butyl piperazine- l-carboxylate (1.5 g, 8.1 mmol) in THF (50 mL) were added tert-butyl 2-bromoacetate (1.4 mL, 9.6 mmol) and TEA (1.1 mL, 8.1 mmol). The mixture was stirred at r.t. for 2 hours. TLC showed the reaction was complete. Water (20 mL) was added and the mixture was extracted with ethyl acetate (50 mL). The organic extract was washed with brine, dried over anhydrous Na2S04, and concentrated under vacuum. The residue was purified by flash chromatography (silica gel, 1 % ~ 20 %ethyl acetate in petroleum ether) to give tert-butyl 4-[2-(tert-butoxy)-2-oxoethyl]piperazine- l-carboxylate (2.1 g) as a white solid. LC-MS (ESI) m/z (M+l): 301. Step 2: Preparation of 2-(piperazin-l-yl)acetic acid
Figure imgf000112_0001
[0238] To a mixture of tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)piperazine-l-carboxylate (500 mg, 1.6 mmol) in DCM (1 mL) was added TFA (3 mL). The reaction mixture was stirred at room temperature for 16 hrs. TLC showed the reaction was complete. The resulting mixture was concentrated to give crude product, which was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 145.
Step 3: Preparation of 2-(4-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetic acid
Figure imgf000112_0002
[0239] To a solution of 2-(piperazin-l-yl)acetic acid, TFA salt (260 mg, 1.8 mmol) in CH3CN (20 mL) was added DIPEA (1.2 mL, 7.2 mmol) drop-wise at 0°C. The mixture was stirred at 0°C for 10 min and Intermediate 1 (806 mg, 1.8 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by prep-HPLC (Cl 8,0 ~ 90 %
acetonitrile in H20 with 0.1 % formic acid) to give 2-(A-((((3R,4S,5S,6R)-S-me\\\oxy-4- ((2i?,3i?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetic acid (320 mg) as a white solid. LC-MS (ESI) m/z (M+l): 453.
Step 4: Preparation of sodium 2-(4-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetate
Figure imgf000113_0001
[0240] To a solution of 2-(4-((((3R, 4S,5S, 6/?)-5-methoxy-4-((2f?, 3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin- 1 -yl)acetic acid (300 mg, 0.67 mmol) in MeCN (2 mL) was added 0.1 N NaOH solution (5.9 mL, 0.0.59 mmol) dropwise at -60°C with stirring. The mixture was lyophilized to give sodium 2-(4- ((((37?, 4S, 5S, 6R)-5-methoxy-4-((2R, 3i?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l-yl)acetate (260 mg) as a white solid. LC-MS (ESI) m/z (M+l): 453. 1H NMR (400 MHz, DMSO-d6 ) d 5.36 (s, 1H), 5.19 (s, 1H), 3.53 (d, J = 10.8 Hz, 1H), 3.28 (s, 4H), 2.85 (s, 1H), 2.68 (s, 2H), 2.57 (s, 2H), 2.39 (d, J = 24.7 Hz, 4H), 2.17 (s, 2H), 1.94 (s, 1H), 1.78 (d, J = 11.7 Hz, 2H), 1.70 (s, 4H), 1.61 (s, 3H), 1.07 (s, 3H),
1.01 (d, J = 13.3 Hz, 1H).
Example 32: Preparation of sodium 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
Figure imgf000113_0002
Step 1: Preparation of ethyl 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol- l-yl)acetate
Boc
Figure imgf000113_0003
Figure imgf000113_0005
CU(OAC)2, Sodium ascorbate
t-BuOH/H20
Figure imgf000113_0004
[0241] A mixture of tert-butyl prop-2-yn-l-ylcarbamate (600 mg, 3.86 mmol), ethyl 2- azidoacetate (648 mg, 5.02 mmol), Cu(OAc)2 (230 mg, 1.15 mmol) and sodium ascorbate (384 mg, 1.93 mmol) in t-BuOH/H20 (10 mL, 1/1, v/v) was stirred at 30°C overnight. TLC showed the reaction was complete. The t-BuOH was removed under vacuum. The aqueous layer was extracted with ethyl acetate (5 mL x 2). The combined organic layers were dried over anhydrous Na2S0 and concentrated. The residue was purified by column chromatography (silica gel, 10 to 50 % ethyl acetate in petroleum ether) to give ethyl 2-(4-(((tert- butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate (850 mg). LC-MS (ESI) found: 285[M+l]+.
Step 2: Preparation of 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l- yl)acetic acid
Figure imgf000114_0001
[0242] To a mixture of ethyl 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l- yl)acetate (500 mg, 1.75 mmol) in THF/MeOH (6 mL, 1/1, v/v) was added LiOH (2.6 mL, 5.2 mmol, 2M). The reaction mixture was stirred at room temperature for 1 h. TLC showed the reaction was complete. The organic volatiles were removed under vacuum. The aqueous layer was adjusted pH to 3 with diluted H2S04 and extracted with CHCl3/i-PrOH (5 ml x 10, 3/1, v/v). The combined organic layers were dried over Na2S04 and concentrated to give 2-(4- (((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetic acid (450 mg) as a colorless oil. LC-MS(ESI) found: 257 [M+l]+.
Step 3: Preparation of 2-(4-(aminomethyl)-lH-l,2,3-triazol-l-yl)acetic acid
Figure imgf000114_0002
[0243] To a solution of 2-(4-(((tert-butoxycarbonyl)amino)methyl)-lH-l,2,3-triazol-l- yl)acetic acid (450 mg, 1.75 mmol) in DCM (4.5 mL) was added TFA (1.5 mL). After the completion of addition, the reaction mixture was stirred at room temperature overnight. LCMS showed the reaction was complete. The mixture was concentrated to give 2-(4-(aminomethyl)- lH-l,2,3-triazol-l-yl)acetic acid TFA salt (800 mg) as a brown oil. LC-MS (ESI) found: 157 [M+l] +. Step 4: Preparation of sodium 2-(4-((((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
Figure imgf000115_0001
[0244] To a solution of 2-(4-(aminomethyl)-lH-l,2,3-triazol-l-yl)acetic acid (105 mg, 0.67 mmol) in CH3CN (3 mL) and water (lmL) was added DIPEA (0.29 mL, 1.78 mmol) drop-wise at 0°C. The mixture was stirred at 0°C for 10 min and Intermediate 1 (200 mg, 0.45 mmol) was added. The reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by prep-HPLC (Cl 8, 0 ~ 90 % MeCN in H20 with 0.1 % formic acid) to give 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)- lH-l,2,3-triazol-l-yl)acetic acid (70 mg, 0.15 mmol). The acid was dissolved in MeCN (1 mL) and aq. NaOH (1.4 mL, 0.1M) was added at 0°C. After lyophilization, sodium 2-(4- {[({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)amino]methyl}-lH-l,2,3-triazol-l-yl)acetate (70 mg) was obtained as a white solid. LC-MS (ESI) m/z(M+l): 465. 1H NMR (400 MHz, DMSO-d6 ) d 8.53 (s, 1H), 7.64 (d, J = 27.7 Hz, 1H), 5.31 (s, 1H), 5.20 (t, J = 7.2 Hz, 1H), 4.54 (s, 2H), 4.18 (t, J = 13.1 Hz, 2H), 3.53 (d, J = 10.8 Hz, 1H), 3.29 (s, 3H), 2.83 (dd, J = 7.7, 4.4 Hz, 1H), 2.58 - 2.53 (m, 2H), 2.22 - 2.14 (m, 2H), 1.87 (dd, J = 26.7, 15.9 Hz, 3H), 1.71 (s, 4H), 1.61 (s, 3H), 1.10 - 1.01 (m, 4H).
Example 33: Preparation of sodium 2-(4-((((((3 R,4S,5S,6R)-5-metho\y-4-((2R,3 R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
Figure imgf000116_0001
Step 1: Preparation of ethyl 2-(4-((methylamino)methyl)-lH-l,2,3-triazol-l-yl)acetate
Figure imgf000116_0002
[0245] The mixture of methyl(prop-2-yn-l-yl)amine (300 mg, 4.34 mmol), ethyl 2- azidoacetate (728 mg, 5.64 mmol), Cu(OAc)2 (259 mg, 1.30 mmol) and sodium ascorbate (432 mg, 2.17 mmol) in t-BuOH/H20 (6 mL, 1/1, v/v) was stirred at 30°C overnight. TLC showed the reaction was complete. The solvent was removed under vacuum. The residue was purified by flash chromatography (silica gel, 5% ~ 20% MeOH in DCM) to afford ethyl 2-(4- ((methylamino)methyl)-lH-l,2,3-triazol-l-yl)acetate (450 mg) as a brown solid. LC-MS (ESI) m/z(M+l): 199.
Step 2: Preparation of 2-(4-((methylamino)methyl)-lH-l,2,3-triazol-l-yl)acetic acid
Figure imgf000116_0003
[0246] To a mixture of ethyl 2-{4-[(methylamino)methyl]-lH-l,2,3-triazol-l-yl}acetate (450 mg, 2.27 mmol) in THF/MeOH (6 mL, v/v = 1/1) was added LiOH (3.4 mL, 6.8 mmol, 2M). The reaction mixture was stirred at room temperature for 1 h. TLC showed the reaction was complete. The reaction mixture was adjusted pH to 3 with diluted H2S04. The volatile was removed under vacuum to give 2-{4-[(methylamino)methyl]-lH-l,2,3-triazol-l-yl}acetic acid (-386 mg) in water which was used directly in the next step without further purification. LC- MS(ESI) found: 171 [M+l]+.
Step 3: Preparation of sodium 2-(4-((((((J ?, A,5A,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate
Figure imgf000117_0001
[0247] To a mixture of 2-(4-((methylamino)methyl)-lH-l,2,3-triazol-l-yl)acetic acid (171 mg, 1.01 mmol) in MeCN (6 mL) and water (2 mL) was added DIPEA (0.22 mL, 1.34 mmol) drop-wise at 0°C. The mixture was stirred at 0°C for 10 min and Intermediate 1 (300 mg, 0.67 mmol) was added. The reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by prep-HPLC (Cl 8, 0 ~ 90 % MeCN in H20 with 0.1 % formic acid) to give 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)- 2 -methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetic acid (240 mg). It was dissolved in MeCN (2 mL) and added aq. NaOH (4.5 mL, 0.1M) at 0°C. After lyophilization, sodium 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3- triazol-l-yl)acetate (240 mg) was obtained as a white solid. LC-MS (ESI) m/z(M+l): 479. 1H NMR (400 MHz, DMSO-d6 ) d 7.81 (d, J = 34.3 Hz, 1H), 5.39 (s, 1H), 5.19 (t, J = 7.4 Hz, 1H), 4.64 - 4.34 (m, 4H), 3.56 (d, J = 11.1 Hz, 1H), 3.29 (s, 3H), 2.84 (d, J = 7.0 Hz, 4H), 2.59 - 2.51 (m, 3H), 2.26 - 2.08 (m, 2H), 2.05 - 1.90 (m, 1H), 1.84 - 1.59 (m, 8H), 1.16 - 0.87 (m, 4H).
Example 34: Preparation of sodium (J/?,5/?)-3,5-dihydroxy-7-(((((J/?, V,5.V,6/?)-5- methoxy-4-((2 ?,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)amino)heptanoate
Figure imgf000118_0001
Step 1: Preparation of (.?/?, 5/?)-7-amino-3,5-dihydroxyheptanoic acid
Figure imgf000118_0003
[0248] To a mixture of tert-butyl 2-((4i?,6 ?)-6-(2-aminoethyl)-2,2-dimethyl-l,3-dioxan-4- yl)acetate (1.35 g, 4.9 mmol) in DCM (30 mL) was slowly added TFA (10 mL). The resulting mixture was stirred at r.t. for 2h. TLC showed the reaction was complete. The mixture was concentrated to give crude (.V)-2-(pyrrol i di n-3 -yl )propan-2-ol (1.3 g), which was used in the next step without further purification. LC -MS (ESI) m/z (M-l): 178.
Step 2: Preparation of ( S,6A)-6-(2-aminoethyl)-4-hydroxytetrahydro-2H-pyran-2-one
Figure imgf000118_0002
[0249] A mixture of (3i?,5i?)-7-amino-3,5-dihydroxyheptanoic acid (1.3 g) in aq. NaHCO-, solution (50 mL) was stirred at r.t. for lh, and extracted with DCM/MeOH (30 mL x 2). The combined organic layers was dried over anhydrous Na2S04 and concentrated under vacuum to give a yellow residue, which was used in the next step without further purification. LC-MS (ESI) m/z (M-l): 160.
Step 3: Preparation of (3/?,5/?)-3,5-dihydroxy-7-(((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)heptanoic acid
Figure imgf000119_0001
[0250] To a solution of (7S',7V)-6-(2-ami noethyl )-4-hydroxytetrahydro-2H-pyran-2-one (310 mg, 1.9 mmol) in DMF (10 mL) was added DIPEA (0.6 mL, 3.8 mmol) drop-wise at 0°C. The mixture was stirred at 0°C for 10 min, and Intermediate 1 (447 mg, 1.1 mmol) was added. The reaction mixture was stirred at r.t. overnight under a N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated, and the residue was purified by prep-HPLC (08,0 ~ 90 % acetonitrile in H20 with 0.1 % formic acid) to give (3R, 5/ )-3,5-dihydroxy-7- (((((37?, 4S, 5S, 6R)-5 -m ethoxy-4 -((2/?, 37?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)heptanoic acid (300 mg) as a white solid. LC-MS (ESI) m/z (M+l): 486.
Step 4: Preparation of sodium (3/?,5/?)-3,5-dih droxy-7-(((((3/?,4S,5A,6/?)-5-methoxy-4-
((2/?,3/?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)heptanoate
Figure imgf000119_0002
[0251] To a mixture of (3R, 5/ )-3,5-dihydroxy-7-[( { [(3/ ,7V,5,.y, ri/ )-5-methoxy-4-[(2R.,3R.)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)amino]heptanoic acid (210 mg, 0.43 mmol) in MeCN (lmL) was added 0.1 M NaOH (3.8 mL) at -60 °C with stirring. The solution was lyophilized to give sodium
(3/?,5/?)-3,5-dihydroxy-7-[({ [(3/?, 4S,5S, 6/?)-5-methoxy-4-[(2/?)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)amino]heptanoate (215 mg) as a white solid. LC-MS (ESI) m/z (M+l): 486. 1H NMR (400 MHz, DMSO-d6 ) d 7.59 (s, 1H),
7.08 (s, 1H), 5.19 (t, 7= 7.4 Hz, 1H), 4.56 (d, J= 2.6 Hz, 1H), 4.30 (m, 1H), 4.21 (s, 1H), 3.71 (s, 1H), 3.37 (d, J= 2.4 Hz, 1H), 3.30 (s, 3H), 3.14 (dd, J= 9.3, 4.2 Hz, 2H), 2.81 (d, J= 4.4 Hz, 1H), 2.22 - 2.10 (m, 3H), 2.07 - 1.91 (m, 4H), 1.89 - 1.76 (m, 3H), 1.70 (d, 7= 8.3 Hz, 5H), 1.61 (s, 4H), 1.57 - 1.43 (m, 3H), 1.07 (s, 3H), 0.90 - 0.83 (m, 1H). Example 35: Preparation of sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoate
Figure imgf000120_0001
Step 1: Preparation of 3-bromo-2,2-dimethylpropanoic acid
Figure imgf000120_0002
[0252] To a solution of 3 -hydroxy-2, 2-dimethylpropanoic acid (3.5 g, 30 mmol) and CBr4 (14.9 g, 45 mmol) in THF (50 mL) was added PPh3 (11.8 g, 45 mmol) at 0°C. The mixture was stirred at 0°C for 30 min. The reaction was stirred at room temperature overnight under N2 atmosphere. The reaction mixture was then quenched with water and extracted with DCM. The dichloromethane solution was dried over Na2S04, filtered and concentrated to give crude product. The crude product was purified by chromatography (silica gel, 1 to ~50 % AcOEt in PE) to give 3 -bromo-2, 2-dimethylpropanoic acid (1.3 g) as a colorless oil.
Step 2: Preparation of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)-2,2- dimethylpropanoic acid
Figure imgf000120_0003
[0253] To a solution of tert-butyl azeti din-3 -ylcarbamate hydrochloride (1.15 g, 5.5 mmol) and K2C03 (1.4 g, 10 mmol) in DMF (20 mL) was added 3 -bromo-2, 2-dimethylpropanoic acid (1.0 g, 5.5 mmol) dropwise at 0°C. The reaction was stirred at room temperature overnight under N2 atmosphere. TLC showed the reaction was complete. Water was added and the pH of the water layer adjusted to 5~6, then the water layer was extracted with DCM. The
dichloromethane solution was dried over anhydrous Na2S0 , filtered and concentrated to give crude product. The crude product was purified by column chromatography (silica gel, 1 to -10 % MeOH in DCM) to give 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)-2,2- dimethylpropanoic acid (550 mg) as a white solid. LC-MS (ESI) found: 273 [M+l]+.
Step 3: Preparation of 3-(3-aminoazetidin-l-yl)-2,2-dimethylpropanoic acid
trifluoroacetic acid salt
Figure imgf000121_0001
[0254] To a mixture of 3-(3-((tert-butoxycarbonyl)amino)azetidin-l-yl)-2,2- dimethylpropanoic acid (550 mg, 2.0 mmol) in dichloromethane (5 mL) was added
trifluoroacetic acid (3 mL) dropwise at 0°C. The reaction was stirred room temperature for 1 h. TLC showed the reaction was complete. The mixture was concentrated under reduced pressure to give 3-(3-aminoazetidin-l-yl)-2,2-dimethylpropanoic acid trifluoroacetic acid salt (440 mg) as a yellow syrup, which was used in the next step without further purification. LC-MS (ESI) found: 173[M+1]+.
Step 4: Preparation of 3-(3-(((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- l-yl)-2,2-dimethylpropanoic acid
Figure imgf000121_0002
[0255] 3-(3-aminoazetidin-l-yl)-2,2-dimethylpropanoic acid trifluoroacetic acid salt (440 mg, 1.9 mmol) was coupled with Intermediate 1 (700 mg, l .6mmol) by following the General Procedure for Coupling. The reaction was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL x 2). The combined organic layers was dried with Na2S04, filtered and concentrated to give crude product. The residue was purified by prep-HPLC (Method B: 0.1% NH4OH) to give 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)- 2 -methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoic acid (155 mg) as a white solid. LC-MS (ESI) found: 481 [M+l]+.
Step 5: Preparation of sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoic acid
Figure imgf000122_0001
[0256] To a solution of 3-(3-(((((3i?, S,,5<S,,6/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- 1 - yl)-2,2-dimethylpropanoic acid (155 mg, 0.33 mmol) in acetonitrile (1 mL) was added aq. NaOH (3 mL, 0.3 mmol, 0.1N) dropwise at -20°C. The mixture was lyophilized to give sodium 3 -(3 ~(((((3R, 4S, 5S, 6R)-5-methoxy-4-((2R, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2-dimethylpropanoic acid (160 mg) as a white solid. LC-MS (ESI) found: 481 [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 7.63 (d, J= 7.5 Hz, 1H), 5.26 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 4.06 (dd, J= 14.1, 7.0 Hz, 1H), 3.57 (t, J= 6.4 Hz, 2H), 3.52 (dd, J= 10.8, 2.4 Hz, 1H), 3.27 (s, 3H), 3.00 (dd, J= 14.8, 7.4 Hz,
2H), 2.84 (d, J= 4.2 Hz, 1H), 2.57 (d, J= 4.3 Hz, 1H), 2.16 (dd, J= 13.7, 7.0 Hz, 2H), 1.99 - 1.74 (m, 3H), 1.69 (d, j= 13.4 Hz, 4H), 1.62 (d, j= 7.6 Hz, 4H), 1.08 (s, 3H), 0.99 (s, 8H).
Example 36: Preparation of sodium ( 3R,5R)-3,5-dihydroxy-7-((l-((((3R,4S,5S,6R)-5 - methoxy-4-((2 ?,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)azetidin-3-yl)oxy)heptanoate
Figure imgf000122_0002
1. Preparation of tert-butyl 2-((4R, 6 ?)-6-(2-h drox ethyl)-2, 2-dimethyl- 1,3-dioxan- 4-yl)acetate
Figure imgf000122_0003
[0257] tert-Butyl 2-((4R, 6/i)-6-(2-aminoethyl)-2,2-dimethyl- 1 ,3 -dioxan-4-yl (acetate (10 g, 36.6 mmol) was dissolved in 200 mL of 4: 1 H20: AcOH and the mixture was placed in ice bath. To this mixture was slowly added aqueous sodium nitrite (150 mL, 2 M) via a syringe pump over 30 min. The reaction was stirred for 4 h at room temperature. TLC showed the reaction was complete. The reaction was extracted with EtOAc (3 c 50 mL). The organic layer was dried with anhydrous MgS04, filtered, and the filtrate was concentrated via rotary evaporation. The residue was purified by flash chromatography (silica gel, 1 % ~ 10% ethyl acetate in petroleum ether) to give tert-butyl 2-((4f?,6/?)-6-(2-hydroxyethyl)-2,2-dimethyl-l,3-dioxan-4- yl)acetate (1.7 g, 16.9 %) as a colorless oil.
2. Preparation of tert-butyl 2-(( /?,6A)-6-(2-iodoethyl)-2,2-dimethyl-l,3-dioxan-4- yl)acetate
Figure imgf000123_0001
[0258] To a solution of tert-butyl 2-((4R, ri/i)-6-(2-hydroxyethyl)-2,2-dimethyl- 1 ,3-dioxan-4- yl)acetate (1.7 g, 6.2 mmol), PPh3 (3.3 g, 12.4 mmol) and imidazole (1.0 g, 15 mmol) in THF (30 mL) at 0°C was added I2 (3.2 g, 12.4 mmol) dropwise. The reaction was stirred at RT overnight. The mixture was washed with water, and the organic layer was separated and washed with brine, dried over anhydrous Na2S0 and concentrated. The residue was purified by flash chromatography (silica gel, 0 ~ 30 % EtOAc in PE) to give tert-butyl 2-((4R,6S)-6-(2- iodoethyl)-2, 2-dimethyl- 1, 3 -dioxan-4-yl)acetate (1.9 g, 79.8%) as a yellow solid.
3. Preparation of tert-butyl 2-(( /?,6/?)-6-(2-((l-benzhydrylazetidin-3-yl)oxy)ethyl)-
2,2-dimethyl-l,3-dioxan-4-yl)acetate
Figure imgf000123_0002
[0259] A solution of l-benzhydrylazeti din-3 -ol (600 mg, 2.51 mmol) in THF (10 mL) was added sodium hydride (200 mg, 5.02 mmol ) at 0°C under nitrogen atmosphere and the mixture was stirred for 15 mins at 0°C. tert-Butyl 2-((4A, riS)-6-(2-iodoethyl)-2, 2-dimethyl- 1,3- dioxan-4-yl)acetate (1.1 g, 3.01 mmol) was added and the mixture was warmed to room temperature and stirred for 18 hours. TLC showed the reaction was complete. Water (20 mL) was added and the mixture was extracted with ethyl acetate (30 mL). The organic extract was washed with brine, dried over anhydrous Na2S04, and evaporated in vacuo. The residue was purified by flash chromatography (silica gel, 1 % ~ 50 % ethyl acetate in petroleum ether) to give tert-butyl2-((4i?,di?)-6-(2-((l-benzhydrylazetidin-3-yl)oxy)ethyl)-2,2-dimethyl-l,3-dioxan- 4-yl)acetate (200 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 496. 4. Preparation of (.?/?, 5/?)-7-(azetidin-3-yloxy)-3,5-dihydroxyheptanoic acid
Figure imgf000124_0001
[0260] To a mixture of tert-butyl 2-[{4R, 6R)-6-{2-\ [\ -(diphenyl methyl )azetidin-3- yl]oxy}ethyl)-2,2-dimethyl-l,3-dioxan-4-yl]acetate (200 mg, 0.4 mmol) and Pd/C (100 mg) in THF (3 mL) was added TFA (5 mL). After the air was replaced by hydrogen three times, the reaction mixture was stirred at room temperature for 16 hrs. The resulting mixture
was concentrated to give crude product, which was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 234.
5. Preparation of (.?/?, 5/?)-7-(azetidin-3-yloxy)-3,5-dihydroxyheptanoic acid
Figure imgf000124_0002
[0261] To a solution of (3R, 5//)-7-(azetidin-3-yloxy)-3,5-dihydroxyheptanoicacid (50 mg, 0.21 mmol) in MeCN (5 mL) was added DIPEA (0.2 mL) dropwise at 0 °C. The mixture was stirred at 0 °C for lOmin, and (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 95 mg, 0.21 mmol) was added. The reaction mixture was stirred at RT overnight under N2 atmosphere. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by prep-HPLC (Cl 8,0 ~ 90 % acetonitrile in H20 with 0.1 % NH3Ή20) to give (3R, 5/i)-3,5-dihydroxy-7-{ [ 1 - ({[(3i?,4ri',5ri',6/?)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]oxy}heptanoic acid (22mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 542.
6. Preparation of sodium (3/?,5/?)-3,5-dihydroxy-7-((l-((((3/?, V,5,V,6/?)-5-methoxy-
4-((2 ?,J ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)heptanoate
Figure imgf000125_0001
[0262] To a mixture of (3R, 5/i)-3,5-dihydroxy-7- { [ l -( J [(3/^-/.S',5.S',/ /^)-5-methoxy-4- [( 2R , 3f?)-2-methyl-3-(3 -methylbut-2-en-l -yl)oxiran-2-yl]- 1 -oxaspiro[2.5]octan-6- yl]oxy}carbonyl)azetidin-3-yl]oxy}heptanoic acid (22mg, 0.04 mmol) in MeCN (1 mL) was added 0.1N NaOH (0.36 mL) at -60°C with stirring. After lyophilization, (3R,4S,5S,6R)-5- methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl3-{[(3f?,5f?)-3,5-dihydroxy-7-oxo-7-(sodiooxy)heptyl]oxy}azetidine-l-carboxylate
(2lmg) was obtained as a colorless oil. LC-MS (ESI) m/z (M-l): 540. 1H NMR (400 MHz, DMSO-d6) d 5.30 (s, 1H), 5.19 (s, 1H), 4.73 (s, 1H), 4.24 (s, 1H), 4.12 - 4.01 (m, 2H), 3.68 (s, 4H), 3.54 - 3.49 (m, 1H), 3.28 (s, 3H), 3.10 (d, J = 7.6 Hz, 1H), 2.89 (d, J = 8.1 Hz, 1H), 2.84
(d, J = 4.3 Hz, 1H), 2.67 (s, 1H), 2.56 (d, J = 4.1 Hz, 2H), 2.33 (s, 1H), 2.17 (d, J = 7.1 Hz, 2H),
1.98 (m, 2H), 1.81 - 1.72 (m, 4H), 1.71 (s, 3H), 1.61 (s, 4H), 1.53 (s, 1H), 1.51 - 1.40 (m, 2H),
1.40 - 1.33 (m, 4H), 1.27 (m, 3H), 1.07 (s, 3H), 1.01 (d, J = 22.2 Hz, 1H). Example 37: Preparation of sodium 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro [3.3] heptan-2-yl)acetate
Figure imgf000125_0002
1. Preparation of tert-butyl 6-(2-(tert-butoxy)-2-oxoethyl)-2,6- diazaspiro [3.3] heptane-2-carboxylate
Figure imgf000126_0001
[0263] A mixture of tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate (1.06 g, 5.35 mmol), tert-butyl 2-bromoacetate (1.03 g, 5.28 mmol) and Cs2C03 (3.49 g,l0.7 mmol) in DMF (10 mL) was stirred at 50°C for l6h. TLC showed the reaction was complete. The slurry was diluted with EA, and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure, and purified by flash
chromatography to give tert-butyl 6-(2-(tert-butoxy)-2-oxoethyl)-2,6-diazaspiro[3.3]heptane-2- carboxylate (45 mg) as a white solid. LC-MS (ESI) found: 313 [M+H] +
2. Preparation of 2-(2,6-diazaspiro[3.3]heptan-2-yl)acetic acid
TFA salt
Boa
TFA, DCM
Figure imgf000126_0003
Figure imgf000126_0002
[0264] To a solution of tert-butyl 6-(2-(tert-butoxy)-2-oxoethyl)-2,6-diazaspiro[3.3]heptane- 2-carboxylate (345 mg, 1.10 mmol) in DCM (2 mL) was added TFA (1 mL). After the completion of addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete, and the mixture was concentrated to give crude 2-(2,6- diazaspiro[3.3]heptan-2-yl)acetic acid TFA salt (1.3 g) as a brown oil. LC-MS (ESI) found: 157 [M+l] +.
3. Preparation of 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro [3.3] heptan-2-yl)acetic acid
TFA salt
Figure imgf000126_0004
Intermediate 1
[0265] Tinder N2 atmosphere, to a solution of 2-(2,6-diazaspiro[3.3]heptan-2-yl)acetic acid 2,2,2-trifluoroacetate salt (crude, 1.3 g, 1.06 mmol), and diisopropyl ethyl amine (650 mg, 5.0 mmol) in CH3CN (5 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 490 mg, 1.09 mmol) in C¾CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, ¾0 (0.1% FA)/ C¾CN) to give 2-(6-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)-2,6-diazaspiro[3.3]heptan-2-yl)acetic acid (138 mg) as a colorless oil. LC-MS (ESI) found: 465 [M+l] +.
4. Preparation of sodium 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro [3.3] heptan-2-yl)acetate
Figure imgf000127_0001
[0266] To a solution of 2-(6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6- diazaspiro[3.3]heptan-2-yl)acetic acid (80 mg, 0.174 mmol) in CH3CN (1 mL) was added 0.1 N NaOH (1.55 mL, 0.155 mmol) solution at -60°C with stirring. After lypholization, sodium 2-(6- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6-diazaspiro[3.3]heptan-2-yl)acetate (80 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 5.27 (s, 1H), 5.19 (s, 1H), 3.97 (s, 4H), 3.63 (s, 4H), 3.51 (dd, J = 11.0, 2.4 Hz, 1H), 3.27 (d, J = 6.2 Hz, 3H), 3.11 (s, 2H), 2.85 (d, J = 4.3 Hz, 1H), 2.56 (dd, J = 11.7, 5.3 Hz, 2H), 2. l7 (s, 2H), 1.97 - 1.87 (m, 1H), 1.80— 1.67 (m, 6H), 1.61 (s, 3H), 1.05 (d, J = 17.9 Hz, 4H).
Example 38: Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- oxopiperazin-l-yl)acetate
Figure imgf000128_0001
1. Preparation of tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)-3-oxopiperazine-l- carboxylate
TFA
^ ° O
Figure imgf000128_0002
OH
[0267] To a solution of tert-butyl 3-oxopiperazine-l-carboxylate (3.9 g, 19.5 mmol) in DMF (10 mL), were added tert-butyl 2-bromoacetate (2.84 mL, 19.5 mmol) and sodium hydride
(0.856 g, 21.4 mmol, 60% in mineral oil). The reaction mixture was stirred at 25 °C for 16 hours. TLC showed the reaction was complete. The slurry was diluted with EA, and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give tert-butyl 4- (2-(tert-butoxy)-2-oxoethyl)-3-oxopiperazine-l-carboxylate (3.89 g) as a white solid. LC-MS (ESI) found: 315 [M+H] +.
2. Preparation of 2-(2-oxopiperazin-l-yl)acetic acid
Figure imgf000128_0003
[0268] To a solution of tert-butyl 4-[2-(tert-butoxy)-2-oxoethyl]-3-oxopiperazine-l- carboxylate (310 mg, 0.98 mmol) in DCM (2 mL) was added TFA (1 mL). After addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was reaction was complete. The reaction mixture was then concentrated to give crude 2-(2- oxopiperazin-l-yl)acetic acid (TFA salt, 500 mg) as a brown oil, which was directly used in the next reaction step. LC-MS (ESI) found: 159 [M+l]+.
3. Preparation of 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-oxopiperazin- l-yl)acetic acid
Figure imgf000129_0001
Intermediate 1
[0269] To a solution of 2-(2-oxopiperazin-l-yl)acetic acid 2,2,2-trifluoroacetate salt (500 mg, 0.98 mmol) and N,N-diisopropylethylamine (707 mg, 5.5 mmol) in CH3CN (3 mL) under a N2 atmosphere was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate
(Intermediate 1, 490 mg, 1.09 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 hrs. TLC showed the reaction was complete. The reaction mixture was then concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- oxopiperazin-l-yl)acetic acid (150 mg) as a colorless oil. LC-MS (ESI) found: 467 [M+l]+.
4. Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- oxopiperazin-l-yl)acetate
Figure imgf000129_0002
[0270] To a solution of 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-oxopiperazin- 1 - yl)acetic acid (103 mg, 0.221 mmol) in CH3CN (1 mL) was added 0.1 N NaOH (1.98 mL, 0.198 mmol) at -60 °C with stirring. After lyophilization, sodium 2-(4-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)-2-oxopiperazin-l-yl)acetate (105 mg) was obtained as white solid. 1H NMR (400 MHz, DMSO-d6 ) d 5.40 (s, 1H), 5.19 (t, J= 7.4 Hz, 1H), 3.89 (d, J= 21.0 Hz, 2H), 3.69 - 3.49 (m, 5H), 3.35 (s, 3H), 3.29 (s, 3H), 2.86 (d, J= 4.3 Hz, 1H), 2.58 (d, J= 4.3 Hz, 1H), 2.17 (dd, j= 13.5, 6.9 Hz, 2H), 1.99 (td, j= 13.5, 4.4 Hz, 1H), 1.85 - 1.77 (m, 2H), 1.73 (d, j =
17.4 Hz, 4H), 1.61 (s, 3H), 1.08 (s, 3H), 1.01 (d, J= 13.6 Hz, 1H).
Example 39: Preparation of sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptan-6-yl)oxy)acetate
Figure imgf000130_0001
equivalent
1. TFA/DCM,
rt, overnight 0.1 M NaOH(aq)
2. Intermediate 1 0 °C to rt
DIPEA, MeCN equivalent
0 °C to rt 6h
Figure imgf000130_0002
Figure imgf000130_0003
1. Preparation of tert-butyl 6-hydroxy-2-azaspiro[3.3]heptane-2-carboxylate
Figure imgf000130_0004
equivalent
[0271] To a solution of tert-butyl 6-oxo-2-azaspiro[3.3]heptane-2-carboxylate (845 mg) in MeOH (20 mL) was added NaBH4 (152 mg) at 0 °C. The mixture was stirred at r.t. for 6 hrs. TLC showed the reaction was complete. The mixture was concentrated and the residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give tert-butyl 6-hydroxy-2-azaspiro[3.3]heptane-2-carboxylate (853 mg, quantitive) as white solid. 1H NMR (400 MHz, DMSO-d6 ) d 5.00 (d, J = 6.2 Hz, 1H), 3.92 (q, J = 7.0 Hz, 1H), 3.75 (d, J = 21.0 Hz, 4H), 2.37 (ddd, J = 11.8, 6.1, 2.6 Hz, 2H), 1.95 - 1.86 (m, 2H), 1.35 (s, 9H). LC MS: m/z (M+l)+ 214. 2. Preparation of tert-butyl 6-(2-(tert-butoxy)-2-oxoethoxy)-2-azaspiro[3.3]heptane- 2-carboxylate
Figure imgf000131_0001
[0272] To a solution of NaH (60% wt, 80 mg) in anhydrous THF (5 mL) was added a solution of tert-butyl 6-hydroxy-2-azaspiro[3.3]heptane-2-carboxylate (213 mg) in anhydrous THF (5 mL) while cooling the reaction in an ice-bath. After 1 h, tert-butyl 2-bromoacetate was added to the mixture at the same temperature. The mixture was stirred at r.t overnight. TLC showed the reaction was complete. The reaction was quenched with NH4Cl (aq) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give tert-butyl 6-(2-(tert-butoxy)-2-oxoethoxy)-2- azaspiro[3.3]heptane-2-carboxylate (236 mg) as a yellow oil. 1H NMR (400 MHz, CDCl3) d 3.93 (m, 1H), 3.87 (d, J = 8.4 Hz, 4H), 3.85 (s, 2H), 2.54 - 2.42 (m, 2H), 2.21 - 2.14 (m, 2H), 1.47 (s, 9H), 1.42 (s, 9H). LC MS: m/z (M+l)+ 328.
3. Preparation of sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptan-6-yl)oxy)acetate
Figure imgf000131_0002
[0273] To a solution of tert-butyl 6-(2-(tert-butoxy)-2-oxoethoxy)-2-azaspiro[3.3]heptane-2- carboxylate (236 mg) in DCM (2.5 mL) was added TFA (2.5 mL), and the resulting mixture was stirred at room temperature overnight. TLC showed the reaction was complete. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling the reaction in an ice-bath. To the resulting solution was added (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 280 mg), and the reaction was allowed to warm to r.t. and be stirred at r.t. for 6 hrs. TLC showed the reaction was complete. The reaction mixture was quenched with aqueous NaHCO, (1 M) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3Ή20) to give 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-azaspiro[3.3]heptan-6- yl)oxy)acetic acid (100 mg ) as a white solid. LC MS: m/z (M-l) 478. To a solution of 2-((2- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-azaspiro[3.3]heptan-6-yl)oxy)acetic acid (100 mg) in H20/MeCN (v/v, 10: 1, 5 mL) was added aqueous NaOH (0.1 M, 2 mL) at -40 °C. The solvent was lyophilized to give sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro[3.3]heptan-6-yl)oxy)acetate (100 mg) as a white solid. 'H NMR (400 MHz, DMSO- d6) d 5.27 (dd, J = 4.2, 2.5 Hz, 1H), 5.19 (td, J = 7.4, 3.9 Hz, 1H), 3.91 (s, 1H), 3.83 (d, J = 17.0 Hz, 4H), 3.54 - 3.47 (m, 2H), 3.27 (s, 3H), 2.84 (d, J = 4.4 Hz, 1H), 2.56 (d, J = 4.3 Hz, 1H), 2.53 (d, J = 6.6 Hz, lH),2.36 (t, J = 9.5 Hz, 2H), 2.17 (q, J = 6.4 Hz, 2H), 2.03 - 1.94 (m, 2H), 1.90 (dd, J = 13.3, 4.6 Hz, 1H), 1.80 - 1.72 (m, 1H), 1.74 (d, J = 11.0 Hz, 1H), 1.70 (s, 3H), 1.61 (s, 3H), 1.07 (s, 3H), 1.04 - 0.96 (m, 1H), 0.94 (d, J = 6.5 Hz, 1H). LC MS: m/z (M- 1) 478.
Example 40: Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3- methylazetidin-3-yl)oxy)acetate
Figure imgf000132_0001
, ,
Figure imgf000132_0003
0 C to rt 6h
1. Preparation of tert-butyl 3-hydroxy-3-methylazetidine-l-carboxylate
Figure imgf000132_0002
[0274] To a solution of 3-methylazetidin-3-ol hydrochloride (500 mg) in DCM (10 mL) was added Et3N (1.63 g) at r.t. After 5 mins, Boc20 (1.77 g) was added, and the reaction was stirred at r.t. for 6hrs. The mixture was then concentrated, and the residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give tert-butyl 3 -hydroxy-3 - methylazetidine-l-carboxylate (757 mg, quantitive) as a yellow oil. LC MS: m/z (M+l)+ 188.
2. Preparation of tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)-3-methylazetidine-l- carboxylate
Figure imgf000133_0001
g
[0275] To a solution of NaH (60% wt, 80 mg) in anhydrous THF (5 mL) was added a solution of tert-butyl 3 -hydroxy-3 -methylazeti dine- l-carboxylate (187 mg) in anhydrous THF (5 mL) while cooling the reaction in an ice-bath. After 1 h, tert-butyl 2-bromoacetate was added to the mixture at the same temperature. The mixture was then allowed to stir at r.t overnight. The reaction was quenched with NH4Cl (aq) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and
concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give tert-butyl 3 -(2-(tert-butoxy)-2-oxoethoxy)-3 -methylazeti dine- 1- carboxylate (220 mg) as a yellow oil. LC MS: m/z (M+l)+ 302.
3. Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3- methylazetidin-3-yl)oxy)acetate
Figure imgf000133_0002
[0276] To a solution of tert-butyl 3-(2-(tert-butoxy)-2-oxoethoxy)-3-methylazetidine-l- carboxylate (220 mg) in DCM (2.5 mL) was added TFA (2.5 mL), and the resulting mixture was stirred at room temperature overnight. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling in an ice-bath. To the resulting solution was added (3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl (4- nitrophenyl) carbonate (Intermediate 1, 282 mg), and the reaction was allowed to stir at room temperature for 6 hrs. The reaction mixture was quenched with aqueous NaHC03 (1 M) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep-HPLC (Cl 8,0 ~ 90 % acetonitrile in ¾0 with 0.1 % NH3Ή2O) to give 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3 -methylazeti din-3 - yl)oxy)acetic acid (210 mg) as a white solid. LC MS: m/z (M-l) 474. To a solution of 2-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3-methylazetidin-3-yl)oxy)acetic acid in
H20/MeCN(v/v, 10: 1, 5 mL) was added aqueous NaOH (0.1 M, 0.9 eq) at -40 °C. The solvent was lyophilized to give sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3-methylazetidin-3- yl)oxy)acetate (220 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 5.30 (s, 1H), 5.23 - 5.11 (m, 1H), 3.86 (d, J = 8.9 Hz, 2H), 3.60 (d, J = 8.9 Hz, 2H), 3.54 - 3.49 (m, 3H), 3.28 (s,
3H), 2.84 (d, J = 4.3 Hz, 1H), 2.55 (dd, J = 7.5, 5.3 Hz, 2H), 2.17 (q, J = 7.3 Hz, 2H), 1.93 (s, 1H), 1.77 (dd, J = 14.4, 10.5 Hz, 2H), 1.72 - 1.69 (m, 4H), 1.61 (d, J = 1.2 Hz, 3H), 1.37 (s, 3H), 1.07 (s, 3H), 1.02 (dd, J = 13.7, 3.7 Hz, 1H), 0.94 (d, J = 6.5 Hz, 1H). LC MS: m/z (M-l) 474.
Example 41: Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate
Figure imgf000134_0002
Intermediate 1 1. Preparation of tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)piperazine-l-carboxylate
Figure imgf000134_0001
[0277] To a solution of tert-butyl piperazine- l-carboxylate (1.5 g, 8.05 mmol) in DMF (10 mL), were added tert-butyl 2-bromoacetate (1.68 g, 8.70 mmol) and Cs2C03 (3.49 g, 10.7 mmol). The reaction mixture was stirred at 25 °C for 16 hours. TLC showed the reaction was complete. The slurry was diluted with EA, and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give tert-butyl 4-(2-(tert-butoxy)-2-oxoethyl)piperazine-l- carboxylate (2.08 g) as a white solid. LC-MS (ESI) found: 301 [M+H] +
2. Preparation of 2-(piperazin-l-yl)acetic acid lt
Figure imgf000135_0002
[0278] To a solution of tert-butyl 4-[2-(tert-butoxy)-2-oxoethyl]piperazine- l-carboxylate (230 mg, 0.766 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The mixture was concentrated to give crude 2-(piperazin-l-yl)acetic acid TFA salt (500 mg ) as a brown oil. LC-MS (ESI) found: 145 [M+l]+.
3. Preparation of 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetic acid
Figure imgf000135_0001
Intermediate 1
[0279] Under N2 atmosphere, to a solution of 2-(piperazin-l-yl)acetic acid 2,2,2- trifluoroacetate salt (110 mg, 0.65 mmol), ethyl diisopropylamine (500 mg, 3.9 mmol) in
CH3CN (3 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate
(Intermediate 1, 290 mg, 0.65 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by adding DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 h. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2- (4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l-yl)acetic acid (150 mg) as a colorless oil. LC-MS (ESI) found: 453 [M+l]+.
4. Preparation of sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l- yl)acetate
Figure imgf000136_0001
[0280] To a solution of 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin- 1 -yl)acetic acid (80 mg, 0.18 mmol) in CH3CN (1 mL) was added 0.1 N NaOH (1.59 mL, 0.159 mmol) solution at -60 °C with stirring. After lyophilization, sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate (82 mg) was obtained as white solid. 1H NMR (400 MHz, DMSO-d6) d 5.36 (s, 1H), 5.19 (t, J= 7.5 Hz, 1H), 3.54 (dd, J= 11.0, 2.5 Hz, 1H), 3.28 (s, 7H), 2.85 (d, j= 4.3 Hz, 1H), 2.66 (s, 2H), 2.58 (dd, j= 7.2, 5.4 Hz, 2H), 2.40 (d, j= 17.4 Hz, 4H), 2.23 - 2.09 (m, 2H), 1.94 (td, 7 = 13.3, 4.3 Hz, 1H), 1.79 (d, 7= 1 1.0 Hz, 2H), 1.71 (s, 4H), 1.61 (s, 3H), 1.08 (s, 3H), 1.02 (d, J= 13.7 Hz, 1H).
Example 42: Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-4-carboxylate
Figure imgf000137_0003
X TFA
Figure imgf000137_0004
1. Preparation of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylat
Figure imgf000137_0001
[0281] To a solution of tert-butyl 3-hydroxyazetidine-l-carboxylate (1.0 g, 5.77 mmol) and TEA (1.15 g, 11.5 mmol) in DCM (15 mL) was added methanesulfonyl chloride (0.67 mL,
8.66 mmol) at 0 °C. After 2 h, TLC showed the reaction was complete, and the reaction was diluted with ice brine. The organic layer was separated, dried over anhydrous MgS04 and concentrated to give tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate (1.2 g) as a colorless oil. LC-MS(ESI) found: 196 [M+H-56]+
2. Preparation of methyl l-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-pyrazole-4- carboxylate
Figure imgf000137_0002
[0282] To a mixture of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate (750 mg, 2.98 mmol) in DMF (10 mL) was added K2C03 (1.2 g, 8.95 mmol) and methyl lH-pyrazole-4- carboxylate (376. 4 mg, 2.98 mmol). The reaction was stirred at 100 °C overnight. The mixture was washed with water and extracted with EA. The organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure to give methyl l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-lH-pyrazole-4-carboxylate (550 mg). LC-MS (ESI) found: 236 [M+H-56]+.
3. Preparation of l-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-pyrazole-4- carboxylic acid
Figure imgf000138_0001
[0283] To a stirred suspension of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH- pyrazole-4-carboxylate (550 mg, 1.95 mmol) in THF (10 mL) and H20 (5 mL) was
added LiOH (93.65 mg, 3.91 mmol). The reaction mixture was stirred at r.t. overnight. The reaction mixture was concentrated to remove the solvent, and 5 mL of water and 5 mL of EtOAc were added. The water layer was adjusted to pH = 3 with 1N HC1 and then extracted with DCM. The organic layer was concentrated to give l-{ l-[(tert-butoxy)carbonyl]azetidin-3- yl}-lH-pyrazole-4-carboxylic acid (430 mg) as a colorless oil. LC-MS(ESI) found: 268
[M+l]+.
4. Preparation of l-(azetidin-3-yl)-lH-pyrazole-4-carboxylic acid trifluoroacetic acid salt
Figure imgf000138_0002
X TFA
[0284] To a mixture of l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH -pyrazole-4- carboxylic acid (430 mg, 1.60 mmol) in DCM (2 mL) was added TFA (1 mL) dropwise at 0 °C. The reaction was stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure to give l-(azetidin-3-yl)-lH-pyrazole-4-carboxylic acid trifluoroacetic acid salt (210 mgas a yellow syrup, which was directly used in the next step without
purification. LC-MS (ESI) found: 168 [M+l]+.
5. Preparation of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- lH-pyrazole-4-carboxylic acid
Figure imgf000139_0001
[0285] To a solution of l-(azetidin-3-yl)-lH-pyrazole-4-carboxylic acid (210 mg, 1.25 mmol) in MeCN (10 mL) was added DIPEA (638.15 mg, 5.03 mmol) dropwise at 0 °C. The mixture was stirred at 0 °C for 10 min, and (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate
(Intermediate 1, 562.12 mg, 1.26 mmol) was added at the same temperature. The reaction was then stirred at room temperature overnight under N2 atmosphere. The mixture was then concentrated under vacuum to remove the solvent while keeping external temperature below 40°C. The residue was diluted with dichlorom ethane and washed with ammonium acetate buffer (pH 4.0, 20 mL). The dichloromethane solution was dried over anhydrous Na2S04, filtered and concentrated to give the crude product. The residue was purified by column chromatography on silica gel (dichloromethane: methanol = 100 : 1 to 10 : 1) and further purified by preparative HPLC (C18, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 H20) to give l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]- l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH-pyrazole-4-carboxylic acid (230 mg) as a white solid. LC-MS (ESI) found: 476 [M+l]+.
6. Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- lH-pyrazole-4-carboxylate
Figure imgf000139_0002
[0286] To a solution of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-lH- pyrazole-4-carboxylic acid (230 mg, 0.48 mmol) in acetonitrile (1 mL) was added 5 mL of 0.1 N aq. NaOH solution at -50 °C. The mixture was lyophilized overnight to give sodium l-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-4-carboxylate (230 mg) as a white solid. LC-MS (ESI) found: 476 [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 8.07 (d, ./ = 2.6 Hz, 1H), 7.72 (s, 1H), 5.34 (s, 1H), 5.25 (dd, J= 9.3, 3.9 Hz, 1H), 5.18 (t, J= 7.4 Hz, 1H), 4.31 (t, J= 8.4 Hz, 2H), 4.18 (s, 2H), 3.55 (dd, 7= 11.0, 2.6 Hz, 1H), 3.30 (s, 3H), 2.84 (d, J = 4.3 Hz, 1H), 2.55 (dd, 7= 13.6, 5.3 Hz, 2H), 2.17 (s, 2H), 2.03 - 1.89 (m, 1H), 1.80 (dd, 7 = 19.7, 10.8 Hz, 3H), 1.70 (s, 3H), 1.60 (s, 3H), 1.08 (s, 3H), 1.03 (d, J= 13.2 Hz, 1H).
Example 43: Preparation of sodium 5-((((((J ?, »V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-
3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinate
Figure imgf000140_0001
1. Preparation of sodium 5-(aminomethyl)picolinate
Figure imgf000140_0002
[0287] To a solution of 5-cyanopicolinic acid (200 mg, 1.35 mmol) in MeOH (2 mL) and 1M NaOH (2 mL) was added Pd(OH)2/C (100 mg). Then the reaction was stirred under hydrogen at r.t. overnight. TLC showed the reaction was complete. The reaction mixture was filtered and concentrated under vacuum to give crude sodium 5-(aminomethyl)picolinate (280 mg, crude) as brown oil, which was used in the next step without further purification. LC-MS (ESI) found: l53[M+l]+.
2. Preparation of 5-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinic acid
Figure imgf000141_0001
[0288] To a solution of sodium 5-(aminomethyl)picolinate (280 mg, crude) in MeCN/H20 (2 mL /2 mL) was added (3f?, ,S,,5S,, d/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 300 mg, 0.68 mmol), and the reaction was then stirred at r.t. overnight. TLC showed the reaction was complete. The resulting mixture was concentrated to give a residue, which was purified by prep-HPLC (Method B: 0.1% NH4OH) to afford 5-((((((3R,4S,5S, 6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinic acid (140 mg) as a colorless oil. LC-MS (ESI) found: 461 [M+l]+.
3. Preparation of sodium 5-((((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinate
Figure imgf000141_0002
[0289] To a solution of 5-((((((3f?,4,S,,5S,, d/?)-5-methoxy-4-((2f?,3f?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinic acid (140 mg, 0.3 mmol) in MeCN (1.0 mL) was added 0.1 N NaOH solution (2.7 mL, 0.27 mmol) at -60°C with stirring, followed by lyophilization to give sodium 5-((((((3i?,4ri,,5ri',6/?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)methyl)picolinate (124.7 mg) as a white solid. LC-MS (ESI) found: 46l [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 8.46 - 8.38 (m, 1H), 7.90 (d, J = 8.0 Hz, 1H), 7.77 (s, 1H), 7.68 (d, J = 7.9 Hz, 1H), 5.33 (s, 1H), 5.20 (s, 1H), 4.25 (d, J = 6.0 Hz, 2H), 3.29 (s, 4H), 2.85 (d, J = 4.3 Hz, 1H), 2.58 (d, J = 4.3 Hz, 1H), 2.19 (dd, J = 13.0, 6.5 Hz, 2H), 1.96 - 1.78 (m, 3H), 1.71 (s, 4H), 1.61 (s, 3H), 1.09 (d, J = 10.1 Hz, 4H). Example 44: Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-3-carboxylate
Figure imgf000142_0001
1. Preparation of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate
Boc — N^>— OH Boc -O OMs
Figure imgf000142_0002
[0290] To a solution of tert-butyl 3-hydroxyazetidine-l-carboxylate (1.0 g, 5.77 mmol) and TEA (1.15 g, 11.5 mmol) in DCM (15 mL) was added methanesulfonyl chloride (0.67 mL, 8.66 mmol) at 0 °C. After 2 h, TLC showed the reaction was complete, and the reaction was diluted with ice brine. The organic layer was dried over anhydrous MgS04 and
concentrated to give tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate (l.2g) as a colorless oil. LC-MS(ESI) found: 196 [M+H-56]+
2. Preparation of l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-lH-pyrazole-3- carboxylate
Figure imgf000142_0003
[0291] To a mixture of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate (750 mg, 2.98 mmol) in DMF (10 mL) was added K2C03 (1.2 g, 8.95 mmol) and methyl lH-pyrazole- 3-carboxylate (376.4 mg, 2.98 mmol). The reaction was stirred at 100 °C overnight. TLC showed the reaction was complete. The mixture was washed with water and extracted with EA. The organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure to give methyl l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-lH-pyrazole-3-carboxylate (650 mg). LC-MS (ESI) found: 236 [M+H-56]+.
3. Preparation of l-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-pyrazole-3- carboxylic acid
Figure imgf000143_0001
[0292] To a stirred suspension of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH- pyrazole-3-carboxylate (650 mg, 2.31 mmol) in THF (10 mL) and H20 (5 mL) was added LiOH (110.66 mg, 4.62 mmol). The reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The reaction mixture was concentrated to remove the solvent, and 5 mL of water and 5 mL of EtOAc were added. The water layer was adjusted to pH = 3 with 1N HC1 and then extracted with DCM. The organic layer was concentrated to give l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-lH-pyrazole-3-carboxylic acid (470 mg) as a colorless oil. LC- MS(ESI) found: 268 [M+l]+.
4. Preparation of l-(azetidin-3-yl)-lH-pyrazole-3-carboxylic acid trifluoroacetic acid salt
Figure imgf000143_0002
[0293] To a mixture of l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH -pyrazole-3- carboxylic acid (470 mg, 1.76 mmol) in DCM (2 mL) was added TFA (1 mL) dropwise at 0 °C. The reaction was stirred at room temperature for 1 h. Then the mixture was concentrated under reduced pressure to give l-(azetidin-3-yl)-lH-pyrazole-3-carboxylic acid trifluoroacetic acid salt (347 mg) as a yellow syrup, which was directly used in the next step without further purification. LC-MS (ESI) found: 168 [M+l]+.
5. Preparation of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- lH-pyrazole-3-carboxylic acid
Figure imgf000143_0003
[0294] To a solution of l-(azetidin-3-yl)-lH-pyrazole-3-carboxylic acid (347 mg, 1.38 mmol) in MeCN (10 mL) was added DIPEA (890.1 mg, 6.9 mmol) dropwise at 0 °C to pH = 8. The mixture was stirred at 0°C for 10 min and (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate
(Intermediate 1, 562.12 mg, 1.26 mmol) was added. After addition, the reaction was stirred at room temperature overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was then concentrated under vacuum to remove the solvent while keeping external temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL). The dichloromethane solution was dried over anhydrous Na2S04, filtered and concentrated to give the crude product. The residue was purified by column chromatography on silica gel (dichloromethane: methanol = 100 : 1 to 10 : 1) and further purified by preparative HPLC (C18, 0 ~ 90 % acetonitrile in H20 with 0.1 %
NH3 H20) to give l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-3- carboxylic acid (210 mg as a white solid. LC-MS (ESI) found: 476 [M+l]+.
6. Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- lH-pyrazole-3-carboxylate
Figure imgf000144_0001
[0295] To a solution of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-lH- pyrazole-3 -carboxylic acid (210 mg, 0.44 mmol) in acetonitrile (1 mL) was added 5 mL of 0.1 N aq. NaOH solution at -50 °C. The mixture was lyophilized overnight to give sodium l-[l- ({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH-pyrazole-4-carboxylate (200 mg) as a white solid. LC-MS (ESI) found: 476 [M+l]+. 1H NMR (400 MHz, DMSO-d6 ) d 7.77 (s, 1H), 6.45 (s, 1H), 5.35 (s, 1H), 5.24 (d, J= 5.7 Hz, 1H), 5.18 (t, J= 7.4 Hz, 1H), 4.32 (t, J= 8.4 Hz, 2H), 4.24 - 4.15 (m, 2H), 3.55 (dd, 7= 11.0, 2.5 Hz, 1H), 3.31 (s, 3H), 2.84 (d, J= 4.3 Hz, 1H), 2.56 (dd, J = 11.5, 5.3 Hz, 2H), 2.25 - 2.10 (m, 2H), 1.95 (dd, 7= 13.4, 3.9 Hz, 1H), 1.83 (s, 1H), 1.76 (t, J= 14.2 Hz, 2H), 1.70 (s, 3H), 1.60 (s, 3H), 1.08 (s, 3H), 1.03 (d, J= 12.9 Hz,
1H). Example 45: Preparation of sodium 6-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate
Figure imgf000145_0001
1. Preparation of tert-butyl 3-(5-bromopyridin-2-yl)-3-hydroxyazetidine-l- carboxylate
1 . isopropyl magnesium chloride,
Figure imgf000145_0002
Figure imgf000145_0003
Figure imgf000145_0004
[0296] To a stirred solution of 5-bromo-2-iodopyridine (5.0 g, 17.6 mmol) in dry THF (30 mL) was added isopropylmagmesium chloride (2.0 M in THF, 9.69 mL, 19.3 mmol) dropwise at -20 °C over a period of 10 min. Then a solution of tert-butyl 3-oxoazetidine-l-carboxylate (3.62 g, 21.13 mmol) in dry THF (10 mL) was added at -20°C to the above reaction mixture. The reaction mixture was stirred at r.t. for 16 hrs under N2. and the starting material was consumed completely by TLC. The reaction mixture was cooled to -l0°C and quenched with 10% citric acid solution, extracted with EA and washed with NaHCO, (aq.). The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo. The residue was purified by flash chromtography on silica gel eluting with PE/EtOAc 10-20/1 to give tert-butyl 3-(5- bromopyridin-2-yl)-3-hydroxyazetidine-l-carboxylate (730 mg) as a white solid. LC-MS (ESI) found: 329 [M+H] + 2. Preparation of methyl 6-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)nicotinate
CO PdCI2(dppf)
Figure imgf000146_0001
[0297] To a stirred solution of tert-butyl 3-(5-bromopyridin-2-yl)-3-hydroxyazetidine-l- carboxylate (200 mg, 0.60 mmol) in MeOH (5mL), Pd(dppf)Cl2 (22.66 mg, 0.031 mmol) was added in one portion at 25 °C, followed by TEA (2 mL). The mixture is purged with carbon monoxide at room temperature for 10 minutes, then it was heated to 90 °C and stirred overnight under 40 psi of carbon monoxide. TLC and LC-MS showed the reaction was complete. The reaction solution is cooled to room temperature, quenched with saturated brine and extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over magnesium sulfate, and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica gel: hexane: ethyl acetate = 15: 1) to give methyl 6-( 1 -(tert- butoxycarbonyl)-3-hydroxyazetidin-3-yl)nicotinate (155 mg) as a brown oil. LC-MS (ESI) found: 309 [M+l]+.
3. Preparation of methyl 6-(l-(tert-butoxycarbonyl)-3-
((methylsulfonyl)oxy)azetidin-3-yl)nicotinate
Figure imgf000146_0002
[0298] To a stirred solution of methyl 6-{ l-[(tert-butoxy)carbonyl]-3-hydroxyazetidin-3- yl}pyridine-3-carboxylate (300 mg, 0.97 mmol) in dry DCM (5 mL), was added triethylamine (0.21 mL, 1.46 mmol) dropwise. Five minutes later, methanesulfonyl chloride (0.083 mL, 1.07 mmol) was added at 0 °C over a period of 10 min. The reaction mixture was stirred at r.t. for 1.0 h under N2 and the starting material was completely consumed by TLC. The reaction mixture was cooled to 0°C and diluted with 10% NaCl solution, extracted with DCM and washed with NaHCO, (aq.). The organic layer was dried over anhydrous
Na2S04 and concentrated in vacuo to give crude methyl 6-(l-(tert-butoxycarbonyl)-3- ((methylsulfonyl)oxy)azetidin-3-yl)nicotinate (410 mg) as a colorless oil. LC-MS (ESI) found: 387 [M+l]+. 4. Preparation of methyl 6-(l-(tert-butoxycarbonyl)azetidin-3-yl)nicotinate
Figure imgf000147_0001
[0299] To a stirred solution of methyl 6-{ l-[(tert-butoxy)carbonyl]-3- (methanesulfonyloxy)azetidin-3-yl}pyridine-3-carboxylate (410 mg, 1.06 mmol) in dry MeOH (5 mL), was added Pd/C (20 mg, 0.188 mmol) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h under N2 and the starting material was completely consumed by TLC.
The reaction mixture was filtered. To the filtrate was added 10% NaCl solution, and the aqueous layer was extracted with EA. The organic layer was dried over anhydrous
Na2S04 and concentrated in vacuo to give methyl 6-(l-(tert-butoxycarbonyl)azetidin-3- yl)nicotinate (105 mg) as a colorless oil. LC-MS (ESI) found: 293 [M+l]+.
5. Preparation of lithium 6-(l-(tert-butoxycarbonyl)azetidin-3-yl)nicotinate
Figure imgf000147_0002
[0300] To a stirred solution of methyl 6-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}pyridine-3- carboxylate (105 mg, 0.36 mmol) in THF (3 mL), was added lithium hydroxide (39.5 mg, 0.89 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed TLC. The reaction mixture was concentrated in vacuo to give lithium 6-(l-(tert-butoxycarbonyl) azeti din-3 -yl) nicotinate (crude, 150 mg) as a colorless oil. LC-MS (ESI) found: 279 [M+l]+.
6. Preparation of methyl 6-(azetidin-3-yl)nicotinic acid
TFA salt
Figure imgf000147_0003
[0301] To a solution of lithium 6-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}pyridine-3- carboxylate (crude, 150 mg, 0.35 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The mixture was concentrated to give 6-(azeti din-3 -yl) nicotinic acid TFA salt (crude, 300 mg) as a brown oil. LC-MS (ESI) found: 177 [M-l] . 7. Preparation of 6-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)nicotinic acid
Figure imgf000148_0001
Intermediate 1
[0302] Under a N2 atmosphere, to a solution of 6-(azetidin-3-yl)nicotinic acid 2,2,2- trifluoroacetate salt (crude, 300 mg) and, ethyldiisopropylamine (462 mg, 3.5 mmol) in CH3CN (3 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate
(Intermediate 1, 160 mg, 0.36 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 hrs. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 6-( 1 -((((3//,4.V,5.V,6//)-5-methoxy-4-((2//,3//)-2- ethyl-3-(3- ethylbut-2-en- l -yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)nicotinic acid (130 mg) as a colorless oil. LC-MS (ESI) found: 487 [M+l]+.
8. Preparation of sodium 6-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)nicotinate
Figure imgf000148_0002
[0303] To a solution of 6-( 1 -((((3//,4.V,5.V,6//)-5-methoxy-4-((2//,3//)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)nicotinic acid (53 mg, 0.11 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (0.98 mL, 0.098 mmol) solution at -60°C with stirring. Sodium 6-(l-((((3i?,4ri,,5ri',6i?)-5-methoxy-4- ((2//,3//)-2-methyl-3-(3-methylbut-2-en- l -yl)oxiran-2-yl)- l -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)nicotinate (55 mg) was obtained as white solid after
lyophilization. 1H MR (400 MHz, DMSO-d6 ) d 8.96 (s, 1H), 8.06 (dd, J= 7.9, 1.7 Hz, 1H), 7.21 (d, J= 7.9 Hz, 1H), 5.34 (s, 1H), 5.19 (t, j= 7.4 Hz, 1H), 4.23 (t, 7= 8.2 Hz, 2H), 4.14 - 3.93 (m, 3H), 3.55 (dd, 7 = 11.0, 2.4 Hz, 1H), 3.31 (s, 3H), 2.85 (d, 7= 4.3 Hz, 1H), 2.56 (dd, J = 10.3, 5.2 Hz, 2H), 2.17 (d, J= 4.9 Hz, 2H), 2.02 - 1.92 (m, 1H), 1.88 - 1.73 (m, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.09 (s, 3H), 1.03 (d, J= 12.9 Hz, 1H).
Example 46: Preparation of sodium 5-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-
((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinate
Figure imgf000149_0001
1. Preparation of tert-butyl 3-(6-bromopyridin-3-yl)-3-hydroxyazetidine-l- carboxylate
Figure imgf000149_0002
Figure imgf000149_0003
[0304] To a stirred solution of 2-bromo-5-iodopyridine (5.0 g, 17.6 mmol) in dry THF (30 mL), was added isopropylmagnesium chloride (9.69 mL, 19.37 mmol) dropwise over a period of 10 min at -20°C. Then a solution of tert-butyl 3-oxoazetidine-l-carboxylate (3.62 g, 21.1 mmol) in dry THF (10 mL) was added at -20°C to the above reaction mixture. The reaction mixture was then stirred at r.t. for 16 hrs under N2. The starting material was completely consumed by TLC . The reaction mixture was cooled to -l0°C and quenched with 10% citric acid solution, extracted with EA and washed with NaHC03 (aq.). The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo. Then the crude product was purified by flash chromatography on silica gel eluting with PE/ EtOAc 10-20/1 to give tert-butyl 3-(6- brom opyri din-3 -yl)-3-hydroxyazeti dine- l-carboxylate (3.56 g) as a white solid. LC-MS (ESI) found: 329 [M+H] +
2. Preparation of methyl 5-{l-[(tert-butoxy)carbonyl]-3-hydroxyazetidin-3- yl}pyridine-2-carboxylate
CO Pd(dppf)CI
Figure imgf000150_0001
[0305] To a stirred solution of tert-butyl 3-(6-bromopyridin-3-yl)-3-hydroxyazetidine-l- carboxylate (1.1 g, 3.34 mmol) in MeOH (5mL), was added Pd(dppf)Cl2 (0.023 g, 0.031 mmol) in one portion at 25°C, followed by the addition of TEA (2 mL). The mixture was purged with carbon monoxide at room temperature for 10 minutes, and it was then heated to 90 °C and stirred overnight under 40 psi of carbon monoxide. TLC showed the reaction was complete. The reaction solution was cooled to room temperature, and diluted with saturated brine and extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous magnesium sulfate, and concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica gel; hexane: ethyl acetate = 15: 1) to give methyl 5-{ l-[(tert-butoxy)carbonyl]-3-hydroxyazetidin-3-yl}pyridine-2-carboxylate (650 mg) as a brown oil. LC-MS (ESI) found: 309 [M+l]+.
3. Preparation of lithium 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3-yl)picolinate
Figure imgf000150_0002
[0306] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)picolinate (120 mg, 0.39 mmol) in THF (3 mL), was added lithium hydroxide (32.9 mg, 0.78 mmol) in H20 (lmL) in one portion at 25 °C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)picolinate (crude, 150 mg) as a colorless oil. LC-MS (ESI) found: 295 [M+l]+.
4. Preparation of 5-(3-hydroxyazetidin-3-yl)picolinic acid
TFA salt
Figure imgf000150_0003
COOH
[0307] To a solution of lithium 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3-yl)picolinate (crude, 150 mg) in DCM (2 mL) at r.t. was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. The mixture was concentrated to give 5- (3-hydroxyazetidin-3-yl)picolinic acid (crude, TFA salt, 150 mg) as a brown oil. LC-MS (ESI) found: 195 [M+l]+.
5. Preparation of 5-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)picolinic acid
Figure imgf000151_0001
Intermediate 1
[0308] Under a N2 atmosphere, to a solution of 5-(3-hydroxyazetidin-3-yl)pyridine-2- carboxylic acid (78.1 mg, 0.4 mmol), DIPEA (0.53 mL, 3.22 mmol) in CH3CN (3 mL) was added dropwise a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 180 mg, 0.40 mmol) in CH3CN (1 mL) at 0°C, followed by adding DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 5-[3-hydroxy-l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]pyridine-2-carboxylic acid (123 mg) as a colorless oil. LC-MS (ESI) found: 503 [M+l]+.
6. Preparation of sodium 5-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinate
Figure imgf000151_0002
[0309] To a solution of 5-[3-hydroxy-l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]pyridine-2-carboxylic acid (123 mg, 0.25 mmol) in CH3CN (0.3 mL) was added 0.1 N NaOH (2.20 mL, 0.22 mmol) solution at -60°C with stirring. After lyophilization, sodium 5- [3-hydroxy- l-({[(3R,4S, 5 S,6R)-5-methoxy-4-[(2R,3R)-2 -methyl-3 -(3-methylbut-2-en-l - yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]pyridine-2-carboxylate (122 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 8.61 (s, 1H), 7.87 (dd, J= 20.4, 8.1 Hz, 2H), 6.62 (s, 1H), 5.37 (s, 1H), 5.18 (s, 1H), 4.12 (s, 4H), 3.56 (dd, J =
11.0, 2.5 Hz, 1H), 3.31 - 3.29 (m, 3H), 2.85 (d, J= 4.3 Hz, 1H), 2.57 (d, J= 4.3 Hz, 1H), 2.54 (d, j= 6.3 Hz, 1H), 2.17 (s, 2H), 1.96 (dd, j= 13.3, 4.6 Hz, 1H), 1.86 - 1.72 (m, 3H), 1.69 (s, 3H), 1.60 (s, 3H), 1.09 (s, 3H), 1.02 (d, j= 12.8 Hz, 1H). Example 47: Preparation of sodium 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate
Figure imgf000152_0002
1. Preparation of 6-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3-yl)nicotinic acid
Figure imgf000152_0001
[0310] To a stirred solution of methyl 6-(l-(tert-butoxycarbonyl)-3-hydroxyazeti din-3 - yl)nicotinate (120 mg, 0.39 mmol) in THF (3 mL), was added lithium hydroxide (32.9 mg, 0.78 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 6-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)nicotinate (crude, 150 mg) as a colorless oil. LC-MS (ESI) found: 295 [M+l]+.
2. Preparation of 6-(3-hydroxyazetidin-3-yl)nicotinic acid TFA salt
Boc
Figure imgf000153_0001
[0311] To a solution of lithium 6-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3-yl)nicotinate (crude, 150 mg) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The mixture was concentrated to give 6-(3-hydroxyazeti din-3 -yl)nicotinic acid (crude, TFA salt, 150 mg) as a brown oil. LC-MS (ESI) found: 195 [M+l]+.
3. Preparation of 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)nicotinic acid
TFA salt
Figure imgf000153_0002
Intermediate 1
[0312] Under a N2 atmosphere, to a solution of 6-(3-hydroxyazeti din-3 -yl)pyridine-3- carboxylic acid (78.12 mg, 0.40 mmol), DIPEA (0.40 mL, 2.42 mmol) in MeCN (3 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 180 mg, 0.40 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by adding DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 6-[3-hydroxy-l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]pyridine-3 -carboxylic acid (25 mg) as a colorless oil. LC-MS (ESI) found: 503 [M+l]+.
4. Preparation of sodium 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate
Figure imgf000154_0001
[0313] To a solution of 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl)oxy)carbonyl)azetidin-3 - yl)nicotinic acid (25 mg, 0.05 mmol) in CH3CN (0.3 mL) was added 0.1 N NaOH
solution(0.45 mL, 0.05 mmol) at -60°C with stirring. After lyophilization, sodium 6-(3- hydroxy- 1 -((((3R,4S,5 S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3 -methylbut-2-en- 1 -yl)oxiran- 2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)nicotinate (26 mg) was obtained as white solid. 1H MR (400 MHz, DMSO-d6 ) d 8.92 (s, 1H), 8.09 (dd, J= 8.0, 1.7 Hz, 1H), 7.49 (d, j= 7.9 Hz, 1H), 6.62 (s, 1H), 5.35 (s, 1H), 5.18 (t, j= 7.4 Hz, 1H), 4.29 (d, j= 26.8 Hz, 2H), 3.97 (s, 2H), 3.55 (dd, j= 11.0, 2.6 Hz, 1H), 3.30 (s, 3H), 2.84 (d, j= 4.3 Hz, 1H), 2.55 (dd, j= 7.6, 5.3 Hz, 2H), 2.16 (d, j= 5.0 Hz, 2H), 1.97 (s, 1H), 1.87 - 1.73 (m, 3H), 1.70 (s, 3H), 1.60 (s, 3H), 1.08 (d, j= 4.6 Hz, 3H), 1.03 (s, 1H).
Example 48: Preparation of sodium 3-fluoro-l-((((.?/?, S,5A,6/?)-5-methoxy-4-((2/?,.?/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidine-3-carboxylate
Figure imgf000154_0002
1. Preparation of l-(tert-butoxycarbonyl)azetidine-3-carboxylic acid
Figure imgf000154_0003
[0314] Di-tert-butyldi carbonate (18 g, 83 mmol) was added to a solution of 3- azetidinecarboxylic acid (7.6 g, 75 mmol) in THF (150 mL), water (75 mL) and 2M NaOH (75 mL) while cooling in an ice-bath. The reaction mixture was then stirred at r.t. for 24 h. TLC showed the reaction was complete. The organic solvent was removed in vacuo and the residual water mixture was acidified to pH = 1 by the addition of 4 M HC1. The water mixture was then extracted with EtOAc and the combined organic layers were washed with brine, dried over anhydrous MgS04 and concentrated in vacuo to give l-(tert-butoxycarbonyl)azetidine-3- carboxylic acid (11 g) as a white solid. LC-MS (ESI) m/z (M+l): 202.
2. Preparation of l-tert-butyl 3-methyl azetidine-l,3-dicarboxylate
Figure imgf000155_0001
[0315] To a solution of azetidine-l,3-dicarboxylic acid mono-tert-butyl ester (11 g, 54.7 mmol) in DMF (75 mL), were added K2C03 (15 g, 109.4 mmol) and CH3I (9.3 g, 65.6 mmol). The reaction mixture was stirred at r.t. for 24 h. TLC showed the reaction was complete. Water was added. The water phase was extracted with EtOAc and the combined organic layers were washed with brine, dried over anhydrous MgS04 and concentrated in vacuo to give azetidine- l,3-dicarboxylic acid l-tert-butyl ester 3-methyl ester (8 g) as a white solid. LC-MS (ESI) m/z (M+l): 216.
3. Preparation of l-tert-butyl3-methyl3-fluoroazetidine-l,3-dicarboxylate
N-Fluorobenzenesulfonimide
LiHMDS
Figure imgf000155_0002
Figure imgf000155_0003
[0316] To a solution of l-tert-butyl 3-methyl azetidine-l,3-dicarboxylate (8 g, 37.2 mmol) in THF (80 mL), was added LiHMDS (44 mL, 44.6 mmol, 1 M in THF) at -78°C. 3 N- fluorobenzenesulfonimide (14 g, 44.6 mmol) was added 30 min later. The reaction mixture was stirred at -78°C for 4 hrs. TLC showed the reaction was complete. Water was added. The water phase was extracted with EtOAc and the combined organic layers were washed with brine, dried over anhydrous MgS04 and concentrated in vacuo to give l-tert-butyl3 -methyl 3- fluoroazetidine-l,3-dicarboxylate (6 g) as a colorless oil. LC-MS (ESI) m/z (M+l): 234.
4. Preparation of l-(tert-butoxycarbonyl)-3-fluoroazetidine-3-carboxylic acid
Figure imgf000155_0004
[0317] To a stirred suspension of l-tert-butyl 3-methyl 3-fluoroazetidine-l,3-dicarboxylate (6 g, 25.7 mmol) in MeOH:H20 (1 : 1, 60 mL) was added lithium hydroxide (10 g, 257 mmol) at room temperature and the mixture was stirred at room temperature for 24 hours. The reaction mixture was concentrated, water (20 mL) was added to the residue, and the resulting mixture was acidified to pH~4 with 1 N HCI. The acidified aqueous layer was extracted with ethyl acetate (5 x 50 mL). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2S04 and evaporated under reduced pressure to give l-(tert-butoxycarbonyl)-3- fluoroazetidine-3 -carboxylic acid (5.5 g) as a white solid. LC-MS (ESI) m/z (M+l): 220.
5. Preparation of 3-fluoroazetidine-3-carboxylic acid
Figure imgf000156_0001
[0318] To a mixture of l-(tert-butoxycarbonyl)-3-fluoroazetidine-3 -carboxylic acid (300 mg, 1.36 mmol) in DCM (2 mL) was added TFA (1 mL). The reaction mixture was stirred at room temperature for 2hrs. TLC showed the reaction was complete. The resulting mixture was concentrated to give crude product, which was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 120.
6. Preparation of 3-fluoro-l-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3- carboxylic acid
Figure imgf000156_0002
[0319] To a solution of 3-fluoroazetidine-3-carboxylic acid (150 mg, 1.25 mmol) in MeCN (5 mL) was added DIPEA (1 mL, 5.03 mmol) dropwise at 0 °C. The mixture was stirred at 0 °C for 10 min, then (3f?, ri',5ri',6/?)-5-methoxy-4-[(2f?,3f?)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl-4-nitrophenylcarbonate (Intermediate 1, 563 mg,
1.25 mmol) was added. The reaction mixture was stirred at r.t. overnight under a N2
atmosphere. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep-HPLC (Cl 8,0 ~ 90 % acetonitrile in H20 with 0.1 % NH3Ή2O) to give 3-fluoro-l-({[(3i?, ,S,,5S,, 6/?)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidine-3- carboxylicacid (300 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 428.
7. Preparation of sodium 3-fluoro-l-(((( /?, S,5»S,6/?)-5-methoxy-4-((2/?, /?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidine-3-carboxylate
Figure imgf000157_0001
[0320] To a mixture of 3-fluoro-l-({[(3i?,4,S,,5S,, d/?)-5-methoxy-4-[(2R,3R) -2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidine-3- carboxylic acid (210 mg, 0.49 mmol) in MeCN (1 mL) was added aq. NaOH (4.4 mL, 0.1 mol/L) at -60 °C with stirring. The reaction mixture was lyophilized to give sodium 3-fluoro-l- ((((3f?, 4S, 5S, 6// )- 5 -m ethoxy-4 -((2//, 3f?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3-carboxylate (195 mg) as a white solid. LC- MS (ESI) m/z (M+l): 428. 1H NMR (400 MHz, DMSO-d6 ) 5 5.31 (s, 1H), 5.19 (t, J = 7.3 Hz, 1H), 4.21 (s, 2H), 3.85 (d, J = 9.1 Hz, 2H), 3.53 (dd, J = 10.9, 2.5 Hz, 1H), 3.29 (s, 3H), 2.84
(d, J = 4.3 Hz, 1H), 2.57 (dd, J = 7.8, 4.6 Hz, 2H), 2.17 (dd, J = 12.8, 6.4 Hz, 2H), 1.99 - 1.88 (m, 1H), 1.78 (dd, J = 18.5, 7.2 Hz, 3H), 1.70 (s, 3H), 1.61 (s, 3H), 1.08 (s, 3H), 1.02 (d, J = 13.6 Hz, 1H).
Example 49: Preparation of sodium 6-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-
2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate
Figure imgf000158_0001
l. Preparation of methyl 6-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3-yl)nicotinate
Figure imgf000158_0002
[0321] To a stirred solution of methyl 6-{ l-[(tert-butoxy)carbonyl]-3-hydroxyazetidin-3- yl}pyridine-3-carboxylate (205 mg, 0.665 mmol) in dry DCM (5 mL), was added
(diethylamino)sulfur trifluoride (0.163 mL, 1.33 mmol) at 0°C over a period of 10 min. The reaction mixture was stirred at r.t. for 2 h under N2, and the starting material was completely consumed by TLC. The reaction mixture was cooled to -l0°C and quenched with water, extracted with EA and washed with NaCl (aq.). The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo, then the crude product was purified by flash
chromatography on silica gel eluting with PE/EtOAc = 20-15/1 to give methyl 6-(l-(tert- butoxycarbonyl)-3-fluoroazeti din-3 -yl)nicotinate (185 mg) as a colorless oil. LC-MS (ESI) found: 311 [M+H] +
2. Preparation of lithium 6-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3- yl)nicotinate
Figure imgf000158_0003
[0322] To a stirred solution of methyl 6-(l-(tert-butoxycarbonyl)-3-fluoroazeti din-3 - yl)nicotinate (185 mg, 0.59 mmol) in THF (3 mL), was added lithium hydroxide (52.44 mg, 1.19 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h, and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 6-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3- yl)ninotinate (crude, 256 mg) as a colorless oil. LC-MS (ESI) found: 297 [M+l]+.
3. Preparation of 6-(3-fluoroazetidin-3-yl)nicotinic acid
TFA salt
Figure imgf000159_0001
[0323] To a solution of lithium 6-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3-yl)nicotinate (crude, mg, 0.57 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The reaction was concentrated to give 6-(3-fluoroazeti din-3 -yl)nicotinic acid (crude, TFA salt, 200 mg) as a brown oil. LC-MS (ESI) found: 197 [M+l]+.
4. Preparation of sodium 6-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate
Figure imgf000159_0002
[0324] Stepl : Under a N2 atmosphere, to a solution of 6-(3-fluoroazeti din-3 -yl)pyridine-3- carboxylic acid (200 mg, 0.51 mmol, 2,2,2-trifluoroacetate salt) and DIPEA (0.42 mL, 2.55 mmol) in CH3CN (3 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 228.0 mg, 0.50 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 6-(3-fluoro-l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinic acid (96 mg) as a colorless oil. LC-MS (ESI) found:
505 [M+l]+. Step2: To a solution of 6-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)nicotinic acid (96 mg, 0.19 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.73 mL) solution at -60°C with stirring. After lypholization, sodium 6-(3-fluoro-l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -yl)nicotinate (lOOmg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 8.99 (s, 1H), 8.17 (dd, J= 8.0, 1.7 Hz, 1H), 7.46 (d, J= 7.9 Hz, 1H), 5.36 (s, 1H), 5.18 (t, J = 7.3 Hz, 1H), 4.46 (s, 2H), 4.29 (dd, j= 22.4, 10.3 Hz, 2H), 3.56 (dd, j = 11.0, 2.5 Hz, 1H), 3.31 (s, 3H), 2.84 (d, 7= 4.3 Hz, 1H), 2.56 (dd, 7= 9.3, 5.3 Hz, 2H), 2.17 (s, 2H), 2.02 - 1.93 (m, 1H), 1.81 (dt, J= 29.8, 14.9 Hz, 3H), 1.70 (s, 3H), 1.60 (s, 3H), 1.08 (s,
3H), 1.03 (d, J= 12.7 Hz, 1H).
Example 50: Preparation of sodium 5-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl) picolinate
Figure imgf000160_0001
Intermediate 1
1. Preparation of methyl 5-(l-(tert-butoxycarbonyl)-3-
((methylsulfonyl)oxy)azetidin-3-yl)picolinate
Figure imgf000161_0001
[0325] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)picolinate (240 mg, 0.778 mmol) and triethylamine (0.163 mL, 1.17 mmol) in dry DCM (5mL) was added methanesulfonyl chloride (0.066 mL, 1.856 mmol) dropwise over a period of 10 min at 0°C. The reaction mixture was stirred at r.t. for 1.0 h under N2 and the starting material was completely consumed by TLC. The reaction mixture was cooled to 0°C, quenched with 10% NaCl solution and extracted with DCM. The combined organic layers were washed with NaHCO, (aq.), dried over anhydrous Na2S04 and concentrated in vacuo to give methyl 5- (l-(tert-butoxycarbonyl)-3-((methylsulfonyl)oxy)azetidin-3-yl)picolinate (294 mg) as a colorless oil. LC-MS (ESI) found: 387 [M+l]+.
2. Preparation of methyl 5-(l-(tert-butoxycarbonyl)azetidin-3-yl)picolinate
Figure imgf000161_0002
[0326] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)-3-
((methylsulfonyl)oxy)azetidin-3-yl)picolinate (290 mg, 0.751 mmol) in dry MeOH (5 mL), was added Pd/C (20 mg, 0.188 mmol, 10%) in one portion at 25 °C. The reaction mixture was stirred at r.t. for 1.0 h under N2, and the starting material was completely consumed by TLC. The reaction mixture was filtered, and 10% NaCl solution was added to the filtrate. The resulting mixture was extracted with EA. The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo to give methyl 5-(l-(tert-butoxycarbonyl)azetidin-3-yl)picolinate (200 mg) as a colorless oil. LC-MS (ESI) found: 293 [M+l]+.
3. Preparation of lithium 5-(l-(tert-butoxycarbonyl)azetidin-3-yl) picolinate
Figure imgf000161_0003
[0327] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)azetidin-3-yl)picolinate (200 mg, 0.68 mmol) in THF 3 (mL), was added lithium hydroxide (33 mg, 0.89 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 5-(l-(tert-butoxycarbonyl)azetidin-3-yl)picolinate (crude, 300 mg) as a colorless oil. LC-MS (ESI) found: 279 [M+l]+.
4. Preparation of 5-(azetidin-3-yl)picolinic acid
Figure imgf000162_0001
[0328] To a solution of lithium 5-(l-(tert-butoxycarbonyl)azetidin-3-yl)picolinate (crude, 200 mg, 0.72 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. Then the mixture was concentrated to afford 5-(azetidin-3-yl)picolinic acid (crude, TFA salt, 200 mg) as a brown oil. LC-MS (ESI) found: 179 [M+l]+.
5. Preparation of sodium 5-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)picolinate
Figure imgf000162_0002
Intermediate 1
[0329] Stepl : Under N2 atmosphere, to a solution of 5-(azetidin-3-yl)pyridine-2-carboxylic acid (51.44 mg, 0.29 mmol, TFA salt), DIPEA (0.239 mL, 1.4 mmol) in CH3CN (3 mL) was added a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1 300 mg, 0.67 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by adding DMAP (5 mg, 0.067 mmol). Then the mixture was stirred at r.t. for 16 hrs. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 5-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azeti din-3 -yl)picolinic acid (140 mg) as a colorless oil. LC-MS (ESI) found: 487 [M+l]+. Step2: To a solution of 5-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)picolinic acid (70 mg, 0.144 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.30 mL) solution at -60°C with stirring. After lypholization, sodium 5-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]pyridine-2-carboxylate (75 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO- d6) d 8.39 (s, 1H), 7.88 (d, 7= 8.1 Hz, 1H), 7.82 - 7.77 (m, 1H), 5.35 (s, 1H), 5.19 (d, J= 7.0 Hz, 1H), 4.32 (s, 2H), 3.91 (s, 3H), 3.55 (dd, 7= 11.0, 2.4 Hz, 1H), 3.31 (s, 3H), 2.85 (d, 7 =
4.3 Hz, 1H), 2.56 (dd, J= 11.1, 5.3 Hz, 2H), 2. l7 (s, 2H), 2.02 - 1.91 (m, 1H), 1.79 (q, J= 19.0 Hz, 3H), 1.69 (s, 3H), 1.60 (s, 3H), 1.09 (s, 3H), 1.03 (d, J= 13.2 Hz, 1H).
Example 51: Preparation of sodium l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)azetidin-3-yl]-lH-imidazole-4-carboxylate
Figure imgf000163_0001
1. Preparation of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate
Figure imgf000163_0002
[0330] To a stirred solution of tert-butyl 3-hydroxyazetidine-l-carboxylate (5.0 g, 28.87 mmol) and TEA (5.83 g, 57.74 mmol) in DCM (100 mL) was added methanesulfonyl chloride (3.35 mL, 43.31 mmol) at 0 °C. The reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The mixture was washed with aq. NaHCO,, and the organic layer was separated. The solvent was removed under reduced pressure to obtain tert-butyl 3- (methanesulfonyloxy)azetidine-l-carboxylate (6.3 g) as a colorless oil. LC-MS(ESI) found: 196 [M+H-56]+
2. Preparation of methyl l-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-imidazole- 4-carboxylate
Figure imgf000164_0001
[0331] To a mixture of tert-butyl 3-(methanesulfonyloxy)azetidine-l-carboxylate (1.5 g, 5.97 mmol) in DMF (28 mL) was added Cs2C03 (5.83 g, 17.90 mmol) and methyl 1H- imidazole-4-carboxylate (1.51 g, 11.94 mmol). The reaction was stirred at 100 °C overnight. TLC showed the reaction was complete. The mixture was washed with water and extracted with EA, and the organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with hexanes : ethyl acetate = 1 : 1 to give methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH- imidazole-4-carboxylate (410 mg) and methyl l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-lH- imidazole-5-carboxylate (130 mg). LC-MS (ESI) found: 282 [M+H]+. methyl l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-lH-imidazole-4-carboxylate: 1H NMR (400 MHz, CDCl3) d 8.84 (s, 1H), 7.99 (s, 1H), 5.38 (s, 1H), 4.57 (t, J = 8.5 Hz, 2H), 4.10 (d, J = 7.1 Hz, 2H), 3.97 (s, 3H), 1.49 (s, 9H). methyl l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-lH-imidazole-5- carboxylate: 1H NMR (400 MHz, CDCI3) d 9.27 (s, 1H), 7.96 (s, 1H), 5.67 - 5.59 (m, 1H), 4.63 - 4.51 (m, 2H), 4.30 (dd, J= 9.9, 5.0 Hz, 2H), 3.93 (s, 3H), 1.48 (s, 9H).
3. Preparation of l-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-imidazole-4- carboxylic acid
Figure imgf000164_0002
[0332] To a stirred suspension of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH- imidazole-4-carboxylate (410 mg, 1.46 mmol) in THF (10 mL) and H20 (5 mL) was added Li OH (93.65 mg, 3.91 mmol). The reaction mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The reaction mixture was concentrated to remove the solvent, then diluted with 5 mL of water and 5 mL of EtOAc. The water phase was adjusted to pH = 3 with 1N HC1 and then extracted with DCM. The organic layer was concentrated to give l-{ 1- [(tert-butoxy)carbonyl]azetidin-3-yl}-lH-imidazole-4-carboxylic acid (250 mg) as a colorless oil. LC-MS(ESI) found: 268 [M+l]+. 4. Preparation of l-(azetidin-3-yl)-lH-imidazole-4-carboxylic acid trifluoroacetic acid salt
Figure imgf000165_0001
[0333] To a solution of l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-lH-imidazole-4- carboxylic acid (250 mg, 0.94 mmol) in DCM (2 mL) was added TFA (1 mL) dropwise at 0 °C The reaction was stirred at room temperature for 1 h. TLC showed the reaction was complete. The mixture was concentrated under reduced pressure to give l-(azetidin-3-yl)-lH-imidazole- 4-carboxylic acid trifluoroacetic acid salt (240 mg) as a yellow syrup, which was used directly in the next step without purification. LC-MS (ESI) found: 168 [M+l]+.
1. Preparation of l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]- lH-pyrazole-4-carboxylic acid
Figure imgf000165_0002
[0334] To a solution of l-(azetidin-3-yl)-lH-imidazole-4-carboxylic acid (240 mg, 0.75 mmol) in MeCN (5 mL) was added DIPEA (385.86 mg, 2.99 mmol) dropwise at 0 °C. The mixture was stirred at 0°C for 10 min, and (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 334.62 mg, 0.75 mmol) was added. The reaction was then stirred at room temperature overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was then concentrated under vacuum to remove the solvent while keeping external temperature below 40°C. The residue was diluted with dichloromethane and washed with ammonium acetate buffer (pH 4.0, 20 mL). The organic layer was dried over anhydrous Na2S04, filtered and concentrated to give the crude product. The residue was purified by column chromatography on silica gel column (eluent: dichloromethane: methanol = 100 : 1 to 10 : 1) and further purified by preparative HPLC (C18, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 H20) to give l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH-imidazole-4- carboxylic acid (186 mg) as a white solid. LC-MS (ESI) found: 476 [M+l]+.
5. Preparation of sodium l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]-lH-imidazole-4-carboxylate
Figure imgf000166_0001
[0335] To a solution of l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH- imidazole-4-carboxylic acid (186 mg, 0.39 mmol) in MeCN (2 mL) was added 0.1 N aq. NaOH (3.52 mL) at -50 °C. The mixture was lyophilized overnight to give sodium l-[l- ({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH-imidazole-4-carboxylate (190 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 7.85 (s, 1H), 7.07 (s, 1H), 5.86 (s, 1H), 5.33 (s, 1H), 5.19 (t, J= 7.3 Hz, 1H), 4.30 (t, J= 8.7 Hz, 2H), 4.17 - 4.03 (m, 2H), 3.54 (dd, J= 11.0, 2.4 Hz, 1H), 3.30 (s, 3H), 2.85 (d, j= 4.3 Hz, 1H), 2.56 (t, j= 5.9 Hz, 2H), 2.17 (dq, j= 15.3, 7.8 Hz, 2H), 2.01 - 1.91 (m, 1H), 1.80 (dd, J= 20.2, 10.9 Hz, 2H), 1.70 (s, 4H), 1.60 (s, 3H), 1.08 (s, 3H), 1.01 (d, J= 13.3 Hz, 1H). Example 52: Preparation of sodium 5-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)- 2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinate
Figure imgf000166_0002
Intermediate 1
1. Preparation of methyl 5-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3-yl)picolinate
Figure imgf000167_0001
[0336] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)-3-hydroxyazetidin-3- yl)picolinate (250 mg, 0.81 mmol) in dry DCM (5 mL), was added (diethylamino)sulfur trifluoride (0.198 mL, 1.62 mmol) drop wise at 0°C over a period of 10 min. The reaction mixture was stirred at r.t. for 2 h under N2, and the starting material was completely consumed by TLC. The reaction mixture was cooled to -l0°C and quenched with water, extracted with
EA, and the combined organic layers washed with NaCl (aq.). The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo, then the crude product was purified by flash chromatography on silica gel eluting with PE/EtOAc 20-15/1 to give methyl 5-(l-(tert- butoxycarbonyl)-3-fluoroazeti din-3 -yl)picolinate (240 mg) as a colorless oil. LC-MS (ESI) found: 311 [M+H] +
2. Preparation of lithium 5-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3- yl)picolinate acid
Figure imgf000167_0002
[0337] To a stirred solution of methyl 5-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3- yl)picolinate (240 mg, 0.774 mmol) in THF (3 mL), was added lithium hydroxide (52.0 mg, 1.19 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 5-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3- yl)picolinate(crude, 226 mg) as a colorless oil. LC-MS (ESI) found: 297 [M+l]+.
3. Preparation of 5-(3-fluoroazetidin-3-yl)picolinic acid
TFA salt
Figure imgf000167_0003
[0338] To a solution of lithium 5-(l-(tert-butoxycarbonyl)-3-fluoroazetidin-3-yl)picolinate (crude, 226 mg, 0.77 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The solvent ws removed to give 5-(3-fluoroazetidin-3-yl)picolinic acid (crude, TFA salt, 200 mg) as a brown oil. LC-MS (ESI) found: 197 [M+l]+.
4. Preparation of sodium 5-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinate
Figure imgf000168_0001
Intermediate 1
[0339] Step 1 : To a solution of 5-(3-fluoroazetidin-3-yl)picolinic acid 2,2,2-trifluoroacetate salt (131 mg, 0.67 mmol) and ethyldiisopropylamine (700 mg, 5.4 mmol) in CH3CN (3 mL) under N2 was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate
(Intermediate 1, 300 mg, 0.67 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 5-(3-fluoro-l-((((3i?,4ri,,5ri,,6i?)-5-methoxy-4- ((2//,3//)-2-methyl-3-(3-methylbut-2-en- l -yl)oxiran-2-yl)- l -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)picolinic acid (169 mg) as a colorless oil. LC-MS (ESI) found: 505 [M+l]+.
Step 2: To a solution of 5-(3-fluoro-l-((((3i?,4ri,,5ri,,6i?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)picolinic acid (69 mg, 0.137 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.23 mL, 0.123 mmol) solution at -60°C with stirring. After lypholization, sodium 5-(3-fluoro-l- ((((3//,4,V,5ri,6//)-5-methoxy-4-((2//,3//)-2- ethyl-3-(3- ethylbut-2-en- l -yl)oxiran-2-yl)- l - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)picolinate (66 mg) was obtained as white solid. 1H NMR (400 MHz, DMSO-d6 ) d 8.58 (s, 1H), 7.91 (dd, J = 17.2, 7.5 Hz, 2H),
5.38 (s, 1H), 5.18 (s, 1H), 4.40 (dd, J = 21.3, 7.3 Hz, 4H), 3.57 (d, J = 10.8 Hz, 1H), 3.32 - 3.30 (m, 3H), 2.85 (d, J = 4.1 Hz, 1H), 2.58 (d, J = 4.2 Hz, 1H), 2.54 (d, J = 6.4 Hz, 1H), 2.17 (s,
2H), 1.97 (d, J = 9.2 Hz, 1H), 1.84 (s, 1H), 1.82 - 1.73 (m, 2H), 1.70 (s, 3H), 1.60 (s, 3H), 1.09 (s, 3H), 1.03 (d, J = 13.3 Hz, 1H). Example 53: Preparation of sodium 2-(l-((((J ?, »V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- ylidene)acetate
Figure imgf000169_0001
1. Preparation of tert-butyl 3-(2-(tert-butoxy)-2-oxoethyl)-3-hydroxyazetidine-l- carboxylate
Figure imgf000169_0002
[0340] TMSC1 (0.5 mL, 6.4 mmol) was added to a suspension of Zn (6.1 g, 93.4 mmol) in THF (200 mL) under N2. The mixture was stirred for 15 mins at room temperature and then heated to reflux (75 °C). The heating was stopped, and tert-butyl 2-bromoacetate (7.9 mL, 71.2 mmol) was added at such a rate that the THF boiled gently. After being heated to reflux for 1 h, the mixture was stirred for 1 h at room temperature. A solution of tert-butyl 3-oxoazetidine-l- carboxylate (10 g, 58.4 mmol) in THF (30 mL) while cooling the reaction in an ice bath. After being stirred for 1 h at room temperature, TLC showed the reaction was complete. The mixture was quenched by dropwise addition of aq. NH4Cl (50 mL) at 0°C and then extracted with EA (50 mL). The organic layer was dried over anhydrous Na2S04 and concentrated under vacuum to give a yellow residue, which was purified by flash chromatography (silica gel, 1 %~ 20 % ethyl acetate in petroleum ether) to give tert-butyl3-(2-(tert-butoxy)-2-oxoethyl)-3- hydroxyazetidine-l-carboxylate (8.5 g) as a colorless oil. LC-MS (ESI) m/z (M+l): 288. 2. Preparation of tert-butyl 3-(2-(tert-butoxy)-2-oxoethylidene)azetidine-l- carboxylate
Figure imgf000170_0001
[0341] To a solution of tert-butyl 3-(2-(tert-butoxy)-2-oxoethyl)-3-hydroxyazetidine-l- carboxylate (300 mg, 1.1 mmol) in DCM (5 mL) was added (diethylamino)sulfur trifluoride (0.3 mL, 2.1 mmol) under ice-bath. The reaction was stirred at r.t. for 16 hrs. The resulting mixture was washed with a NaHCO, solution and brine, dried over anhydrous Na2S04, filtered and concentrated to give crude product, which was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 270.
3. Preparation of 2-(azetidin-3-ylidene)acetic acid
Figure imgf000170_0002
[0342] To a mixture of tert-butyl 3-(2-(tert-butoxy)-2-oxoethylidene)azetidine-l-carboxylate (260 mg, 0.89 mmol) in DCM (1 mL) was added TFA (2 mL). The reaction mixture was stirred at room temperature for 2 hrs. The resulting mixture was concentrated to give crude product, which was used in the next step without further purification.
4. Preparation of 2-(l-((((3/?, V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- ylidene)aceticacid
Figure imgf000170_0003
[0343] To a solution of 2-(azeti din-3 -ylidene)acetic acid (300 mg, 1.74 mmol) in MeCN (5 mL) was added DIPEA (1.2 mL, 6.9 mmol) drop wise at 0 °C. The mixture was stirred at 0 °C for 10 min, then (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 779 mg, 1.74 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 Ή2O) to give (3/^-/.V,5*SA/ -5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en- l - yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl4-[(l,3-dioxolan-2-yl)methyl]piperazine-l- carboxylate (150 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 422.
5. Preparation of sodium 2-(l-((((.?/?, S,6/?)-5-methoxy-4-((2/?,.?/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- ylidene)acetate
Figure imgf000171_0001
[0344] To a mixture of 2-(l-((((3i?, S',6 ?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-ylidene)acetic acid (100 mg, 0.23 mmol) in MeCN (1 mL) was added aq. NaOH (2.1 mL, 0.1 M) at -60 °C with stirring. The reaction mixture was lyophilized to give sodium 2-(l-((((3f?, ri', 6/?)-5-methoxy-4- ((27?, 37?)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azeti din-3 -ylidene)acetate (103 mg) as a white solid. LC-MS (ESI) m/z (M+l): 422. 1H NMR (400 MHz, DMSO-d6 ) d 5.50 (s, 1H), 5.32 (s, 1H), 5.19 (t, J = 7.4 Hz, 1H), 4.53 (d, J = 81.6 Hz, 4H), 3.53 (m, 1H), 3.29 (d, J = 3.8 Hz, 4H), 2.85 (d, J = 4.3 Hz, 1H),
2.60 - 2.54 (m, 2H), 2.17 (m, 2H), 2.01 - 1.91 (m, 1H), 1.77 (d, J = 11.0 Hz, 2H), 1.71 (s, 4H), 1.61 (s, 3H), 1.08 (s, 3H), 1.01 (d, J = 13.3 Hz, 1H).
Example 54: Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)thio)acetate
Figure imgf000171_0002
Intermediate 1 1. Preparation of tert-butyl 3-((2-ethoxy-2-oxoethyl)thio)azetidine-l-carboxylate
Figure imgf000172_0001
[0345] To a stirred solution of ethyl 2-mercaptoacetate (, 889 mg, 7.38 mmol) and tert-butyl 3-iodoazetidine-l-carboxylate (1.9 g, 6.711 mmol) in dry DMF (15 mL), was added K2C03 (1.85 g, 13.4 mmol) at 0°C over a period of 10 min. The reaction mixture was stirred at 50 °C for 16 hrs under N2, and the starting material was completely consumed by TLC. The reaction mixture was cooled to 0°C, quenched with water, extracted with EA and washed with
NaCl (aq.). The organic layer was dried over anhydrous Na2S04 and concentrated in vacuo. Then the resulting crude product was purified by flash chromatography on silica gel eluting with PE/EtOAc 10-5/1 to give tert-butyl 3-((2-ethoxy-2-oxoethyl)thio)azetidine-l-carboxylate (1.7 g) as a colorless oil. LC-MS (ESI) found: 276 [M+H] +
2. Preparation of lithium 2-((l-(tert-butoxycarbonyl)azetidin-3-yl)thio)acetate
Figure imgf000172_0002
[0346] To a stirred solution of tert-butyl 3 -((2-ethoxy -2-oxoethyl)thio)azeti dine- 1- carboxylate (240 mg, 0.872 mmol) in THF (3 mL), lithium hydroxide (109 mg, 2.62 mmol) in H20 (1 mL) was added in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h, and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give lithium 2-((l-(tert-butoxycarbonyl)azeti din-3 -yl)thio)acetate (crude, 400 mg) as a colorless oil. LC-MS (ESI) found:248 [M+l]+.
3. Preparation of 2-(azetidin-3-ylthio)acetic acid
TFA salt
Figure imgf000172_0003
[0347] To a solution of lithium 2-((l-(tert-butoxycarbonyl)azeti din-3 -yl)thio)acetate (crude, 400 mg, 0.87 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. It was concentrated to give 2-(azeti din-3 - ylthio)acetic acid (crude, TFA salt, 500 mg) as a brown oil. LC-MS (ESI) found: 148 [M+l]+.
4. Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)thio)acetate TFA salt
Figure imgf000173_0001
Intermediate 1
[0348] Stepl : To a solution of 2-(azeti din-3 -ylthio)acetic acid 2,2,2-trifluoroacetate salt (500 mg, 0.87 mmol), DIPEA (700 mg, 5.4 mmol) in CH3CN (3 mL) under N2 was added a solution of (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 390 mg, 0.87 mmol) in CH3CN (1 mL) dropwise at 0°C, followed by addition of DMAP (5 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran- 2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)thio)acetic acid (220 mg) as a colorless oil. LC-MS (ESI) found: 456 [M+l]+.
Step2: To a solution of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)thio)acetic acid (86 mg, 0.189 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.7 mL, 0.17 mmol) solution at -60°C with stirring. After lypholization, sodium 2-((l-((((3R,4S,5S,6R)-
5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)azetidin-3-yl)thio)acetate 85 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-£¾) d 5.29 (dt, J= 4.6, 2.7 Hz, 1H), 5.19 (td, J= 7.4, 3.7 Hz, 1H), 4.16 (t, J = 8.2 Hz, 2H), 3.79 - 3.61 (m, 3H), 3.52 (dd, 7= 11.0, 2.6 Hz, 1H), 3.28 (s, 3H), 2.93 (s, 2H),
2.84 (d, j= 4.4 Hz, 1H), 2.57 (t, j= 5.5 Hz, 2H), 2.18 (hept, j= 7.6, 7.1 Hz, 2H), 1.93 (td, j = 13.1, 12.2, 6.7 Hz, 1H), 1.82 - 1.67 (m, 6H), 1.61 (d, J= 1.3 Hz, 3H), 1.07 (s, 3H), 1.04 - 0.97 (m, 1H). Example 55: Preparation of sodium 2-((l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfonyl)acetate
Figure imgf000174_0001
5. Preparation of tert-butyl 3-((2-ethoxy-2-oxoethyl)sulfonyl)azetidine-l- carboxylate
Figure imgf000174_0002
[0349] To a solution of tert-butyl 3-[(2-ethoxy-2-oxoethyl)sulfanyl]azetidine-l-carboxylate (305 mg, 1.11 mmol) in DCM (4 mL), was added 3-chloroperoxybenzoic acid (477.85 mg, 2.77 mmol). The reaction mixture was stirred at 25 °C for 16 hours. TLC showed the reaction was complete. The slurry was diluted with DCM and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04 and concentrated under reduced pressure.
The residue was purified by flash chromatography to give tert-butyl 3-(2-ethoxy-2- oxoethanesulfonyl)azetidine-l-carboxylate (300 mg) as a white solid. LC-MS (ESI) found: 308 [M+H] +
6. Preparation of lithium 2-((l-(tert-butoxycarbonyl)azetidin-3-yl)sulfonyl)acetate
LiOH*H20 ^OLi
THF/H2O
Figure imgf000174_0003
.N'T 'O
Boc Boc
[0350] To a stirred solution of tert-butyl 3-(2-ethoxy-2-oxoethanesulfonyl)azetidine-l- carboxylate (100 mg, 0.32 mmol) in THF (3 mL) was added lithium hydroxide monohydrate (68.31 mg, 1.63 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h, and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give crude lithium 2-({ l-[(tert-butoxy)carbonyl]azetidin- 3-yl}sulfonyl)acetate (100 mg) as a colorless oil. LC-MS (ESI) found:280 [M+l]+.
7. Preparation of 2-(azetidin-3-ylsulfonyl)acetic acid
Boc
Figure imgf000175_0001
[0351] To a solution of lithium 2-({ l-[(tert-butoxy)carbonyl]azetidin-3- yl}sulfonyl)acetate (100 mg, 0.3 mmol) in DCM (2 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. It was concentrated under vacuum to give 2-(azeti din-3 - ylsulfonyl)acetic acid (70 mg) as a brown oil. LC-MS (ESI) found: 180[M+1]+.
8. Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfonyl)acetate
Figure imgf000175_0002
[0352] Step 1 : To a solution of 2-(azetidine-3-sulfonyl)acetic acid 2,2,2-trifluoroacetate salt (75.08 mg, 0.33 mmol), and DIPEA (0.33 mL, 2.0 mmol) in CH3CN (3 mL) under N2 was added dropwise a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 150 mg, 0.34 mmol) in CH3CN (1 mL) at 0°C, followed by addition of DMAP (4.1 mg, 0.033 mmol). The mixture was then stirred at r.t. for 16 h. The reaction mixture was concentrated under vacuum and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2-((l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)sulfonyl)acetic acid (72 mg) as a colorless oil. LC-MS (ESI) found: 488 [M+l]+.
Step 2: To a solution of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfonyl)acetic acid (72 mg, 0.15 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.34 mL) at -60°C with stirring. After lyophilization, sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfonyl)acetate (75.3 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6) d 5.31 (s, 1H), 5.20 (t, J= 7.5 Hz, 1H), 4.65 (dd, J= 14.1, 6.8 Hz, 1H), 4.08 (d, j= 9.0 Hz, 4H), 3.63 (s, 2H), 3.54 (dd, j= 11.0, 2.6 Hz, 1H), 3.28 (d, j= 4.6 Hz, 3H), 2.86 (d, j= 4.3 Hz, 1H), 2.59 (dd, j= 9.6, 5.2 Hz, 2H), 2.18 (dd, j= 14.9, 7.5 Hz, 2H), 1.96 (dd, J= 13.4, 9.2 Hz, 1H), 1.80 (dd, 7= 16.6, 7.1 Hz, 2H), 1.73 - 1.66 (m, 4H), 1.62 (s, 3H), 1.08 (s, 3H), 1.02 (d, J= 13.6 Hz, 1H).
Example 56: Preparation of sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptane-6-carboxylate
Figure imgf000176_0001
1. Preparation of 2-((((3/?,4,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptane-6-carboxylic acid
Figure imgf000176_0002
[0353] To a solution of 2-azaspiro[3.3]heptane-6-carboxylic acid 2,2,2-trifluoroacetate salt (94.64 mg, 0.67 mmol), and DIPEA (0.69 mL, 4.02 mmol) in C¾CN (4 mL) under N2 was added a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 300 mg, 0.67 mmol) in C¾CN (1 mL) dropwise at 0°C, followed by adding DMAP (8.24 mg, 0.067 mmol). The mixture was then stirred at r.t. for 16 hrs. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give 2-({[(3R,4S,5S,6R)-
5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-
6-yl]oxy}carbonyl)-2-azaspiro[3.3]heptane-6-carboxylic acid (209 mg) as a colorless oil. LC- MS (ESI) found: 450 [M+l]+.
2. Preparation of sodium 2-((((3/?,4,V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2- azaspiro [3.3] heptane-6-carboxylate
Figure imgf000177_0003
[0354] To a solution of 2-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)-2- azaspiro[3.3]heptane-6-carboxylic acid (100 mg, 0.223 mmol) in CH3CN (0.3 mL) was added 0.1 N NaOH (2.00 mL, 0.20 mmol) solution at -60°C with stirring. After lyophilization, sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran- 2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-azaspiro[3.3]heptane-6-carboxylate (100 mg) was obtained as white solid. 1H NMR (400 MHz, DMSO-d6) 85.28 (s, 1H), 5.20 (t, J = 7.5 Hz, 1H), 3.80 (d, J= 31.8 Hz, 4H), 3.51 (dd, J= 1.0, 2.5 Hz, 1H), 3.27 (s, 3H), 3.26 - 3.22
(m, 1H), 2.85 (d, J= 4.3 Hz, 1H), 2.56 (t, J= 5.8 Hz, 2H), 2.25 - 2.09 (m, 6H), 1.99 - 1.87 (m, 1H), 1.80 - 1.66 (m, 6H), 1.62 (s, 3H), 1.08 (s, 3H), 1.01 (d, J= 13.5 Hz, 1H).
Example 57: Preparation of sodium 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoate
Figure imgf000177_0001
1. Preparation of tert-butyl 3-formylazetidine-l-carboxylate
Figure imgf000177_0002
[0355] To the solution of tert-butyl 3-(hydroxymethyl)azetidine-l-carboxylate (7.0 g) in DCM (100 mL) was added Dess-Martin periodinane (24.0 g). The reaction was allowed to be stirred at r.t overnight. TLC showed the reaction was complete. Then the reaction was quenched with 10% Na2S203(aq), and extracted with EA. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 20 % ~ 50 % EA in PE) to give tert-butyl 3- formylazetidine-l-carboxylate (S-2, 6.0 g) as colorless oil. 1H NMR (400 MHz, CDCI3) d 9.78
(t, J = 1.7 Hz, 1H), 4.09 - 3.92 (m, 4H), 3.28 (dddd, J = 12.8, 8.1, 4.6, 2.3 Hz, 1H), 1.37 (d, J = 1.5 Hz, 9H).
2. Preparation of (Z)-3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3-oxoprop-l-en- l-yl)azetidine-l-carboxylate
Figure imgf000178_0001
[0356] To the solution of ethyl 2-((tert-butyldimethylsilyl)oxy)-2-(dimethoxyphosphoryl) acetate (15.9 g, prepared using literature procedure) in anhydrous THF (100 mL) was added LiHMDS (2 mol/L in THF) dropwise under N2 at -70 °C. After 30 min, tert-butyl 3- formylazetidine-l-carboxylate (S2, 6.0 g) in anhydrous THF (50 mL) was added to the mixture drop-wise at the same temperature. After 4 hours, TLC showed the reaction was complete. The reaction was quenched with NH4Cl (aq), and extracted with EA. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 5 % ~ 10 % EA in PE) to give (Z)-3 -^-((tert- butyl dimethylsilyl)oxy)-3 -ethoxy-3 -ox oprop-l-en-l -yl)azeti dine- l-carboxylate (S-3, 4.5 g) as a colorless oil. 1H NMR (400 MHz, CDCl3) d 6.06 (d, J = 9.4 Hz, 1H), 4.07 (q, J = 7.1 Hz, 2H), 4.00 (t, J = 8.4 Hz, 2H), 3.59 (dd, J = 8.3, 5.9 Hz, 2H), 3.54 - 3.40 (m, 1H), 1.30 (s, 9H), 1.18 (t, J = 7.1 Hz, 3H), 0.81 (s, 9H), 0.00 (s, 6H).
3. Preparation of tert-butyl 3-(3-ethoxy-2,3-dioxopropyl)azetidine-l-carboxylate
Figure imgf000178_0002
[0357] A solution of (Z)-3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3-oxoprop-l-en-l- yl)azetidine-l-carboxylate (800 mg) in DCM (20 mL) was treated with TBAF (1 M in THF, 4.0 mL) and stirred at r.t. for 4 hours. TLC showed the reaction was complete. The reaction was extracted with EA and washed with water. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 5 % ~ 10 % EA in PE) to give tert-butyl 3 -(3 -ethoxy-2,3 - dioxopropyl)azetidine-l-carboxylate (600 mg) as a colorless oil. 1H NMR (400 MHz,
Chloroform-d) d 4.21 (qd, J = 7.2, 1.1 Hz, 2H), 4.02 (td, J = 8.6, 1.1 Hz, 2H), 3.55 - 3.36 (m, 2H), 3.08 (dd, J = 7.5, 0.9 Hz, 2H), 2.81 (ddt, J = 13.3, 7.8, 3.8 Hz, 1H), 1.31 (d, J = 1.2 Hz, 9H), 1.26 (td, J = 7.1, 1.1 Hz, 3H).
4. Preparation of tert-butyl 3-(3-ethoxy-2,2-difluoro-3-oxopropyl)azetidine-l-carboxylate
Figure imgf000179_0001
[0358] A solution of tert-butyl 3 -(3 -ethoxy-2,3 -di oxopropyl)azeti dine- l-carboxylate (500 mg) in DCM (10 mL) was treated with (diethylamino)sulfur triflouride (594 mg) at 0 °C. The reaction was allowed to stir at r.t for 6 hrs, and was monitored with TLC till complete. The reaction was quenched with NaHCO,(aq) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The crude product, was used in the next step without further purification.
2. Preparation of lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2,2- difluoropropanoate
Figure imgf000179_0002
[0359] The residue from last step was dissolved in MeOH/H20(v/v, 3/1, 10 mL). To the solution was added LiOH H20 (77 mg) and the mixture was stirred at r.t for 6 hrs. TLC showed the reaction was complete. The solvent was removed under vacuum to give lithium 3-(l-(tert- butoxycarbonyl)azeti din-3 -yl)-2,2-difluoropropanoate (300 mg) as a yellow solid. LC MS: m/z (M-Li) 264.
3. Preparation of 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoic acid
Figure imgf000179_0003
[0360] To a solution of lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2,2- difluoropropanoate (300 mg) in DCM(5 mL) was added TFA (2.5 mL). The resulting mixture was stirred at room temperature for 3 hrs. TLC showed the reaction was complete. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.75 mL) was added dropwise while cooling the reaction in an ice bath. To the resulting solution was added (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 440 mg), and the mixture was stirred at r.t. for 6 hrs. TLC showed the reaction was complete. The reaction mixture was quenched with aqueous NaHC03 (1 M) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoic acid (36 mg). LC MS: (M-H) 472.
4. Preparation of sodium 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-
2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoate
Figure imgf000180_0001
[0361] To a solution of 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoic acid (36 mg) in MeCN (20 mL) was added aqueous NaOH (0.1 M) at -20 °C. The solvent was removed by lyophilization to give sodium 2,2- difluoro-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate (36 mg) as a white solid. LC MS: m/z (M-Na) 472. 1H MR (400 MHz, DMSO-d6) d 5.28 (s, 1H), 5.19 (ddq, J= 8.9, 7.3, 1.5 Hz, 1H), 3.93 (t, J= 8.4 Hz, 2H), 3.58 (t, J= 7.4 Hz, 2H), 3.51 (dd, J = 11.0, 2.6 Hz, 1H), 3.27 (s, 3H), 2.84 (d, J= 4.4 Hz, 1H), 2.74 (hept, J= 7.5 Hz, 1H), 2.58 - 2.53 (m, 2H), 2.16 (td, J= 16.3, 8.5 Hz, 5H), 1.93 (td, = 13.3, 4.5 Hz, 1H), 1.74 (d, J= 11.0 Hz, 2H), 1.70 (d, j= 1.4 Hz, 3H), 1.61 (d, j= 1.3 Hz, 3H), 1.07 (s, 3H), 1.00 (dt, j= 14.2, 3.6 Hz, 1H). 19F NMR (377 MHz, DMSO-d6) d -101.45, -101.53. Examples 58(a) and 58(b): Preparation of sodium 2-((l-((((3/?, S,5»S,6/?)-5-methoxy-4-
((2 ?,3 ?)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfinyl)acetate
Figure imgf000181_0003
1. Preparation of tert-butyl 3-((2-ethoxy-2-oxoethyl)sulfinyl)azetidine-l-carboxylate
Figure imgf000181_0001
[0362] To a solution of tert-butyl 3-[(2-ethoxy-2-oxoethyl)sulfanyl]azetidine-l-carboxylate (lg, 3.6 mmol) in DCM (10 mL), was added 3-chloroperoxybenzoic acid (0.63 g, 3.6 mmol). The reaction mixture was stirred at 25 °C for 16 hours. TLC showed the reaction was complete.
The slurry was diluted with DCM and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give tert-butyl 3-(2-ethoxy-2-oxoethanesulfmyl)azetidine-l- carboxylate (830 mg) as a white solid. LC-MS (ESI) found: 292 [M+H] +
2. Preparation of lithium 2-((l-(terf-butoxycarbonyl)azetidin-3-yl)sulfinyl)acetic acid
Figure imgf000181_0002
[0363] To a stirred solution of tert-butyl 3-(2-ethoxy-2-oxoethanesulfmyl)azetidine-l- carboxylate (350 mg, 1.20 mmol) in THF (3 mL), was added lithium hydroxide monohydrate (151.3 mg, 3.60 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give crude lithium 2-({ l-[(tert-butoxy)carbonyl]azetidin- 3-yl}sulfmyl)acetate (mg) as a colorless oil. LC-MS (ESI) found:264 [M+l]+. 3. Preparation of 2-(azetidin-3-ylsulfinyl)acetic acid
Figure imgf000182_0001
k OH
Bocri .
[0364] To a solution of lithium 2-({l-[(tert-butoxy)carbonyl]azetidin-3-yl}sulfmyl)acetate (300 mg, 1.14 mmol) in DCM (5 mL) was added TFA (1 mL). After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The mixture was concentrated to give 2-(azetidine-3-sulfmyl)acetic acid (200 mg) as a brown oil. LC-MS (ESI) found: 164 [M+l]+.
4. Preparation of sodium 2-((l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfinyl)acetate
Figure imgf000182_0002
[0365] Step 1 : To a solution of 2-(azetidine-3-sulfmyl)acetic acid 2,2,2-trifluoroacetate salt (200 mg, 1.10 mmol), DIPEA (1.10 mL, 6.62 mmol) in MeCN (5 mL) under N2 was added dropwise a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 494 mg, 1.10 mmol) in CH3CN (1 mL) at 0°C, followed by addition of DMAP (13.6 mg, 0.110 mmol). The mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated. The residue was purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) and chiral SFC resolution (Instrument: Waters Thar 80 preparative SFC; Column: ChiralPak IG, 250x21.2mm I.D., 5pm; Mobile phase: C02 for A and methanol (0.1%NH3Ή20) for B; Gradient: B 30%; Flow rate: 50 mL /min; Back pressure: 100 bar; Column temperature: 35°C; Wavelength: 220nm) to give separated diasteriomers of 2-((l- ((((3//,4k,5k,6//)-5-methoxy-4-((2//,3//)-2-methyl-3-(3-methylbut-2-en- l -yl)oxiran-2-yl)- l - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)sulfmyl)acetic acid (70 mg and 65 mg) as colorless oils. LC-MS (ESI) found: 472 [M+l]+.
Step 2: To a solution of 2-(( l -((((3//,4k,5k,6//)-5-methoxy-4-((2//,3//)-2- ethyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfmyl)acetic acid (70 mg, 0.14 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.34 mL) solution at -60°C with stirring. After lyophilization, sodium 2-((l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfmyl)acetate (70 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6 ) d 5.29 (s, 1H), 5.20 (t, J= 7.3 Hz, 1H), 4.25 (d, J= 27.4 Hz, 1H), 4.13 (s, 1H), 4.00 (dd, j= 11.5, 6.1 Hz, 2H), 3.92 (s, 1H), 3.53 (dd, j= 11.0, 2.4 Hz, 1H), 3.28 (s, 3H), 3.25 (s, 1H), 3.15 (d, J= 14.0 Hz, 1H), 2.85 (d, J= 4.2 Hz, 1H), 2.59 (t, J= 6.0 Hz, 2H), 2.24 - 2.11 (m, 2H), 1.97 (s, 1H), 1.78 (d, 7= 10.4 Hz, 2H), 1.71 (s, 4H), 1.62 (s, 3H), 1.07 (d,
J= 9.0 Hz, 3H), 1.02 (d, J= 13.9 Hz, 1H).
Step 3 : To a solution of 2-(( l -((((3/^4.V,5.V,6/^)-5-methoxy-4-((2/^3/^)-2- ethyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfmyl)acetic acid (65 mg, 0.13 mmol) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.20 mL) solution at -60°C with stirring. After lyophilization, sodium 2-((l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)sulfmyl)acetate (65 mg) was obtained as a white solid. 1H MR (400 MHz, DMSO-d6) d 5.29 (s, 1H), 5.20 (t, J = 7.5 Hz, 1H), 4.23 (s, 1H), 4.13 (t, J = 8.1 Hz, 1H), 4.01 (s, 3H), 3.53 (dd, J = 11.0, 2.5 Hz, 1H), 3.29 (s, 3H), 3.13 (d, J = 13.8 Hz, 2H), 2.85 (d, J = 4.3 Hz, 1H), 2.57 (d, J = 4.3 Hz, 2H), 2.18 (dd, J = 14.4, 7.4 Hz, 2H), 1.99 - 1.91 (m,
1H), 1.78 (d, J = 10.7 Hz, 2H), 1.69 (d, J = 19.1 Hz, 4H), 1.62 (s, 3H), 1.08 (s, 3H), 1.02 (d, J = 12.0 Hz, 1H).
Example 59: Preparation of sodium l-(l-((((3/?, S,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)-6-oxo- 1 ,6-dih dropyridine-3-carboxylate
Figure imgf000183_0001
1. Preparation of methyl l-(l-(terf-butoxycarbonyl)azetidin-3-yl)-6-oxo-l,6- dihydropyridine-3-carboxylate
Figure imgf000184_0001
[0366] To a solution of tert-butyl 3-iodoazetidine-l-carboxylate (1.25 g, 4.41 mmol) and methyl 6-hydroxypyridine-3-carboxylate (0.676 g, 4.42 mmol) in DMF (5 mL), was added Cs2C03 (2.16 g, 6.62 mmol). The reaction mixture was stirred at 25 °C for 16 hours. TLC showed the reaction was complete. The slurry was diluted with EA, and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give methyl l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-6-oxo-l,6-dihydropyridine-3-carboxylate (270 mg) as a white solid. LC-MS (ESI) found: 309 [M+H] +.
2. Preparation of l-(azetidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid, trifluoroacetate salt
Figure imgf000184_0002
[0367] Step 1 : To a stirred solution of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-6- oxo-l,6-dihydropyridine-3-carboxylate (217 mg, 0.71 mmol) in THF (3 mL), was added LiOH H20 (88.68 mg, 2.11 mmol) in H20 (lmL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give crude target product as a colorless oil. LC- MS (ESI) found: 295 [M+l]+. Used in the next reaction step without further purification.
Step 2: The crude target product was dissolved in DCM (3 mL), then TFA (1 mL) was added. After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The reaction mixture was concentrated to give l-(azetidin- 3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid, TFA salt (150 mg) as a brown oil. LC-MS (ESI) found: 195 [M+l]+.
3. Preparation of l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-6- oxo-1, 6-dihydropyridine-3-carboxylic acid
Figure imgf000185_0002
[0368] To a solution of l-(azetidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid trifluoroacetate salt (150 mg, 0.69 mmol), and DIPEA (0.69 mL, 4.03 mmol) in CH3CN (4 mL) was added dropwise a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate
(Intermediate 1, 300 mg, 0.67 mmol) in CH3CN (2 mL) at 0°C, followed by the addition of DMAP (8.24 mg, 0.067 mmol). Then the mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)azetidin-3-yl]-6-oxo-l,6-dihydropyridine-3-carboxylic acid (160 mg) as a colorless oil. LC-MS (ESI) found: 503 [M+l]+.
4. Preparation of sodium l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- 6-oxo- 1 ,6-dih dropyridine-3-carboxylate
Figure imgf000185_0001
[0369] To a solution of l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-6- oxo-l,6-dihydropyridine-3 -carboxylic acid (lOOmg, 0.199 mmol) in CH3CN (0.3 mL) was added 0.1 N NaOH (1.79 mL, 0.179 mmol) solution at -60°C with stirring. After
lyophilization, sodium l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-6-oxo-l,6- dihydropyridine-3-carboxylate (100 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6) d 8.07 (s, 1H), 7.84 (dd, J= 9.2, 2.2 Hz, 1H), 6.24 (d, J= 9.3 Hz, 1H), 5.31 (d, J = 20.8 Hz, 2H), 5.19 (t, J= 7.4 Hz, 1H), 4.30 (t, J= 8.6 Hz, 2H), 4.09 (dd, J= 9.1, 5.7 Hz, 2H), 3.55 (dd, J = 10.9, 2.6 Hz, 1H), 3.31 (d, 7= 3.4 Hz, 3H), 2.85 (d, 7= 4.4 Hz, 1H), 2.58 (t, j = 5.9 Hz, 2H), 2.16 (dt, J= 14.3, 6.9 Hz, 2H), 1.98 (t, J= 11.6 Hz, 1H), 1.86 (d, 7= 11.5 Hz, 1H), 1.79 (d, J= 10.9 Hz, 1H), 1.73 (d, J= 15.8 Hz, 4H), 1.61 (s, 3H), 1.09 (d, J= 3.5 Hz, 3H),
1.03 (d, J= 12.8 Hz, 1H).
Example 60: Preparation of sodium l-(l-((((3/?, S,5»S,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)-2-oxo- 1 ,2-dih dropyridine-4-carboxylate
Figure imgf000186_0001
1. Preparation of methyl l-(l-(terf-butoxycarbonyl)azetidin-3-yl)-2-oxo-l,2- dihydropyridine-4-carboxylate
Figure imgf000186_0002
[0370] To a solution of tert-butyl 3-iodoazetidine-l-carboxylate (1.0 g, 3.5 mmol) and methyl 2-hydroxypyridine-4-carboxylate (0.54 g, 3.53 mmol) in DMF (15 mL), was added CS2CO3 (1.73 g, 5.30 mmol). The reaction mixture was stirred at 60 °C for 16 hours. TLC showed the reaction was complete. The slurry was diluted with EA and washed with water and brine. The combined organic layers were dried over anhydrous Na2S04, concentrated under reduced pressure and purified by flash chromatography to give methyl l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-2-oxo-l,2-dihydropyridine-4-carboxylate (208 mg) as a white solid. LC-MS (ESI) found: 309 [M+H] +
2. Preparation of l-(azetidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid
Figure imgf000186_0003
[0371] Step 1 : To a stirred solution of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-2- oxo-l,2-dihydropyridine-4-carboxylate (200 mg, 0.65 mmol) in THF (3 mL), was added Li0H H20 (81.74 mg, 1.95 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give the crude product as a colorless oil which was used in the next reaction step without further purification. LC-MS (ESI) found: 295
[M+l]+.
Step 2: The crude product was dissolved in DCM (3 mL), TFA (1 mL) was added and the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The reaction mixture was concentrated to give l-(azeti din-3 -yl)-2-oxo- 1,2- dihydropyridine-4-carboxylic acid, TFA salt (150 mg) as a brown oil. LC-MS (ESI) found: 195 [M+l]+.
3. Preparation of sodium l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- 2-oxo- 1 ,2-dih dropyridine-4-carboxylate
Figure imgf000187_0001
[0372] Step 1 : Under N2 atmosphere, to a solution of l-(azeti din-3 -yl)-2-oxo- 1,2- dihydropyridine-4-carboxylic acid trifluoroacetate salt (141.03 mg, 0.58 mmol) and DIPEA (524.6 mg, 4.07 mmol) in MeCN (8 mL) was added dropwise a solution of (3R,4S,5S,6R)-5- methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl 4-nitrophenyl carbonate (Intermediate 1, 260 mg, 0.58 mmol) in CH3CN (1 mL) at 0°C, followed by adding DMAP (3.58 mg, 0.029 mmol). The mixture was then stirred at r.t. for 16 h. TLC showed the reaction was complete. The reaction mixture was concentrated under vacuum and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give l-(l- ((((3//,4,V,5ri,6//)-5-methoxy-4-((2//,3//)-2-methyl-3-(3-methylbut-2-en- l -yl)oxiran-2-yl)- l - oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2-oxo-l,2-dihydropyridine-4-carboxylic acid (100 mg) as a colorless oil. LC-MS (ESI) found: 503 [M+l]+.
Step 2: To a solution of l-(l-((((3i?,4ri',5ri,,6i?)-5-methoxy-4-((2i?,3i?)-2-methyl-3-(3-methylbut- 2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azeti din-3 -yl)-2-oxo- 1 ,2- dihydropyridine-4-carboxylic acid (100 mg, 0.199 mmoL) in CH3CN (0.5 mL) was added 0.1 N NaOH (1.8 mL) solution at -60°C with stirring. After lyophilization, sodium l-[l- ({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l- oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-2-oxo-l,2-dihydropyridine-4-carboxylate (100 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6) d 7.63 (d, J= 7.4 Hz, 1H), 6.64 - 6.57 (m, 2H), 5.31 (d, J= 18.2 Hz, 2H), 5.19 (t, J= 7.4 Hz, 1H), 4.26 (t, = 8.3 Hz, 2H), 4.12 (s, 2H), 3.55 (dd, J= 11.0, 2.5 Hz, 1H), 3.31 (s, 3H), 2.85 (d, J= 4.3 Hz, 1H), 2.57 (dd, J= 9.8, 5.3 Hz, 2H), 2.18 (dd, J= 12.9, 6.3 Hz, 2H), 2.02 - 1.92 (m, 1H), 1.89 - 1.73 (m, 3H), 1.71 (s, 3H), 1.61 (s, 3H), 1.09 (d, j= 4.1 Hz, 3H), 1.03 (d, j= 13.0 Hz, 1H).
Example 61: Preparation of sodium l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)-2-oxo- 1 ,2-dih dropyridine-3-carboxylate
Figure imgf000188_0001
1. Preparation of methyl l-(l-(terf-butoxycarbonyl)azetidin-3-yl)-2-oxo-l,2- dihydropyridine-3-carboxylate
Figure imgf000188_0002
[0373] Step 1 : A heterogeneous mixture of methyl 2-oxo-2H-pyran-3-carboxylate (100 mg, 0.65 mmol) and tert-butyl 3-aminoazetidine-l-carboxylate (111 mg, 0.65 mmol) in DMF (5 mL) were stirred at room temperature for 3 h.
Step 2: The reaction mixture was then treated with 1 -Ethyl-3 -(3
dimethylaminopropyl)carbodiimide (172.97 mg, 0.90 mmol) and DMAP (5.5 mg, 0.045 mmol) at room temperature and the resulting solution was stirred for 12 h. TLC showed the reaction was complete. The reaction mixture was quenched with 1N aqueous HC1 and the solution was extracted with ethyl acetate (4 x 5 mL). The combined organic extracts were washed with 10% aqueous LiCl (3 x 7 mL), dried over anhydrous Na2S04 and filtered. The filtrate
was concentrated under reduced pressure, and the residue was purified by flash
chromatography (silica gel, 20 % ~ 50 % EA in PE) to afford methyl l-{ l-[(tert- butoxy)carbonyl]azetidin-3-yl}-2-oxo-l,2-dihydropyridine-3-carboxylate (130 mg) as a solid. 2. Preparation of lithium l-(l-(ter#-butoxycarbonyl)azetidin-3-yl)-2-oxo-l,2- dihydropyridine-3-carboxylate
Figure imgf000189_0001
[0374] To a stirred solution of methyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-2-oxo-l,2- dihydropyridine-3-carboxylate (130 mg, 0.42 mmol) in THF (3 mL), was added LiOH H20 (53.0 mg, 1.27 mmol) in H20 (1 mL) in one portion at 25°C. The reaction mixture was stirred at r.t. for 1.0 h, and the starting material was completely consumed by TLC. The reaction mixture was concentrated in vacuo to give crude lithium l-{ l-[(tert-butoxy)carbonyl]azetidin- 3-yl}-2-oxo-l,2-dihydropyridine-3-carboxylate (150 mg) as a colorless oil. LC-MS (ESI) found: 295 [M+l]+.
3. Preparation of l-(azetidin-3-yl)-2-oxo-l,2-dihydropyridine-3-carboxylic acid, trifluoroacetate salt
Figure imgf000189_0002
[0375] The crude lithium l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-2-oxo-l,2- dihydropyridine-3-carboxylate (150 mg, 0.51 mmol) was dissolved in DCM (3 mL), and TFA (1 mL) was added. After the addition, the reaction mixture was stirred at room temperature for 3 hours. TLC showed the reaction was complete. The reaction mixture was concentrated under vacuum to give l-(azeti din-3 -yl)-2-oxo-l,2-dihydropyridine-3 -carboxylic acid, TFA salt (100 mg) as a brown oil. LC-MS (ESI) found: 195 [M+l]+.
4. Preparation of l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2- oxo-1, 2-dihydropyridine-3-carboxylic acid
Figure imgf000189_0003
[0376] To a solution of l-(azeti din-3 -yl)-2-oxo-l,2-dihydropyridine-3 -carboxylic acid, trifluoroacetate salt (86.78 mg, 0.45 mmol) and DIPEA (0.46 mL, 2.68 mmol) in C¾CN (4 mL) was added dropwise a solution of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate
(Intermediate 1, 200 mg, 0.447 mmol) in CH3CN (1 mL) at 0°C, followed by addition of DMAP (5.50 mg, 0.045 mmol). Then the mixture was stirred at r.t. for 16 h. The reaction mixture was concentrated, and the residue was purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-2-oxo-l,2- dihydropyridine-3 -carboxylic acid (69 mg) as a colorless oil. LC-MS (ESI) found: 503 [M+l]+.
5. Preparation of sodium l-(l-((((3/?,4A,5A,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- 2-oxo- 1 ,2-dih dropyridine-3-carboxylate
Figure imgf000190_0001
[0377] To a solution of l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-2- oxo-l,2-dihydropyridine-3 -carboxylic acid (69 mg, 0.14 mmol) in CH3CN (0.3 mL) was added 0.1 N NaOH (1.24 mL, 0.124 mmol) solution at -60°C with stirring. After lyophilization, sodium l-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-2-oxo-l,2- dihydropyridine-3-carboxylate (60 mg) was obtained as a white solid. 1H NMR (400 MHz, DMSO-d6) d 7.68 (d, J= 24.2 Hz, 2H), 6.28 (s, 1H), 5.34 (s, 1H), 5.19 (s, 2H), 4.26 (t, J= 8.5 Hz, 2H), 4.17 - 4.10 (m, 2H), 3.55 (d, J= 10.6 Hz, 1H), 3.31 (s, 3H), 2.85 (d, J= 4.2 Hz, 1H), 2.59 - 2.55 (m, 2H), 2.18 (d, J= 5.2 Hz, 2H), 1.95 (d, J= 9.2 Hz, 1H), 1.80 (d, 7= 11.9 Hz, 2H), 1.76 (s, 1H), 1.71 (s, 4H), 1.61 (s, 3H), 1.09 (s, 3H), 1.02 (d, J= 12.5 Hz, 1H).
Example 62: Preparation of sodium 4-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl] oxy} carbonyl)azetidin-3-yl]- 1 ,3-thiazole-2-carboxylate
Figure imgf000191_0001
1. Preparation of benzyl 3-(carbonochloridoyl)azetidine-l-carboxylate
Figure imgf000191_0002
[0378] To a stirred solution of l-[(benzyloxy)carbonyl]azetidine-3-carboxylic acid (2.35 g, 9.99 mmol) in DCM (5 mL) was added oxalyl chloride (4.75 g, 29.97 mmol) at 0 °C, followed by a catalytic amount of DMF (2 drops). The reaction mixture was stirred at 0°C for 15 min and then stirred at room temperature for 1 h. The solvent was removed under reduced pressure to afford benzyl 3-(carbonochloridoyl)azetidine-l-carboxylate (2.4 g) as a colorless oil.
2. Preparation of benzyl 3-(2-diazoacetyl)azetidine-l-carboxylate
Figure imgf000191_0003
[0379] To a stirred solution of benzyl 3-(carbonochloridoyl)azetidine-l-carboxylate (2.1 g, 8.28 mmol) in THF (50 mL) was added (diazomethyl)trimethylsilane (12.41 mL, 12.42 mmol) at 0 °C. The reaction mixture was stirred at 0°C for 15 min and then stirred at room temperature overnight. TLC showed the reaction was complete. After removal of the solvent under reduced pressure, EtOAc and sat. aq. NaHC03 were added. The aqueous layer was extracted with EtOAc and the combined organic layers was washed with brine and dried over anhydrous Na2S04. The solvent was removed under reduced pressure to afford benzyl 3-(2- diazoacetyl)azetidine-l-carboxylate (1.8 g) as a colorless oil.
3. Preparation of benzyl 3-(2-bromoacetyl)azetidine-l-carboxylate
Figure imgf000192_0001
[0380] To a stirred solution of benzyl 3-(2-diazoacetyl)azetidine-l-carboxylate (1.8 g, 6.92 mmol) in Et20 (5 mL) was added HBr (1.43 g, 8.30 mmol) at 0 °C . The reaction mixture was stirred at 0°C for 1 hr. TLC showed the starting material had disappeared. The reaction mixture was neutralized with NaHCO, aq, and the aqueous layer was extracted with EtOAc. The combined organic layers was washed with brine and dried over anhydrous Na2S04. The solvent was removed under reduced pressure to afford benzyl 3-(2-bromoacetyl)azetidine-l- carboxylate (1.9 g) as a colorless oil.
4. Preparation of benzyl 3-propanoylazetidine-l-carboxylate
Figure imgf000192_0002
[0381] To a stirred solution of benzyl 3-(2-bromoacetyl)azetidine-l-carboxylate (1.1 g, 3.52 mmol) in ethanol (20 mL) was added ethyl 2-amino-2-thioxoacetate (0.46 g, 3.52 mmol). The reaction mixture was stirred at l00°C for 3 hr. TLC showed the reaction was complete. After removal of the solvent under reduced pressure, the residue was partitioned between water and EtOAc. The aqueous layer was extracted with EtOAc, and the combined organic layer was washed with brine and dried over anhydrous Na2S04. The solvent was removed under reduced pressure, and the residue was purified by silica gel column eluting with PE/EtOAc (100:1 to 1 : 1) to afford ethyl 4-{ l-[(benzyloxy)carbonyl]azetidin-3-yl}-l,3-thiazole-2-carboxylate (330 mg) as a colorless oil. LC-MS (ESI) found: 347 (M+H)+.
5. Preparation of ethyl 4-(azetidin-3-yl)-l,3-thiazole-2-carboxylate
Figure imgf000192_0003
[0382] To a solution of ethyl 4-{ l-[(benzyloxy)carbonyl]azetidin-3-yl}-l,3-thiazole-2- carboxylate (330 mg, 0.95 mmol) in DCM (10 mL) was added trichloroborane (2.85 mL,
2.85 mmol, 1 M DCM solution) at 0°C under N2 atmosphere, and the reaction was stirred at room temperature overnight. TLC showed the reaction was complete. The reaction mixture was neutralized with aq. NaHCO, and the solvent was removed under reduced pressure to give ethyl 4-(azetidin-3-yl)-l,3-thiazole-2-carboxylate (180 mg) as a colorless oil. LC-MS (ESI) found: 213 (M+H)+.
6. Preparation of 4-{l-[(tert-butoxy)carbonyl]azetidin-3-yl}-l,3-thiazole-2- carboxylic acid
Figure imgf000193_0001
[0383] To a stirred solution of ethyl 4-(azetidin-3-yl)-l,3-thiazole-2-carboxylate (180 mg, 0.85 mmol) in THF (5 mL) was added di-tert-butyl dicarbonate (0.27 mL, 1.27 mmol) followed by NaOH (1.27 mL, 2.54 mmol). The mixture was stirred at room temperature for 30 min.
TLC showed the reaction was complete. EtOAc and water were added to the reaction mixture, and the two layers were separated. The aqueous layer was was acidified with 1N HC1 to pH=3 and then extracted with DCM. The combined organic layer was washed with brine and dried over anhydrous Na2S04. The solvent was removed under reduced pressure and the residue was purified by silica gel chromatography eluting with DCM/MeOH (40 : 1) to afford 4-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-l,3-thiazole-2-carboxylic acid (160 mg) as a colorless oil.
7. Preparation of 4-(azetidin-3-yl)-l,3-thiazole-2-carboxylic acid, trifluoroacetic acid salt
TFA
Figure imgf000193_0002
[0384] To a solution of 4-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-l,3-thiazole-2-carboxylic acid (150 mg, 0.52 mmol) in DCM (2 mL) was added TFA (1 mL) dropwise at 0 °C. The reaction was stirred at room temperature for lhr. TLC showed the reaction was complete. The mixture was concentrated under reduced pressure to give 4-(azeti din-3 -yl)- 1,3 -thiazole-2- carboxylic acid trifluoroacetic acid salt (150 mg) as a yellow syrup, which was directly used in the next step without further purification. LC-MS (ESI) found: 185 (M+H)+. 8. Preparation of (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 8-ethyl-5-oxa-2,8-diazaspiro[3.5]nonane-2- carboxylate
Figure imgf000194_0001
[0385] To a solution of 4-(azetidin-3-yl)-l,3-thiazole-2-carboxylic acid (150 mg, 0.49 mmol) in MeCN (4 mL) was added DIPEA (315.14 mg, 2.44 mmol) dropwise at 0°C. The mixture was stirred at 0°C for 10 min, and (3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate
(Intermediate 1, 218.64 mg, 0.49 mmol) was added. After addition was complete, the reaction was stirred at room temperature overnight under a N2 atmosphere. TLC showed the reaction was complete. The mixture was then concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with dichloromethane, and the DCM solution was washed with ammonium acetate buffer (pH 4.0, 20 mL) and 5% sodium bicarbonate solution (20 mL X 1). The organic layer was dried over anhydrous Na2S04, filtered and concentrated to give the crude product. The residue was purified by column
chromatography on silica gel (eluent: dichloromethane: methanol = 100 : 1 to 10 : 1) and further purified by prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 H20) to give 4-[ 1 -({ [(3R,4S, 5 S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3 -(3 -methylbut-2-en- 1 -yl)oxiran- 2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-l,3-thiazole-2-carboxylic acid (51 mg) as a colorless oil. LC-MS (ESI) found: 493 [M+l]+.
9. Preparation of sodium 4-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]-l ,3-thiazole-2-carboxylate
Figure imgf000194_0002
[0386] To a solution of 4-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-l,3- thiazole-2-carboxylic acid (51 mg, 0.10 mmol) in acetonitrile (1 mL) was added 5 mL of 0.1 N aq. NaOH (0.93 mL, 0.09 mmol) solution at -50 °C. The mixture was lyophilized overnight to give sodium 4-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-l,3-thiazole-2- carboxylate(50 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) d 7.52 (d, J = 31.7 Hz, 2H), 5.87 (s, 1H), 5.32 (s, 1H), 5.18 (s, 2H), 4.03 (d, J = 5.4 Hz, 2H), 3.54 (dd, J = 10.7, 2.3 Hz, 1H), 3.29 (s, 3H), 2.84 (d, J = 4.3 Hz, 1H), 2.57 (d, J = 4.3 Hz, 1H), 2.23 - 2.12 (m, 2H), 1.99 - 1.78 (m, 3H), 1.78 - 1.63 (m, 5H), 1.61 (s, 3H), 1.14 - 0.99 (m, 4H).
Example 63: Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-2-oxopropanoate
Figure imgf000195_0001
1 . TFA, DCM 0.1 M NaOH(aq)
2. DIPEA, Intermediate 1
MeCN
Figure imgf000195_0002
Figure imgf000195_0003
1. Preparation of tert-butyl 3-(3-ethoxy-2,3-dioxopropyl)azetidine-l-carboxylate
Figure imgf000195_0004
[0387] A solution of (Z)-3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3-oxoprop-l-en-l- yl)azetidine-l-carboxylate (800 mg) in DCM (20 mL) was treated with TBAF (1 M in THF, 4.0 mL) and stirred at r.t. for 4 hours. TLC showed the reaction was complete. The reaction was extracted with EA and washed with water. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 5 % ~ 10 % EA in PE) to give tert-butyl 3 -(3 -ethoxy-2,3 - dioxopropyl)azetidine-l-carboxylate (600 mg) as a colorless oil. 1H NMR (400 MHz,
Chloroform-d) d 4.21 (qd, J = 7.2, 1.1 Hz, 2H), 4.02 (td, J = 8.6, 1.1 Hz, 2H), 3.55 - 3.36 (m, 2H), 3.08 (dd, J = 7.5, 0.9 Hz, 2H), 2.81 (ddt, J = 13.3, 7.8, 3.8 Hz, 1H), 1.31 (d, J = 1.2 Hz, 9H), 1.26 (td, J = 7.1, 1.1 Hz, 3H). 2. Preparation of lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2-oxopropanoate
Figure imgf000196_0001
[0388] To a solution of tert-butyl 3 -(3 -ethoxy-2,3 -di ox opropyl)azeti dine- l-carboxylate (135 mg) in MeOH/H20(v/v, 3/1, 10 mL) was added LiOH H20 (22 mg). The mixture was stirred at r.t for 6 hrs. TLC showed the reaction was complete. The solvent was removed under vacuum to give lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2-oxopropanoate (123 mg) as a white solid. LC MS: m/z (M-Li) 242.
3. Preparation of 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2- oxopropanoic acid
Figure imgf000196_0002
[0389] To a solution of lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2-oxopropanoate (123 mg) in DCM (2.5 mL) was added TFA (2.5 mL). The resulting mixture was stirred at room temperature overnight. TLC showed the reaction was complete. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling the reaction in an ice-bath. To the resulting solution was added
(3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 282 mg), and the mixture was stirred at room temperature for 6 hrs. TLC showed the reaction was complete. The reaction mixture was quenched with aqueous NaHCO, (1 M) to pH = 6-7 and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2- oxopropanoic acid (42 mg) as a colorless oil. LC MS: m/z (M+l)+ 452.
4. Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-
2-oxopropanoate
Figure imgf000197_0001
[0390] To a solution of 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2- oxopropanoic acid (42 mg) in MeCN (20 mL) was added aqueous NaOH (0.1 M, ) at -20 °C. The solvent was removed by lyophilization to give sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-2-oxopropanoate (46 mg) as a white foam. LC MS: m/z (M-Na) 450. 1H NMR (400 MHz, DMSO-d6) d 5.28 (s, 1H), 5.20 (t, J= 7.6 Hz, 1H), 3.97 (t, J= 8.1 Hz, 2H), 3.52 (dd, j= 11.0, 2.6 Hz, 3H), 3.28 (s, 3H), 2.85 (d, j= 4.4 Hz, 1H), 2.80 (s, 2H), 2.57 (dd, J= 8.1, 5.1 Hz, 2H), 2.17 (q, J= 15.3, 1 1.3 Hz, 3H), 1.93 (dt, J= 13.6, 6.8 Hz, 1H), 1.75 (d, 7= 11.0 Hz, 2H), 1.71 (s, 4H), 1.61 (d, 7= 1.3 Hz, 3H), 1.08 (s, 3H), 1.01 (d, 7= 13.6 Hz, 1H).
Example 64: Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3-carboxylate
Figure imgf000197_0002
1. Preparation of tert-butyl (Z)-2-((dimethylamino)methylene)-3-oxobutanoate
Figure imgf000197_0003
[0391] To a mixture of tert-butyl 3-oxobutanoate (5.4 g, 6.3 mmol) in dioxane (30 mL) was added l, l-dimethoxy-N,N-dimethylmethanamine (19.5 g, 164 mmol). The reaction mixture was stirred at r.t. for 16 h under N2 atmosphere. After completion of the reaction, the resulting mixture was added to water and extracted with EA. The combined organic layers was washed with brine, dried over anhydrous Na2S04, and concentrated to give crude tert-butyl (Z)-2- ((dimethylamino)methylene)-3-oxobutanoate (, 7.2 g), which was used in the next step without further purification.
2. Preparation of tert-butyl 4-oxo-4H-pyran-3-carboxylate
Figure imgf000198_0001
[0392] To a solution of tert-butyl (2Z)-2-[(dimethylamino)methylidene]-3-oxobutanoate (2.4 g, 11.25 mmol) in 70 mL THF was added potassium /-butoxide (3.15 g, 28.1 mmol) at 0°C. The mixture was stirred at 0°C for 30 mins. Then ethyl formate (0.9 mL, 11.3 mmol) was added dropwise to the mixture. The reaction mixture was stirred at r.t. overnight. The reaction was monitored by TLC and LCMS. It showed the start material was not entirely consumed. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with EA and washed with water and brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 20: 1 to 10: 1) to give tert-butyl 3-[N-methyl(2R,3R)-oxolane-2-amido]azetidine-l-carboxylate (145 mg).
3. Preparation of tert-butyl l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-4-oxo-l,4- dihydropyridine-3-carboxylate
Figure imgf000198_0002
[0393] To a solution of tert-butyl 4-oxo-4H-pyran-3-carboxylate (145 mg, 0.74 mmol) in 5 mL of EtOH was added tert-butyl 3-aminoazetidine-l-carboxylate (127.2 mg, 0.74 mmol) at r.t. The reaction mixture was then stirred at 80 °C for 16 hours. The reaction was monitored by TLC and LCMS and showed the start material was consumed. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was diluted with EA, washed with water and brine, dried over anhydrous Na2S0 and concentrated under reduced pressure to give a residue, which was
purified by column chromatography (DCM/ MeOH, 20: 1) on silica gel to afford tert-butyl 1- (l-(tert-butoxycarbonyl)azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3-carboxylate (200 mg) as a colorless oil. 4. Preparation of l-(azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3-carboxylic acid
Figure imgf000199_0001
[0394] To a solution of tert-butyl l-{ l-[(tert-butoxy)carbonyl]azetidin-3-yl}-4-oxo-l,4- dihydropyridine-3-carboxylate (200 mg, 0.57 mmol) in DCM (4 mL) was added TFA (2 mL) at 0 °C. The reaction mixture was then slowly warmed to r.t. and stirred for 1.5 h. TLC showed the reaction was complete. The reaction mixture was concentrated to give a residue, which was used in the next step without further purification.
5. Preparation of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-4- oxo-1, 4-dihydropyridine-3-carboxylic acid
Figure imgf000199_0002
[0395] To a solution of l-(azeti din-3 -yl)-4-oxo-l,4-dihydropyridine-3 -carboxylic acid (200 mg, 0.65 mmol) in MeCN (5 mL) was added DIPEA (0.4 mL, 2.4 mmol) dropwise at 0 °C. The mixture was stirred at 0 °C for 10 min, then (3i?,4ri',5ri', 6/?)-5-methoxy-4-[(2i?,3i?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenylcarbonate
(Intermediate 1, 232.5 mg, 0.6 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3Ή20) to give l-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azeti din-3 -yl)-4-oxo-l,4-dihydropyridine-3 -carboxylic acid (65 mg) as a colorless oil.
6. Preparation of sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)- 4-oxo- 1 ,4-dih dropyridine-3-carboxylate
Figure imgf000200_0001
[0396] To a mixture of l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-4-oxo- l,4-dihydropyridine-3 -carboxylic acid (65 mg, 0.12 mmol) in MeCN (1 mL) was added 0.1 M NaOH (1.2 mL) at -60 °C with stirring. The reaction mixture was lyophilized to give sodium 1- (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3-carboxylate (51 mg) as a white solid. LC-MS (ESI) m/z (M+l): 503. 1H NMR (400 MHz, DMSO-d6) d 8.27 (s, 1H), 7.87 (s, 1H), 6.24 (s, 1H), 5.35 (s, 1H), 5.19 (t, J= 7.3 Hz, 1H), 5.07 (m, 1H), 4.35 (t, J = 8.9 Hz, 2H), 4.08 (m, 2H), 3.56 (dd, J= 11.0, 2.4 Hz, 1H), 3.31 (s, 4H), 2.85 (d, j= 4.3
Hz, 1H), 2.58 (d, J= 4.3 Hz, 1H), 2.55 (t, J= 6.3 Hz, 1H), 2.17 (t, J= 5.5 Hz, 2H), 1.97 (m, 1H), 1.86 (m, 1H), 1.79 (d, = 11.0 Hz, 1H), 1.70 (s, 3H), 1.61 (s, 3H), 1.09 (s, 3H), 1.03 (d, J = 13.3 Hz, 1H). Examples 65(a) and 65(b): Preparation of sodium 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)azetidin-3-yl)propanoate
Figure imgf000200_0002
1. Preparation of tert-butyl 3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3- oxopropyl)azetidine-l-carboxylate
Figure imgf000200_0003
[0397] To a solution of (Z)-3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3-oxoprop-l-en-l- yl)azetidine-l-carboxylate (500 mg) in MeOH (20 mL) was added Pd/C and the mixture was stirred at r.t. under H2 overnight. TLC showed the reaction was complete. The reaction mixture was filtered and concentrated, and the concentrate was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give tert-butyl 3-(2-((tert-butyldimethylsilyl)oxy)-3- ethoxy-3-oxopropyl)azetidine-l-carboxylate (400 mg) as a colorless oil. 1H NMR (400 MHz, Chloroform-ύ d 4.21 - 4.12 (m, 3H), 4.01 (td, 7= 8.4, 5.2 Hz, 2H), 3.60 (ddd, J= 14.0, 8.6,
5.7 Hz, 2H), 2.68 (ddt, j= 10.1, 7.9, 4.0 Hz, 1H), 2.00 (dd, j= 7.5, 6.1 Hz, 2H), 1.43 (s, 9H), 1.28 (t, J= 7.1 Hz, 3H), 0.89 (s, 9H), 0.04 (d, J= 12.8 Hz, 6H).
2. Preparation of tert-butyl 3-(3-ethoxy-2-hydroxy-3-oxopropyl)azetidine-l- carboxylate
Figure imgf000201_0001
[0398] A solution of tert-butyl 3-(2-((tert-butyldimethylsilyl)oxy)-3-ethoxy-3- oxopropyl)azetidine-l-carboxylate (400 mg) in DCM (20 mL) was treated with TBAF (1 M in THF, 4.0 mL) and stirred at r.t. for 4 hours. TLC showed the reaction was complete. The reaction was extracted with EA and washed with water. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 5 % ~ 10 % EA in PE) to give tert-butyl 3-(3-ethoxy-2-hydroxy-3- oxopropyl)azetidine-l-carboxylate (209 mg) as a colorless oil. 1H NMR (400 MHz,
Chloroform-d) d 4.18 (qd, J = 7.1, 2.7 Hz, 2H), 4.06 (dd, J = 8.1, 4.0 Hz, 1H), 3.97 (td, J = 8.4, 2.4 Hz, 2H), 3.55 (ddd, J = 16.0, 8.6, 5.7 Hz, 2H), 2.67 (ddt, J = 10.2, 7.9, 4.1 Hz, 1H), 2.03 (ddd, J = 13.9, 8.1, 4.1 Hz, 1H), 1.85 (ddd, J = 13.9, 8.1, 6.9 Hz, 2H), 1.36 (s, 9H), 1.25 (t, J = 7.1 Hz, 3H).
3. Preparation of lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2- hydroxypropanoate
Figure imgf000201_0002
[0399] To a solution of tert-butyl 3-(3-ethoxy-2-hydroxy-3-oxopropyl)azetidine-l- carboxylate (209 mg) in MeOH/H20(v/v, 3/1, 10 mL) was added LiOH H20 (22 mg) and the mixture was stirred at r.t for 6 hrs. TLC showed all the starting material consumed. The solvent was removed under vacuum to give lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2- hydroxypropanoate (200 mg) as a white solid. LC MS: m/z (M-Li) 244.
4. Preparation of 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoic acid
Figure imgf000202_0001
[0400] To lithium 3-(l-(tert-butoxycarbonyl)azetidin-3-yl)-2-hydroxypropanoate (200 mg) in DCM(2.5 mL) was added TFA (2.5 mL). The resulting mixture was stirred at room temperature overnight. TLC showed the reaction was complete. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling the reaction in an ice bath. To the resulting solution was added
(3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 282 mg), and the reaction was stirred at room temperature for 6 hrs. TLC showed the reaction was complete. The reaction mixture was quenched with aqueous NaHC03 (1 M) to pH = 6-7 and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) and SFC (IG - 40% MEOH + 0.1% NH3 H20) to give 2-hydroxy-3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoic acid (29 mg) as a white solid and 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoic acid 54 mg) as a white solid. LC MS: m/z (M+H)+ 454.
5. Preparation of sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-
2-oxopropanoate
Figure imgf000202_0002
[0401] To a solution of 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl- 3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)propanoic acid in MeCN (20 mL) was added aqueous NaOH (0.1 M, 0.9 eq, 0.57 mL/l.07 mL) at -20 °C. The solvent was removed via lyophilization to give sodium 2-hydroxy-3-(l- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate (29 mg) as white solid. LC MS: m/z (M-Na) 452. 1H NMR (400 MHz, DMSO-d6) d 5.28 (s, 2H), 5.20 (t, J= 7.7 Hz, 2H), 4.42 (d, j= 3.2 Hz, 2H), 3.91 (t, j= 8.4 Hz, 3H), 3.51 (dd, j= 11.0, 2.6 Hz, 2H), 3.28 (s, 5H), 2.85 (d, J= 4.4 Hz, 2H), 2.70 - 2.61 (m, 2H), 2.62 - 2.53 (m, 2H), 2.18 (s, 2H), 2.23 - 2.10 (m, 1H), 1.94 (s, 1H), 1.79 (dd, J = 26.6, 11.6 Hz, 2H), 1.71 (s, 5H), 1.61 (s, 6H), 1.08 (s, 5H), 1.01 (d, J = 13.5 Hz, 2H). and sodium 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoate (55 mg) as white solid. LC MS: m/z (M-Na) 452. 1H NMR (400 MHz, DMSO-d6) d 5.28 (s, 1H), 5.20 (t, J= 7.6 Hz, 1H), 4.40 (s, 1H), 3.60 - 3.47 (m, 2H), 3.28 (s, 3H), 2.85 (d, J= 4.4 Hz, 1H), 2.71 - 2.60 (m, 1H), 2.60 - 2.53 (m, 2H), 2.23 - 2.13 (m, 2H), 1.99 - 1.88 (m, 1H), 1.75 (d, j= 10.8 Hz, 1H), 1.71 (s, 3H), 1.61 (s, 3H), 1.08 (s, 3H), 1.01 (d, J= 13.7 Hz, 1H).
Example 66: Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-2H-l,2,3-triazole-4-carboxylate
Figure imgf000203_0001
1. Preparation of methyl 2-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-2H-l,2,3- triazole-4-carboxylate
Figure imgf000203_0002
[0402] To a solution of tert-butyl 3-(hydroxymethyl)azetidine-l-carboxylate (936 mg), methyl lH-l,2,3-triazole-4-carboxylate (763 mg) and PPh3 (2.62 g) in DCM (10 mL) was added diisopropyl azodi carboxyl ate (1.2 g) very slowly at 0 °C. The reaction was then stirred at r.t which monitored by TLC. The reaction mixture was quenched with water and extracted with DCM (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give methyl 2-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-2H- l,2,3-triazole-4-carboxylate (mg) as a white solid. LC MS: m/z (M+l)+ 297. 1H MR (400 MHz, Chloroform -ί/) d 8.13 (s, 1H), 4.93 (d, J= 7.6 Hz, 2H), 4.03 (dd, J= 9.0, 8.2 Hz, 2H), 3.94 (s, 3H), 3.80 (dd, j= 9.0, 5.2 Hz, 2H), 3.17 (tt, j= 7.9, 5.2 Hz, 1H), 1.43 (s, 9H).
2. Preparation of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)-2H-l,2,3-triazole-4-carboxylic acid
Figure imgf000204_0001
[0403] To a solution of methyl 2-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-2H-l,2,3- triazole-4-carboxylate (280 mg) in MeOH/H20(v/v, 3: 1, 10 mL) was added Li0H ¾0 (40 mg). The mixture was stirred at r.t for 6 hrs. TLC showed the reaction was complete. The solvent was then removed under vacuum to give lithium 2-((l-(tert-butoxycarbonyl)azetidin-3- yl)methyl)-2H-l,2,3-triazole-4-carboxylate (290 mg) as white solid. LC MS: m/z (M-l) 281. The crude product was dissolved in DCM (2.5 mL), and TFA (2.5 mL) was added. The resulting mixture was stirred at room temperature overnight. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling the reaction in an ice bath. To the resulting solution was added (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 282 mg), and the reaction was stirred at room temperature for 6 hrs. The reaction mixture was quenched with aqueous NaHC03 (1 M) and extracted with EA (3 c 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give 2-((l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-2H-l,2,3-triazole-4-carboxylic acid (88 mg) as a white solid LC MS: m/z (M+l)+ 491.
3. Preparation of sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)-2H-l,2,3-triazole-4-carboxylate
Figure imgf000205_0001
[0404] To a solution of 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)-2H-l,2,3-triazole-4-carboxylic acid (88 mg) in MeCN (20 mL) was added aqueous NaOH (0.1 M, 0.9 eq, 0.57 mL) at -20 °C. The solvent was removed by lyophilization to give sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)-2H-l,2,3- triazole-4-carboxylate (88 mg) as a white solid. LC MS: m/z (M-Na) 489. 1H MR (400 MHz, DMSO-d6) d 7.59 (s, 1H), 5.29 (s, 1H), 5.18 (t, J= 7.6 Hz, 1H), 4.93 (s, 2H), 3.86-4.00 (m, 2H), 3.74 (s, 2H), 3.51 (d, = 10.8 Hz, 1H), 3.27 (s, 3H), 3.14 - 3.01 (m, 2H), 2.84 (d, 7= 4.4 Hz, 1H), 2.57 - 2.58 (m, 1H), 2.55 (d, J= 4.2 Hz, 1H), 2.08-2.23 (m, 2H), 1.99 - 1.87 (m, 3H), 1.65-1.80 (m, 3H), 1.70 (s, 3H), 1.60 (s, 3H), 1.07 (s, 3H), 1.00 (d, J= 13.9 Hz, 1H).
Example 67: Preparation of sodium 6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-6, 7-dihydro-
5H-pyrrolo [3,4-b] pyridine-3-carboxylate
Figure imgf000205_0002
1. Preparation of dimethyl 5-bromopyridine-2,3-dicarboxylate
Figure imgf000206_0001
[0405] To a solution of 5-bromopyridine-2,3-dicarboxylic acid (5 g, 20.3 mmol) in MeOH (50 mL) was added concentrated H2S04 (2 mL). Then the mixture was heated at reflux for 20 h, and TLC showed the reaction was complete. After cooling to r.t., the reaction was quenched with saturated aqueous NaHC03 and concentration in vacuo. The residue was diluted with water (50 mL), and then extracted with DCM (50 mL x 3). The combined organic layers were dried over anhydrous Na2S04, filtered, and concentrated in vacuo to give the title compound as a white solid (4.3 g).LC-MS (ESI) found: 275 [M+H]+.
2. Preparation of (5-bromopyridine-2,3-diyl)dimethanol
Figure imgf000206_0002
[0406] NaBLL (1.4 g, 36.5 mmol) was added portion-wise to a solution of 5-bromopyridine- 2,3-dicarboxylate (2 g,7.3 mmol) in EtOH (20 mL, pre-cooled to 0 °C). A solution of CaCl2 (18.2 mL,2L9 mmol) in ethanol was added slowly at 0 °C, and the reaction mixture was warmed to r.t. and stirred overnight. TLC showed the reaction was complete. Excess sodium borohydride was quenched by slow addition of aqueous 2 N HC1 solution (3 mL), followed by stirring at r.t. for 0.5 h. Saturated aqueous sodium bicarbonate solution was added until a pH of ~ 7 was reached. The mixture was concentrated in vacuo, and the residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 10/1) to give (5-bromopyridine-2,3- diyl)dimethanol (lg). LC-MS (ESI) found: 219 [M+l]+.
3. Preparation of 5-bromo-2,3-bis(chloromethyl)pyridine
Figure imgf000206_0003
[0407] To a solution of (5-bromopyridine-2,3-diyl)dimethanol (900 mg, 4.13 mmol) in DCM (20 mL) was added SOCl2 (15 mL) dropwise at 0 °C. After 3 h, TLC showed the reaction was complete. The reaction was concentrated and the residue was used in the next step directly. LC-MS (ESI) found: 256 [M+l]+.
4. Preparation of 3-bromo-6-(3,4-dimethylbenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridine
Figure imgf000207_0001
[0408] To a solution of 5-bromo-2,3-bis(chloromethyl)pyridine (900 mg, 3.53 mmol) in DCM (15 mL) was added DIPEA (3.5 mL). Then (2,4-dimethoxyphenyl)methanamine (0.64 mL, 4.24 mmol) was added. And the mixture was stirred at r.t. overnight. TLC showed the reaction was complete. The reaction was concentrated under vacuum. The residue was purified by a silica gel column chromatography (PE/EA (v/v) = 5/1) to give 3-bromo-6-(3,4- dimethylbenzyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine (610 mg). LC-MS (ESI) found:350 [M+l]+.
5. Preparation of methyl 6-(3,4-dimethylbenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b] pyridine-3-carboxylate
Figure imgf000207_0002
[0409] To a solution of 3-bromo-6-(3,4-dimethylbenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridine (610 mg, 1.75 mmol) in MeOH (5.5 mL) and TEA (2 mL) was added Pd(dppf)Cl2 (100 mg). Then, the reaction was degassed with CO, and the resulting mixture was stirred at 80 °C overnight. TLC showed the reaction was complete. The reaction mixture was concentrated, and purified on a silica gel column eluting with PE/EA (2.5/1) to give the title product (130 mg). LC-MS (ESI) found:329 [M+l]+.
6. Preparation of lithium 6-(2,4-dimethoxybenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b] pyridine-3-carboxylate
Figure imgf000207_0003
[0410] To a solution of methyl 6-(3,4-dimethylbenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridine-3-carboxylate (127 mg, 0.39 mmol) in THF (4.5 mL) and H20 (1.5 mL) was added Li0H ¾0 (16.2 mg, 0.39 mmol). After stirring at r.t. overnight, TLC showed the reaction was complete. The solvent was removed and the crude product was used in the next step directly. LC-MS (ESI) found:3 l5 [M+l]+. 7. Preparation of 6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxylic acid
Figure imgf000208_0001
[0411] A solution of lithium 6-(2,4-dimethoxybenzyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridine-3-carboxylate (122 mg, 0.39 mmol) in TFA (5 mL) and DCM (1 mL) was stirred at 65 °C overnight. TLC showed the reaction was complete. The solvent was removed and the crude product was used in the next step directly. LC-MS (ESI) found: 165 [M+l]+.
8. Preparation of sodium 6-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-6,7-dihydro- 5H-pyrrolo [3,4-b] pyridine-3-carboxylate
Figure imgf000208_0002
[0412] To a solution of 6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxylic acid (64 mg, 0.39 mmol) in MeCN (3 mL) was added DIPEA (0.27 mL, 1.56 mmol) at 0 °C. After 30 min, (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1 139 mg, 0.31 mmol) was added in one portion and the reaction was warmed to r.t. under N2. After 2 hrs, TLC showed the reaction was complete. The reaction was concentrated under vacuum to remove the solvent while keeping the temperature under 50 °C. The ammonium salt product (75 mg) was obtained through the prep-HPLC (Cl 8, 0-90% MeCN in H20 with 0.1% NH3 H20) as a white solid. To a solution of ammonium salt product (75 mg) in MeCN (2mL) was added 0.1 N NaOH solution (1.7 mL) dropwise at -60°C with stirring, followed by lyophilization to give sodium 6- ((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxylate (70 mg) as a white solid. LC-MS (ESI) found: 473 [M+l]+. 1H NMR (400 MHz, DMSO-d6) 5:8.92 (s, 1H), 8.15-8.20 (m, 1H), 5.46 (s, 1H), 5.20 (t, J = 7.2 Hz, 1H), 4.55-4.80 (m, 4H), 3.58 (dt, Ji = 10.8 Hz, J2 = 2.8 Hz, 1H), 3.32-3.33 (m, 3H), 2.90 (d, J = 4.4 Hz, 1 H), 2.57-2.65 (m, 2H), 2.03-2.21 (m, 3H), 1.75-1.93 (m, 3H), 1.71 (s, 3H), 1.62 (s, 3H), 0.98-1.12 (m, 4H). Example 68: Preparation of sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2, 3-dihydro- lH-pyrrolo [3,4-c] pyridine-6-carboxylate
Figure imgf000209_0001
1. Preparation of dimethyl 6-cyanopyridine-3,4-dicarboxylate
cCoOoOrMe COOMe
,COOMe Zn(CN)2, dppf, Pd2(dba)3 ^J^^COOMe
CV^ DMF, 100 °C, 3h
NC N
[0413] A mixture of dimethyl 6-chloropyridine-3,4-dicarboxylate (3.9 g, 17.0 mmol),
Zn(CN)2 (2.4 g, 20.4 mmol), Pd2(dba)3 (1.5 g, 1.7 mmol), and dppf (1.9 g, 3.4 mmol) in DMF (70 mL) was stirred for 4 h at 100 °C under nitrogen. TLC showed the reaction was complete. The residue was diluted with ethyl acetate, washed with brine, dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was purified by silica gel column eluting with petroleum ether/ethyl acetate (5: 1). The resulting dimethyl 6-cyanopyridine-3,4- dicarboxylate (2.4 g) was obtained as a light yellow solid. LC-MS (ESI) found:22l [M+l]+.
2. Preparation of 4,5-bis(hydroxymethyl)picolinonitrile
Figure imgf000209_0002
[0414] 2 M LiBH4 (10.4 mL) THF solution was added portion-wise to a solution of dimethyl
6-cyanopyridine-3,4-dicarboxylate (2.4 g, 10.4 mmol) in EtOH (40 mL, pre-cooled to 0°C).
The reaction mixture was warmed to r.t. and stirred for 1.5 h. TLC showed the reaction was complete. The mixture was then quenched by slow addition of 5 mL of water. The mixture was concentrated and then purified by silica gel column chromatography (DCM/MeOH (v/v) = 10/1) to give 4,5-bis(hydroxymethyl)picolinonitrile (1.45 g). LC-MS (ESI) found: 165 [M+l]+.
3. Preparation of 4,5-bis(chloromethyl)picolinonitrile
Figure imgf000210_0001
[0415] To a solution of 4,5-bis(hydroxymethyl)picolinonitrile (1.45 g, 8.83 mmol) in DCM (50 mL) was added SOCl2 (50 mL) dropwise at 0 °C. After 3 h, TLC showed the reaction was complete. The volatiles were removed, and the residue was used in the next step without purification. LC-MS (ESI) found: 202 [M+l]+.
4. Preparation of 2-(3,4-dimethylbenzyl)-2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6- carbonitrile
Figure imgf000210_0002
[0416] To a solution of 4,5-bis(chloromethyl)picolinonitrile (1.8 g, 8.8 mmol) in DCM (50 mL) was added DIPEA (7.7 mL, 44 mmol) and then(2,4-dimethoxyphenyl)methanamine (1.6 mL, 10.6 mmol) was added. And the mixture was stirred overnight at r.t. TLC showed the reaction was complete. The reaction was concentrated under vacuum. The residue was purified by silica gel column chromatography (PE/EA (v/v) = 2/1) to give 2-(3,4-dimethylbenzyl)-2,3- dihydro-lH-pyrrolo[3,4-c]pyridine-6-carbonitrile (1.5 g). LC-MS (ESI) found: 296 [M+l]+.
5. Preparation of 2-(3,4-dimethylbenzyl)-2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6- carboxylic acid
Figure imgf000210_0003
[0417] 2 M NaOH (20 mL) solution was added to 2-(3,4-dimethylbenzyl)-2,3-dihydro-lH- pyrrolo[3,4-c]pyridine-6-carbonitrile (550 mg, 1.86 mmol). Then, the mixture was heated to reflux for 18 hours. TLC showed the reaction was complete. The mixture was cooled to 0 °C and acidified to pH 5-6 with concentrated HC1. Then the mixture was concentrated in vacuo to give crude 2-(3,4-dimethylbenzyl)-2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6-carboxylic acid (619 mg). LC-MS (ESI) found:3 l5 [M+l]+.
6. Preparation of 2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6-carboxylic acid
Figure imgf000211_0001
[0418] A a solution of 2-(3,4-dimethylbenzyl)-2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6- carboxylic acid (619 mg, 1.97 mmol) in TFA (5 mL) and DCM (2 mL) was stirred at 65 °C overnight. TLC showed the reaction was complete. The solvent was removed and the residue was used in the next step directly. LC-MS (ESI) found: 165 [M+l]+.
7. Preparation of sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2, 3-dihydro- lH-pyrrolo [3,4-c] pyridine-6-carboxylate
Figure imgf000211_0002
[0419] To a solution of 2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6-carboxylic acid (320 mg, 1.95 mmol) in MeCN (10 mL) was added DIPEA (1.36 mL, 7.80 mmol) at 0 °C. After 30 min, (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl (4-nitrophenyl) carbonate (Intermediate 1, 436 mg, 0.97 mmol) was added, and the reaction was warmed to r.t. under N2. After 2 h, TLC showed the reaction was complete. The reaction was concentrated under vacuum to remove the solvent while keeping the temperature under 50 °C. The ammonium salt product (68 mg) was obtained by prep-HPLC (Cl 8, 0-90% MeCN in H20 with 0.1% NH3Ή20) as a white solid. Then to the solution of ammonium salt product (68 mg) in MeCN (2 mL) was added 0.1 N NaOH solution (l .6mL) dropwise at -60°C with stirring, followed by lyophilization to give sodium 2-((((3R,4S,5S,6R)-
5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-
6-yl)oxy)carbonyl)-2,3-dihydro-lH-pyrrolo[3,4-c]pyridine-6-carboxylate (71 mg) as a white solid. LC-MS (ESI) found: 473 [M+l]+. 1H NMR (400 MHz, DMSO-d6) 5:8.47-8.50 (m, 1H), 7.95-7.99 (m, 1H), 5.52 (s, 1H), 5.27 (t, J = 7.2 Hz, 1H), 4.66-4.83 (m, 4H), 3.65 (dt, Ji = 10.8 Hz, J2 = 2.8 Hz, 1H), 3.38 (s, 3H), 2.95 (d, J = 4.4 Hz, 1 H), 2.64-2.70 (m, 2H), 2.11-2.27 (m, 3H), 1.82-1.98 (m, 3H), 1.78 (s, 3H), 1.68 (s, 3H), 1.16 (s, 3H), 1.10 (d, J = 13.6 Hz, 1H). Example 69: Preparation of sodium l-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylate
C1JSO4
Sodium ascorbate
rnnivi
Figure imgf000212_0003
1. Preparation of tert-butyl 3-(azidomethyl)azetidine-l-carboxylate
Figure imgf000212_0001
[0420] To a solution of tert-butyl 3-(iodomethyl)azetidine-l-carboxylate, 900 mg) in DMF was added NaN3 (295 mg, 1.5 eq). The reaction mixture was stirred at r.t. and monitored by TLC. After the reaction completed, the reaction was quenched with water and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 10 % ~ 50 % EA in PE) to give tert-butyl 3-(azidomethyl)azetidine-l-carboxylate (500 mg).
2. Preparation of methyl l-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-lH-l,2,3- triazole-4-carboxylate
CuS04
Sodium ascorbate
Figure imgf000212_0002
[0421] /ert-Butyl 3-(azidomethyl)azetidine-l-carboxylate (500 mg) and methyl propiolate were suspended in a 1 :1 mixture of water and tert-butyl alcohol (6 mL/6mL). Sodium ascorbate (0.3 mmol, 300 pL of freshly prepared 1 M solution in water) was added, followed by copper(II) sulfate pentahydrate (7.5 mg, 0.03 mmol, in 100 pL of water). The heterogeneous mixture was stirred vigorously overnight, at which point the mixture had become clear, and TLC indicated complete consumption of the reactants. The reaction mixture was diluted with water (50 mL), cooled in ice, and the white precipitate was collected by filtration. After washing the precipitate with cold water (2x25 mL), it was dried under vacuum to afford methyl l-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylate (500 mg, 94%) as a white solid LC MS: m/z (M+l)+ 297. 1H NMR (400 MHz, Chloroform -if) d 8.04 (s, 1H), 4.56 (d, J= 7.6 Hz, 2H), 4.01 (t, J= 8.5 Hz, 2H), 3.89 (s, 3H), 3.68 (dd, J= 9.0, 5.0 Hz, 2H), 3.06 (ddd, j= 10.5, 7.9, 5.0 Hz, 1H), 1.37 (s, 9H).
3. Preparation of sodium l-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)-lH-l,2,3-triazole-4-carboxylate
1. TFA, DCM MeCN te 1
Figure imgf000213_0001
[0422] To a solution of methyl l-((l-(tert-butoxycarbonyl)azetidin-3-yl)methyl)-lH-l,2,3- triazole-4-carboxylate (500 mg) in MeOH/H20 (v/v, 3 : 1, 10 mL) was added LiOHH20 (40 mg). The mixture was stirred at r.t for 6 hrs. Then the solvent was removed under vacuum to give lithium 1 -(( 1 -(tert-butoxycarbonyl)azeti din-3 -yl )m ethyl)- 1H- 1 ,2,3 -triazole-4-carboxylate (290 mg) as a white solid without purification. LC MS: m/z (M-l) 281. The residue was dissolved in DCM (2.5 mL), then TFA (2.5 mL) was added. The resulting mixture was stirred at room temperature overnight. The solvent was removed under vacuum. The residue was dissolved in MeCN (10 mL), and DIPEA (0.5 mL) was added dropwise while cooling the reaction in an ice bath. To the resulting solution was added (3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl (4- nitrophenyl) carbonate (Intermediate 1, 282 mg), and the reaction was stirred at room temperature for 6 hrs. The reaction mixture was quenched with aqueous NaHC03 (1 M) and extracted with EA (3 x 20 mL). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography (silica gel, 1 % ~ 10 % MeOH in DCM) to give l-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylic acid (110 mg) as a white solid. LC MS: m/z (M+l)+ 491. To a solution of l-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R, 3R)-2-methyl -3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylic acid (110 mg) in MeCN (20 mL) was added aqueous NaOH (0.1 M) at -20 °C. The solvent was removed after lyophilization to give impure product (115 mg, analyzed by 1HNMR). The product was re- purified with prep-HPLC (Cl8,0 ~ 90 % acetonitrile in H20 with 0.1 % NH3H20) and SFC (AD column-20%EtOH+0.l%NH3H2O) to give ammonium l-((l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylate (30 mg) . To this product was added aqueous NaOH (0.1 M) at -20 °C, and the solvent was removed after lyophilization to give sodium l-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)methyl)-lH-l,2,3-triazole-4-carboxylate (29 mg) as a white solid LC MS: m/z (M-Na) 489. 1H NMR (400 MHz, DMSO-i¾) d 8.02 (d, J= 8.8 Hz, 1H), 5.29 (d, J= 3.7 Hz, 1H), 5.24 - 5.05 (m, 1H), 4.55 (d, J= 7.5 Hz, 2H), 3.91 (dt, J= 20.8, 8.4 Hz, 3H), 3.72 (s, 2H), 3.57 - 3.48 (m, 2H), 3.27 (s, 3H), 3.14 - 3.01 (m, 2H), 2.84 (d, J= 4.4 Hz, 1H), 2.55 (d, J= 4.0 Hz, 2H), 2.16 (q, j= 7.6 Hz, 2H), 1.94 (td, j= 13.4, 4.7 Hz, 1H), l.70(s, 3H), l.65-l.79(m, 3H), 1.60 (s, 3H), 1.07 (s, 3H), 0.99 (d, j= 13.8 Hz, 1H).
Example 70: Preparation of sodium 3-fluoro-l-((((.?/?, S,5»S,6/?)-5-methoxy-4-((2/?,.?/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidine-3-carboxylate
Figure imgf000214_0001
1. Preparation of 3-fluoro-l-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidine-3- carboxylic acid
Figure imgf000214_0002
[0423] To a solution of 3-fluoropyrrolidine-3-carboxylicacid (200 mg, 1.5 mmol) in MeCN (5 mL) was added DIPEA (0.5 mL, 3.1 mmol) dropwise at 0 °C. The mixture was stirred at 0 °C for 10 min, then (5f?,4,S,,5S,,6/?)-5-methoxy-4-[(2f?,5f?)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenylcarbonate (Intermediate 1 806 mg, 1.8 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40 °C. The residue was purified by column chromatography (silica gel, 1 % ~ 10% MeOH in DCM) and prep- HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 H20) to give 3-fluoro- 1 -( { [(3/(-/N5.5', 6//)-5-rnethoxy-4- [(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)pyrrolidine-3-carboxylic acid (210 mg) as a colorless oil. LC-MS (ESI) m/z (M+l): 442.
2. Preparation of sodium 3-fluoro- l-((((J/?, V,5,V,6/?)-5-methoxy-4-((2/?,J/?)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)pyrrolidine-3-carboxylate
Figure imgf000215_0001
[0424] To a mixture of 3-fluoro-l-({[(3f?,4ri,,5ri,, 6/?)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)pyrrolidine-3- carboxylic acid (160 mg, 0.36 mmol) in MeCN (1 mL) was added aq. 0.1 M NaOH (3.2 mL) at -60 °C with stirring. The reaction mixture was lyophilized to give l-(3R,4S,5S, 6R)-5- methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl 3 -sodium 3-fluoropyrrolidine-l,3-dicarboxylate (130 mg) as a white solid. LC-MS (ESI) m/z (M+l): 442. 1H MR (400 MHz, DMSO-d6) d 5.36 (s, 1H), 5.19 (t, J= 7.0 Hz, 1H), 3.77 - 3.37 (m, 4H), 3.29 (s, 3H), 2.85 (d, J= 4.1 Hz, 1H), 2.57 (dd, 7= 11.1, 6.4 Hz, 2H), 2.42 - 2.08 (m, 3H), 1.98 (dd, 7 = 18.5, 10.1 Hz, 2H), 1.85 - 1.73 (m, 2H), 1.70 (s, 3H), 1.61 (s, 3H), 1.08 (d, 7= 3.7 Hz, 3H), 0.99 (d, J= 12.3 Hz, 1H). Example 71: Preparation of sodium 2-((((J ?, V,5»V,6 ?)-5-methoxy-4-((2 ?,J ?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-5-oxa-2- azaspiro [3.4] octane-6-carboxylate
Figure imgf000216_0001
1. Preparation of 5-oxa-2-azaspiro[3.4]octane-6-carboxylic acid
Figure imgf000216_0002
[0425] To a mixture of 2-[(tert-butoxy)carbonyl]-5-oxa-2-azaspiro[3.4]octane-6-carboxylic acid (250 mg, 0.97 mmol) in DCM (3 mL) was added TFA (1 mL). The reaction mixture was stirred at room temperature for 2 hrs. TLC showed the reaction was complete. The resulting mixture was concentrated to give crude product, which was used in the next step without further purification. LC-MS (ESI) m/z (M+l): 158.
2. Preparation of 2-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-5-oxa-2- azaspiro [3.4] octane-6-carboxylic acid
Figure imgf000216_0003
[0426] To a solution of 5-oxa-2-azaspiro[3.4]octane-6-carboxylic acid (150 mg, 0.95 mmol) in MeCN (5 mL) was added DIPEA (0.6 mL, 3.8 mmol) dropwise at 0 °C. The mixture was stirred at 0 °C for 10 min, then (3i?,4ri,,5ri,,6/?)-5-methoxy-4-[(2i?,3i?)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl 4-nitrophenyl carbonate (Intermediate 1, 426 mg, 0.95 mmol) was added. The reaction mixture was stirred at r.t. overnight under N2 atmosphere. TLC showed the reaction was complete. The mixture was concentrated under vacuum to remove the solvent while keeping the temperature below 40°C. The residue was purified by column chromatography (silica gel, 1 % ~ 10% MeOH in DCM) and prep-HPLC (Cl 8, 0 ~ 90 % acetonitrile in H20 with 0.1 % NH3 H20) and SFC (ChiralPak IG
250x21.2mm I.D., 5pm - 30% MeOH + 0.1% NH3 H20) to give 2-({[(3R,4S,5S, 6R)-5- methoxy-4-[((2R,3R) -2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6- yl]oxy}carbonyl)-5-oxa-2-azaspiro[3.4]octane-6-carboxylic acid as 2 separate diasteriomers A and B (102 mg and 70 mg) as colorless oil. LC-MS (ESI) m/z (M+l): 466.
3. Preparation of sodium 2-((((3/?, V,5,V,6/?)-5-methoxy-4-((2/?,3/?)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-5-oxa-2- azaspiro [3.4] octane-6-carboxylate (71)
Figure imgf000217_0001
[0427] To a mixture of 2-({[(3i?, S',5<S,,d/?)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)-5-oxa-2-azaspiro[3.4]octane- 6-carboxylic acid diastereomer A (102 mg, 0.22 mmol) in MeCN (1 mL) was added aq. 0.1 M NaOH (1.9 mL) at -60 °C with stirring. The reaction mixture was lyophilized to give 2- ( 3R , 4S, 5S, d ?)-5-methoxy-4-[(2R,3R)-2-methyl-3 -(3-methylbut-2-en- 1 -yl)oxiran-2-yl]-l - oxaspiro[2.5]octan-6-yl 6-sodium 5-oxa-2-azaspiro[3.4]octane-2,6-dicarboxylate (71) (94 mg) as a white solid. LC-MS (ESI) m/z (M+l): 466. 1H NMR (400 MHz, DMSO-d6) d 5.30 (s, 1H), 5.19 (t, J= 6.7 Hz, 1H), 4.16 - 4.09 (m, 1H), 4.02 (t, = 6.5 Hz, 1H), 3.86 (dd, J= 16.1, 8.8 Hz, 2H), 3.77 (t, J= 7.6 Hz, 1H), 3.52 (d, J= 10.8 Hz, 1H), 3.28 (s, 3H), 3.23 (d, J= 7.5 Hz, 1H), 2.84 (d, J= 4.3 Hz, 1H), 2.55 (dd, J= 13.0, 5.3 Hz, 2H), 2.17 (s, 2H), 2.07 - 1.88 (m, 4H), 1.76 (dd, J = 14.2, 9.7 Hz, 3H), 1.70 (s, 3H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, = 11.1 Hz, 1H). Example 72: Preparation of sodium 2-(((( /?, S,5A,6/?)-5-methoxy-4-( 2/?, /?)-2-methyl-3- (3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-5-oxa-2- azaspiro [3.4] octane-6-carboxylate
Figure imgf000217_0002
[0428] To a mixture of 2-({[(3f?, S,,5S,,d/?)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut- 2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)-5-oxa-2-azaspiro[3.4]octane- 6-carboxylic acid diastereomer B (75 mg, 0.16 mmol) in MeCN (1 mL) was added aq. 0.1 M NaOH (1.4 mL) at -60 °C with stirring. The reaction mixture was lyophilized to give 2- (3R , 4S, 5S, d/?)-5-methoxy-4-[(2R,3R)-2-methyl-3 -(3-methylbut-2-en- 1 -yl)oxiran-2-yl]-l - oxaspiro[2.5]octan-6-yl 6-sodium 5-oxa-2-azaspiro[3.4]octane-2,6-dicarboxylate (72) (65 mg) as a white solid. LC-MS (ESI) m/z (M+l): 466. 1H NMR (400 MHz, DMSO-d6) d 5.30 (s, 1H), 5.19 (t, 7= 6.7 Hz, 1H), 4.16 - 4.09 (m, 1H), 4.02 (t, 7= 6.5 Hz, 1H), 3.86 (dd, 7= 16.1, 8.8 Hz, 2H), 3.77 (t, 7= 7.6 Hz, 1H), 3.52 (d, 7= 10.8 Hz, 1H), 3.28 (s, 3H), 3.23 (d, 7= 7.5 Hz, 1H), 2.84 (d, 7= 4.3 Hz, 1H), 2.55 (dd, 7= 13.0, 5.3 Hz, 2H), 2.17 (s, 2H), 2.07 - 1.88 (m, 4H), 1.76 (dd, 7 = 14.2, 9.7 Hz, 3H), 1.70 (s, 3H), 1.61 (s, 3H), 1.07 (s, 3H), 1.01 (d, 7= 11.1 Hz, 1H).
Example 73: Preparation of sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)ethane-l-sulfonate
Figure imgf000218_0001
1. Preparation of tert- butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate
Figure imgf000218_0002
[0429] To a solution of 2-(l-(tert-butoxycarbonyl)azetidin-3-yl)acetic acid (6.5 g, 30.2 mmol) in THF (80 mL) was added 1 M B¾ THF solution (121 mL) at 0°C under N2. The mixture was stirred at r.t. overnight. Then, 100 mL of MeOH was added dropwise and the solution was refluxed for 2 h. TLC showed the reaction was complete. The reaction was concentrated and purified by column chromatography (petroleum ether/ethyl acetate, 1 : 1) on silica gel to provide tert- butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate (5 g) as a colorless oil. LC-MS (ESI) found: 202 [M+l]+.
2. Preparation of terf-butyl 3-(2-((methylsulfonyl)oxy)ethyl)azetidine-l-carboxylate
Figure imgf000218_0003
[0430] To a solution of /cvV-butyl 3-(2-hydroxyethyl)azetidine-l-carboxylate (1 g, 4.97 mmol) in DCM (10 mL) was added Et3N (2 mL) and MsCl (0.6 mL) at 0°C. The mixture was stirred for 4 h. TLC showed the reaction was complete. The reaction was concentrated and the residue was purified by column chromatography (petroleum ether/ethyl acetate, 1 :1) on silica gel to afford /cvV-butyl 3-(2-((methylsulfonyl)oxy)ethyl)azetidine-l-carboxylate (1 g) as a colorless oil. LC-MS (ESI) found: 280 [M+l]+.
3. Preparation of sodium 2-(l-(tert-butoxycarbonyl)azetidin-3-yl)ethane-l- sulfonate
Figure imgf000219_0001
[0431] To a solution of /cvV-butyl 3-(2-((methylsulfonyl)oxy)ethyl)azetidine-l-carboxylate (400 mg, 1.43 mmol) in EtOH (10 mL) was added Na2S03 (1.8 g, 14.3 mmol). The mixture was heated to reflux overnight. TLC showed the reaction was complete. The reaction was then cooled to r.t., filtered, and concentrated to provide 380 mg crude sodium 2-(l-(tert- butoxycarbonyl)azetidin-3-yl)ethane-l -sulfonate. LC-MS (ESI) found: 266 [M+l]+.
4. Preparation of 2-(azetidin-3-yl)ethane-l-sulfonic acid
Figure imgf000219_0002
[0432] To a solution of sodium 2-(l-(tert-butoxycarbonyl)azetidin-3-yl)ethane-l-sulfonate (380 mg, 1.43 mmol) in DCM (4 mL) was added TFA (1.5 mL) at 0 °C. Then, the reaction mixture was slowly warmed to r.t. and stirred for 1.5 h. TLC showed the reaction was complete. The reaction mixture was concentrated, and used directly in the next reaction. LC-MS (ESI) found: 166 [M+l]+.
5. Preparation of sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-
(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethane-l-sulfonate
Figure imgf000219_0003
[0433] To a solution of 2-(azeti din-3 -yl)ethane-l -sulfonic acid (237 mg, 1.43 mmol) in MeCN (5 mL) was added DIPEA (1.0 mL,5.74 mmol) at 0 °C. After 30 min, (3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl (4-nitrophenyl) carbonate (Intermediate 1, 321 mg, 0.72 mmol) was added in one portion and the reaction was warmed to r.t. under N2. After 2 h, TLC showed the reaction was complete. Then, the reaction was concentrated under vacuum to remove the solvent and purified by prep-HPLC (Method A, H20 (0.1% FA)/ CH3CN) to give product as ammonium salt (220 mg). Then to a solution of ammonium salt product (150 mg, 0.31 mmol) in MeCN (2 mL) was added 0.1 N NaOH solution (3.1 mL) dropwise at -60°C with stirring, followed by lyophilization to give sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)ethane- l-sulfonate (130 mg) as a white solid. LC-MS (ESI) found: 474 [M+l]+.1H NMR (400 MHz, DMSO-d6) 5:5.29 (s, 1H), 5.19 (t, J = 7.6 Hz, 1H), 3.91 (t, J = 8.0 Hz, 2H), 3.46-3.53 (m, 3H), 3.28 (s, 3H), 2.83 (d, J = 4.0 Hz, 1H), 2.55-2.64 (m, 3H), 2.35 (t, J = 7.6 Hz, 2H), 2.10-2.22 (m, 2H), 1.74-2.03 (m, 6H), 1.71 (s, 3H), 1.61 (s, 3H), 1.08 (s, 3H), 1.01 (d, J = 13.6 Hz, 1H).
Biological Examples:
Methionine Aminopeptidase 2 (MetAP2) Inhibition
[0434] Enzyme inhibition was assessed with Flag tagged Human recombinant MetAP2.
Assay buffer was prepared as a 2x stock containing 100 mM HEPES containing 200 mM NaCl, pH adjusted to 7.5, when required, assay buffer was created by 1 : 1 dilution with double distilled water. 10 mM stock solutions of compounds were prepared in 100% DMSO and further diluted in 100% DMSO required concentration to 1 mM stocks. The stock solutions and DMSO vehicle controls were diluted 1 :20 in assay buffer prior to addition to the test plate (final in well concentration 10 mM). MAS peptide was formulated to a 7.5 mM stock in distilled water and prior to use further diluted 1 :4. Amino acid oxidase was prepared as a stock solution (6.2 mg/ml) and prior to use further diluted 1 :49.6 in distilled water. A 250 pM solution of MnCl2 was prepared in advance of thawing an aliquot of MetAP2 enzyme. 40 pl of enzyme was mixed with 100 pl of MnCl2 then further diluted in assay buffer to a final concentration of 16 pg/ml. 5pl of test compound, lOpl of MAS substrate / amino acid oxidase mixture, 10 pl of MetAP2 was added to test wells in a 384 well black plate with blank wells containing no enzyme, replaced with 10 pl of assay buffer. All compounds we tested in duplicate on two occasions on the same day. The final in well concentrations of the assay are 1% DMSO, 0.272 pg/ml MetAP2, 10 pM MnCl2, 50.0 pg/ml (0.225 U/ml) amino acid oxidase, 0.75 mM MAS.
[0435] The plate was sealed with a TopSeal A cover and mixed briefly on an orbital mixer at 900 rpm. The plate was incubated for a further 25 minutes at 25 °C A 5x stock of Amplex buffer was prepared (0.25 M sodium phosphate, pH 7.4) and stored at 4 °C. When preparing for use the stock was diluted with distilled water. Amplex Ultraread stock solution was prepared at 2.57 mg/ml in 100% DMSO and stored in 50 mΐ aliquots at -20 °C. 20m1 of 505 U/ml Horse radish peroxidase was diluted in 990 ml of Amplex buffer, 100 mΐ of this was combined with 50 mΐ of Amplex Ultrared in 4850 ml of lx Amplex buffer to generate sufficient detection reagent for a 384 well plate. 25 mΐ detection reagent was added to each well of the test plate, which was re-sealed and mixed briefly on an orbital shaker. The plate was transferred to an Envision Multi-label reader and RFU measured corresponding to excitation 531 nm and emission 595 nm. The raw data file was exported as a .csv file for further analysis. At the end of the MetAP2 incubation add 25 mΐ Amplex/HRP mixture per well and read plate on Perkin Elmer Envision.
Human Vein Umbilical Cell (HUVEC) Proliferation
[0436] HUVECs are cells derived from the endothelium of veins from the umbilical cord. EGM-2 cell culture medium was used to grow cells. Cells were passaged by seeding at 3 x 103 cells/mL on T175 flasks and allowed to reach approximately 80% confluence before used in the experiment. After overnight culture cells were placed in fresh EGM-2 complete medium containing the appropriate concentration of compound in 100 pL/well. After a short incubation (1 or 2 hours), cells were washed with DPBS and placed in fresh EGM-2 complete medium without compound. After 72 hours of culture, cell proliferation was evaluated with CyQuant cell proliferation reagent according to manufacturer’s specifications for 60 minutes in a 37 °C, 5% C02 incubator. Fluorescence intensity of samples was measured using a fluorescent microplate reader with excitation at 485 nm and emission detection at 530 nm. Fluorescence intensity values were exported from the plate reader and average fluorescence was calculated across samples. Inhibition of cell proliferation was calculated using GraphPad Prism 7
(GraphPad Software, Inc.).
Diet Induced Obese Mouse Experiments
[0437] Taconic Biosciences maintains an inventory of C57BL/6NTac male mice on 60% kcal high fat diet (D12492; Research Diets). Mice are put on the diet at 6 weeks of age. Male DIO C57B1/6 mice at -19-21 weeks of age (13 - 15 weeks on high fat diet) were received from Taconic approximately 2 weeks prior to study start. Animals were singly housed in normal cages and continued on a high fat chow (D 12492; Research Diets) from the day they were received and throughout the study. Test articles were provided by the Sponsor. Compounds were formulated into a 100% DMSO stock (at 9 mg/mL) prior to the start of the study. Daily aliquots were formulated (to the dosing vehicle of 10% DMSO in water) each morning on Days 1-10 prior to dosing. A daily aliquot for each group was removed from the freezer and thawed on Days 1-10 prior to dosing and made up to the desired concentration in 10% DMSO. Animals were randomized into study groups (N=4) based on body weights collected on Day -3. All animals were administered vehicle via oral gavage on Days -3 to -1. For all groups on the mornings of Days 1-10, test compounds or vehicle were administered via oral gavage. Animals were dosed, based on a body weight taken each morning. Starting on Day -3, individual animal body weights were collected daily. Body weights collected throughout the study will be compared to the pre-dose body weight measured on Day -1.
[0438] On Day 11, all mice were euthanized via C02 asphyxiation and had blood and tissues collected at approximately 24 hours after the Day 10 dose. Animals were sacrificed by C02 and tissues collected and snap frozen in liquid nitrogen. For the liver, 2 aliquots were generated (-200 mg each into 2 mL conical tube). Brain was collected, snap frozen, and placed in a 50 mL conical tube. All tissues were stored on dry ice/at -70 °C.
Tissue N-Terminal Thioredoxin in DIO Mouse
[0439] The tissue samples were homogenized 1 :3 (w/v) in T-PER. The homogenized samples were centrifuged at 10,000 rpm for 10 min. The supernatant (lysate) was transferred to another tube. The total protein concentration of the supernatant was measured by BCA assay. The samples were mixed with 1 mM CaCl2 in 100 mM Tris buffer, pH 8.0 and then reduced with TCEP at room temperature for 10 min. The samples were denatured at 100 °C for 10 min and cooled to room temperature. The samples were mixed with iodoacetamide (IAM) and alkylated at 300 rpm on the incubation shaker for 30 min in the dark. DTT was added to react with excess IAM at room temperature for 10 min. Samples were mixed (25: 1, protein/enzyme) with endoproteinase Glu-C solution and digested overnight (16 hr) at 37 °C. The reaction was quenched by adding 10% formic acid. After digestion, the samples were centrifuged at 10,000 rpm for 10 min. The supernatant (20 pL) was transferred to a new sample plate and mixed with 10 pL of 0.1% formic acid in 90/10 water/acetonitrile (v/v). IS spiking solution (3 pL) was added. The samples were mixed well by pipetting. The sample plate was cover with heating foil and submitted for LC -MS/MS analysis. The analytes and standards were measured by LC- MS/MS. Injections (25 pL) were made onto an Acquity EIPLC BEH 300 C18 column, using a Shimadzu SIL-30ACMP autosampler and Shimadzu LC-20AD pump. The total running time per sample was 15 min. The analytes were detected with an API Sciex TripleTOF 6600 mass spectrometer. The levels of the N-terminal peptide of thioredoxin (amino acid 1-6 MVKLIE, or THRX) was read off of a standard curve and reported as ng/mg total protein (tp) in tissues or as a ratio of the concentration related to total protein in multiple tissues.
[0440] The results are shown in Table 1 below. Table 1
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
BQL = Below Quantifiable Limit
INCORPORATION BY REFERENCE
[0441] All publications and patents mentioned herein, including those items listed below, are hereby incorporated by reference in their entirety for all purposes as if each individual publication or patent was specifically and individually incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.
EQUIVALENTS
[0442] While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification. The full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
[0443] Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term“about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention.
[0444] What is claimed is:

Claims

1. A pharmaceutically acceptable base salt of a compound represented by:
Figure imgf000226_0001
wherein:
L1 is -NRXR2;
R1 is H or Ci-3alkyl, and R2 is selected from the group consisting of: a 4-9 membered monocyclic heterocyclic ring, heteroaryl, -Ci-2alkylene-heteroaryl, phenyl, -Ci-2alkylene- phenyl and -Ci-6alkylene; wherein -Ci-6alkylene may be straight or branched and may optionally be substituted with one or more substituents each independently selected from Rp; or
R1 and R2 taken together with the nitrogen to which they are attached form a 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring; wherein the 4-9 membered monocyclic, fused bicyclic, or spirocyclic heterocyclic ring is optionally substituted with one or more substituents each independently selected from Rh;
L2 is selected from the group consisting of: Ci-4alkylene, -0-Ci-6alkylene, -NRa-Ci. 4alkylene, -S-Ci-4alkylene, - S(0)2-Ci-4alkylene, -S(0)-Ci-4alkylene, =CRa-, -Ci-2alkylene- heteroaryl, a 4-9 membered monocyclic heterocyclic ring, and a bond; wherein Ci-4alkylene and -0-Ci-6alkylene may be straight or branched and may optionally be substituted with one or more substituents each independently selected from Rp;
T is selected from the group consisting of: -C(0)0H, tetrazole, -S03H, isoxazol-3-ol, isothiazol-3-ol, l-alkyl-lH-pyrazol-3-ol, l,2,4-oxadiazol-5-ol, l,2,4-thiadiazol-5-ol, 1H- imidazole-2,5-dione, oxazolidine-2,4-dione-5-yl, thiazolidine-2,4-dione-5-yl, 1,2,4- oxadiazolidine-3,5-dione-2-yl, 4-hydroxy-l,5-dihydro-2H-pyrrol-2-one-l-yl, 6-hydroxy-4H- l,3-dioxin-4-one-2-yl, -C(0)NHS(0)2-Ci-3alkyl, -NH-S(0)2-Ci-3alkyl, -S(0)2-NH-Ci-3alkyl, and -P(0)(0H)(0Rb); wherein -C(0)NHS(0)2-Ci-3alkyl, -NH-S(0)2-Ci-3alkyl and -S(0)2-NH- Cl-3alkyl may optionally be substituted with one or more substituents each independently selected from Rp;
Rp is independently selected, for each occurrence, from the group consisting of fluorine, hydroxyl, cyano, and -C(0)NRaRb; or two Rp are taken together to form =0; Rh is independently selected, for each occurrence, from the group consisting of hydrogen, Ci-3alkyl, fluoro and hydroxyl; or two Rh are taken together to form =0; and
Ra and Rb are independently selected, for each occurrence, from the group consisting of hydrogen and Cl-3 alkyl.
2. The pharmaceutically acceptable base salt of claim 1, wherein L1 is selected from the group consisting of:
Figure imgf000227_0001
wherein independently for each occurrence p is 1 or 2, q is 1 or 2, r is 0, 1 or 2, Z is CH2 or O, and Y is N or CH.
3. The pharmaceutically acceptable base salt of claim 1 or 2, wherein L1 is selected from the group consisting of:
Figure imgf000227_0002
, ; p y .
5. The pharmaceutically acceptable base salt of any one of claims 1-4, wherein the base salt is an alkali metal salt.
6. The pharmaceutically acceptable base salt of any one of claims 1-5, wherein the alkali metal is selected from the group consisting of sodium, potassium, and lithium.
7. The pharmaceutically acceptable base salt of any one of claims 1-6, wherein T is selected from the group consisting of:
Figure imgf000228_0001
8. The pharmaceutically acceptable base salt of any one of claims 1-7, selected from the group consisting of: sodium 3-((S)-2-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)-3-methylbutyl)-5-oxo-l,2,4-oxadiazol-4-ide; sodium 2-(l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)acetate; sodium 1- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l -oxaspiro[2.5 ] octan-6-yl)oxy)carbonyl)piperi dine-4-carboxylate; sodium 5 -(( 1 - ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)tetrazol-2-ide; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)acetate; sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate;
sodium 5-(2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)tetrazol-2-ide; sodium (2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)acetyl)(methylsulfonyl)amide; sodium ethyl(2-(l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)ethyl)phosphonate; sodium 2- ((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-
2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)-2-methylpropanoate; sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2,2- dimethylpropanoate; sodium (3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-
3 -(3 -methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoyl)(methylsulfonyl)amide; sodium 5-((l-
((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)methyl)isoxazol-3-olate; sodium 5-(2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethyl)isoxazol-3-olate; sodium 3-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)oxy)propanoate; sodium 2-(((S)-l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate; sodium 2-((l-
((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)amino)acetate; sodium 2-
(((R)-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidin-3-yl)oxy)acetate; sodium 2-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-
1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin- 1 -yl)acetate; sodium 4-((((((3R,4S,5S,6R)-5-methoxy-4-((2S,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)benzoate;
sodium 4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)benzoate; sodium 6-
((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)nicotinate; sodium 1-
((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3-carboxylate; sodium 2-((R)-3-
(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate; sodium 2-
((S)-3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)pyrrolidin-l-yl)acetate; sodium 2-(4-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en- l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)piperidin-l-yl)acetate; sodium ethyl (l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)phosphonate; sodium 3 -(3 -(((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3 -(3 - methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)amino)azetidin-l-yl)propanoate; sodium 2-(4-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)piperazin-l-yl)acetate; sodium 2-(4- ((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)- 1 -oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)- 1H- 1,2,3 -triazol- 1 - yl)acetate; sodium 2-(4-((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)(methyl)amino)methyl)-lH-l,2,3-triazol-l-yl)acetate; sodium (3R,5R)- 3,5-dihydroxy-7-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2- en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)heptanoate; sodium 3-(3-(((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)azetidin-l-yl)-2,2- dimethylpropanoate; sodium (3R, 5R)-3 , 5 -dihydroxy-7 -(( 1 -((((3R,4S, 5 S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)oxy)heptanoate; sodium 2-(6- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,6-diazaspiro[3.3]heptan-2-yl)acetate; sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-oxopiperazin-l-yl)acetate; sodium 2-((2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-
1-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2-azaspiro[3.3]heptan-6- yl)oxy)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-3- methylazetidin-3-yl)oxy)acetate; sodium 2-(4-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-
2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)piperazin-l-yl)acetate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2 -yl)-l -oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-4-carboxylate; sodium 5- ((((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)amino)methyl)picolinate; sodium l-(l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-lH-pyrazole-3-carboxylate; sodium 6-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)nicotinate;
sodium 5-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)picolinate; sodium 6-(3-hydroxy-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)nicotinate; sodium 3-fluoro-l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidine-3-carboxylate; sodium 6-(3-fluoro-l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)nicotinate; sodium 5-(l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl) picolinate; sodium l-[l- ({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2- yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3-yl]-lH-imidazole-4-carboxylate; sodium 5-(3-fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)picolinate; sodium 2-(l-((((3R,4S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- ylidene)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)thio)acetate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)sulfonyl)acetate; sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en- 1 -yl)oxiran-2-yl)- 1 -oxaspiro[2.5 ] octan-6-yl)oxy)carbonyl)-2- azaspiro[3.3]heptane-6-carboxylate; sodium 2,2-difluoro-3-(l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)propanoate; sodium 2-((l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)sulfmyl)acetate; sodium l-(l- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en- l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxylate; sodium l-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)-2-oxo-l,2-dihydropyridine-4-carboxylate; sodium l-(l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-2-oxo-l,2-dihydropyridine-3- carboxylate; sodium 4-[l-({[(3R,4S,5S,6R)-5-methoxy-4-[(2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl]-l-oxaspiro[2.5]octan-6-yl]oxy}carbonyl)azetidin-3- yl]-l,3-thiazole-2-carboxylate; sodium 3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)-2-oxopropanoate; sodium l-(l-((((3R,4S,5S,6R)-5- methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l- oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3-yl)-4-oxo-l,4-dihydropyridine-3- carboxylate; sodium 2-hydroxy-3-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2- methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)propanoate; sodium 2-((l-((((3R,4S,5S,6R)-5-methoxy- 4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-2H-l,2,3-triazole-4-carboxylate; sodium 6- ((((3R,4S, 5 S,6R)-5-methoxy-4-((2R,3R)-2 -methyl -3-(3-methylbut-2-en-l-yl)oxiran-2- yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3- carboxylate; sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-2,3-dihydro- lH-pyrrolo[3,4-c]pyridine-6-carboxylate; sodium l-((l-((((3R,4S,5S,6R)-5-methoxy-4- ((2R,3R)-2-methyl-3-(3-methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6- yl)oxy)carbonyl)azetidin-3-yl)methyl)-lH-l,2,3-triazole-4-carboxylate; sodium 3- fluoro-l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)pyrrolidine-3-carboxylate; sodium 2-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-en-l- yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)-5-oxa-2-azaspiro[3.4]octane-6- carboxylate ; and sodium 2-(l-((((3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3- methylbut-2-en-l-yl)oxiran-2-yl)-l-oxaspiro[2.5]octan-6-yl)oxy)carbonyl)azetidin-3- yl)ethane-l -sulfonate, and a pharmaceutically acceptable salt or stereoisomer thereof.
9. A method of treating or mitigating a liver, intestine, and/or kidney disorder in a patient in need thereof, comprising administering an effective amount of a compound of any one of claims 1-8 to the patient.
10. The method of claim 9, wherein the liver disorder is one or more of: non-alcoholic steato- hepatitis, alcoholic steato-hepatitis, hepato-cellular carcinoma, liver cirrhosis, and hepatitis B.
11. The method of claim 9, wherein the kidney disorder is one or more of chronic kidney
disease, glomerular disease, IGA nephropathy, lupus nephritis, and polycystic kidney disease.
12. A method of treating hepatocellular carcinoma in a patient in need thereof, comprising adminsetering an effective amount of a compound of any one of claims 1-8 to the patient.
13. The method of any one of claims 9-11, wherein the patient suffers from type 2 diabetes, hepatitis B, cirrhosis, alpha-l antitrypsin deficiency, and/or is obese.
14. A method of treating, or preventing the development of, hepatic preneoplastic lesions in patient in need thereof, comprising administering an effective amount of a compound of any one of claims 1-8.
15. The method of claim 14, wherein the patient is at risk for but not yet suffering from NASH.
16. The method of claim 14, wherein the patient has at least one of: minimal liver fibrosis, minimal hepatic steatosis, lobular inflammation, and/or balloon degeneration.
17. The method of any one of claims 1-16, wherein the patient suffers from type 2 diabetes and/or is obese.
PCT/US2018/065225 2017-12-12 2018-12-12 Targeting compounds WO2019118612A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762597598P 2017-12-12 2017-12-12
US62/597,598 2017-12-12
US201862654710P 2018-04-09 2018-04-09
US62/654,710 2018-04-09

Publications (1)

Publication Number Publication Date
WO2019118612A1 true WO2019118612A1 (en) 2019-06-20

Family

ID=65139129

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/065225 WO2019118612A1 (en) 2017-12-12 2018-12-12 Targeting compounds

Country Status (1)

Country Link
WO (1) WO2019118612A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0357061A1 (en) * 1988-09-01 1990-03-07 Takeda Chemical Industries, Ltd. Angiogenesis inhibitory agent
WO2002042295A2 (en) * 2000-11-01 2002-05-30 Praecis Pharmaceuticals Inc. Peptides as met-ap2 inhibitors
WO2009073445A2 (en) * 2007-11-28 2009-06-11 Mersana Therapeutics, Inc. Biocompatible biodegradable fumagillin analog conjugates
WO2010065877A2 (en) * 2008-12-04 2010-06-10 Zafgen Corporation Methods of treating an overweight or obese subject
WO2011150088A1 (en) * 2010-05-25 2011-12-01 SynDevRX Optimized drug conjugates
WO2017027684A1 (en) * 2015-08-11 2017-02-16 Zafgen, Inc. Fumagillol heterocyclic compounds and methods of making and using same

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0357061A1 (en) * 1988-09-01 1990-03-07 Takeda Chemical Industries, Ltd. Angiogenesis inhibitory agent
WO2002042295A2 (en) * 2000-11-01 2002-05-30 Praecis Pharmaceuticals Inc. Peptides as met-ap2 inhibitors
WO2009073445A2 (en) * 2007-11-28 2009-06-11 Mersana Therapeutics, Inc. Biocompatible biodegradable fumagillin analog conjugates
WO2010065877A2 (en) * 2008-12-04 2010-06-10 Zafgen Corporation Methods of treating an overweight or obese subject
WO2011150088A1 (en) * 2010-05-25 2011-12-01 SynDevRX Optimized drug conjugates
WO2017027684A1 (en) * 2015-08-11 2017-02-16 Zafgen, Inc. Fumagillol heterocyclic compounds and methods of making and using same

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
CARREIRA; KVAERNO: "Classics in Stereoselective Synthesis", 2009, WILEY-VCH
CHRISTOPHER ARICO-MUENDEL ET AL: "Antiparasitic activities of novel, orally available fumagillin analogs", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 19, no. 17, 1 September 2009 (2009-09-01), AMSTERDAM, NL, pages 5128 - 5131, XP055555890, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2009.07.029 *
CHRISTOPHER C. ARICO-MUENDEL ET AL: "Carbamate Analogues of Fumagillin as Potent, Targeted Inhibitors of Methionine Aminopeptidase-2", JOURNAL OF MEDICINAL CHEMISTRY, vol. 52, no. 24, 24 December 2009 (2009-12-24), US, pages 8047 - 8056, XP055311910, ISSN: 0022-2623, DOI: 10.1021/jm901260k *
DONG Z. ET AL.: "A Substrate Pharmacophore for the Human Sodium Taurocholate Co-transporting Polypeptide", INT J PHARM., vol. 478, no. 1, 15 January 2015 (2015-01-15), pages 88 - 95
DORING, B. ET AL.: "The SLC10 carrier family: transport functions and molecular structure", CURR TOP MEMBR, vol. 70, 2012, pages 105 - 68
FENNER KS; JONES HM; ULLAH M; KEMPSHALL S; DICKINS M; LAI Y; MORGAN P; BARTON HA: "The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: from obscure liver transporters to key determinants of hepatobiliary clearance", XENOBIOTICA, vol. 42, no. 1, 11 November 2011 (2011-11-11), pages 28 - 45
GIACOMINI KM; HUANG SM; TWEEDIE DJ; BENET LZ; BROUWER KL; CHU X ET AL.: "Membrane transporters in drug development", NAT REV DRUG DISCOV., vol. 9, no. 3, 2010, pages 215 - 36
HAGENBUCH B.; MEIER PJ: "The superfamily of organic anion transporting polypeptides", BIOCHIM BIOPHYS ACTA, vol. 1609, no. 1, 2003, pages 1 - 18, XP004400940, DOI: doi:10.1016/S0005-2736(02)00633-8
KLEINER ET AL., HEPATOLOGY, vol. 41, 2005
KUMPULAINEN ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 7, 2008, pages 255
LETSCHERT, K. ET AL.: "Molecular characterization and inhibition of amanitin uptake into human hepatocytes", TOXICOL SCI, vol. 91, no. 1, 2006, pages 140 - 9
RIZWAN, AN.; BURCKHARDT, G: "Organic anion transporters of the SLC22 family: biopharmaceutical, physiological, and pathological roles", PHARM RES, vol. 24, no. 3, 2007, pages 450 - 70, XP019483495, DOI: doi:10.1007/s11095-006-9181-4
SAHI, J. ET AL.: "Metabolism and transporter-mediated drug-drug interactions of the endothelin-A receptor antagonist CI-1034", CHEM BIOL INTERACT, vol. 159, no. 2, 2006, pages 156 - 68, XP025058335, DOI: doi:10.1016/j.cbi.2005.11.001
SIMPLICIO ET AL., MOLECULES, vol. 13, 2008, pages 519
STIEGER, B. ET AL.: "In situ localization of the hepatocytic Na+/Taurocholate cotransporting polypeptide in rat liver", GASTROENTEROLOGY, vol. 107, no. 6, 1994, pages 1781 - 7
TU M; MATHIOWETZ AM; PFEFFERKORN JA; CAMERON KO; DOW RL; LITCHFIELD J; DI L; FENG B; LIRAS S: "Medicinal chemistry design principles for liver targeting through OATP transporters", CURR TOP MED CHEM., vol. 13, no. 7, 2013, pages 857 - 66

Similar Documents

Publication Publication Date Title
US11547704B2 (en) Chemical compounds
AU2021202973B2 (en) Isoindolinone inhibitors of the MDM2-p53 interaction having anticancer activity
JP2022078121A (en) Substitution type heterocyclyl derivative as cdk inhibitor
TWI434843B (en) Kinase inhibitors and methods of their use
JP6837482B2 (en) 2,4-Dihydroxy-nicotinamide as an APJ agonist
US10730863B2 (en) Bridged bicyclic compounds as farnesoid X receptor modulators
JP2021520350A (en) BRM Targeted Compounds and Related Usage
ES2716531T3 (en) 1-arylcarbonyl-4-oxy-piperidine compounds useful for the treatment of neurodegenerative diseases
EP3672949A1 (en) 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
CN107614499B (en) Pyrido [3,4-d ] pyrimidine derivatives and pharmaceutically acceptable salts thereof
JP2019532995A (en) Tau protein targeted PROTAC and related methods of use
EA019206B1 (en) Pyrazine compounds as phosphodiesterase 10 inhibitors
EP2225247A1 (en) 2-fluoropyrazolo[1,5-a]pyrimidines as protein kinase inhibitors
US20230265116A1 (en) Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use
US11365205B2 (en) Imidazolidin-2-one compounds as PRMT5 modulators
MX2014011373A (en) Diacylglycerol acyltransferase 2 inhibitors.
US10344017B2 (en) Pyrrolcarboxamide derivatives for the inhibition of ERK5
AU2014234907B2 (en) Geminally substituted cyanoethylpyrazolo pyridones as Janus kinase inhibitors
CA3140257C (en) Next-generation modulators of stimulator of interferon genes (sting)
KR20180035907A (en) PUMA GILROL HETEROCYCLIC COMPOUND AND METHODS OF MAKING AND USING THE SAME
TW202045498A (en) Substituted bicyclic compounds as farnesoid x receptor modulators
WO2019089665A1 (en) Alkene spirocyclic compounds as farnesoid x receptor modulators
JP2023529908A (en) BRM-Targeting Compounds and Related Methods of Use
CN103459382A (en) Heterocyclic compounds for the inhibition of pask
US20230097358A1 (en) Indazole based compounds and associated methods of use

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: 18836986

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 30/09/2020)

122 Ep: pct application non-entry in european phase

Ref document number: 18836986

Country of ref document: EP

Kind code of ref document: A1