US20190167604A1 - Arylfluorosulfate compounds and methods - Google Patents

Arylfluorosulfate compounds and methods Download PDF

Info

Publication number
US20190167604A1
US20190167604A1 US16/243,761 US201916243761A US2019167604A1 US 20190167604 A1 US20190167604 A1 US 20190167604A1 US 201916243761 A US201916243761 A US 201916243761A US 2019167604 A1 US2019167604 A1 US 2019167604A1
Authority
US
United States
Prior art keywords
compound
fluorosulfate
cancer
patient
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/243,761
Inventor
Jie Li
Zilei LIU
Suhua Li
Peng Wu
K. Barry Sharpless
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Scripps Research Institute
Original Assignee
Scripps Research Institute
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
Priority claimed from PCT/US2015/034516 external-priority patent/WO2015188120A1/en
Application filed by Scripps Research Institute filed Critical Scripps Research Institute
Priority to US16/243,761 priority Critical patent/US20190167604A1/en
Publication of US20190167604A1 publication Critical patent/US20190167604A1/en
Assigned to THE SCRIPPS RESEARCH INSTITUTE reassignment THE SCRIPPS RESEARCH INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WU, PENG, LI, JIE, LIU, Zilei, SHARPLESS, K. BARRY, LI, SUHUA
Priority to US16/871,902 priority patent/US11779548B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/136Amines having aromatic rings, e.g. ketamine, nortriptyline having the amino group directly attached to the aromatic ring, e.g. benzeneamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/14Quaternary ammonium compounds, e.g. edrophonium, choline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41681,3-Diazoles having a nitrogen attached in position 2, e.g. clonidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • A61K31/515Barbituric acids; Derivatives thereof, e.g. sodium pentobarbital
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/65Tetracyclines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/31Somatostatins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C305/00Esters of sulfuric acids
    • C07C305/26Halogenosulfates, i.e. monoesters of halogenosulfuric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/088Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/22Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with hetero atoms directly attached to ring nitrogen atoms
    • C07D295/26Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/02General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length in solution
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/113General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure
    • C07K1/1136General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure by reversible modification of the secondary, tertiary or quarternary structure, e.g. using denaturating or stabilising agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/06Methods of screening libraries by measuring effects on living organisms, tissues or cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B50/00Methods of creating libraries, e.g. combinatorial synthesis
    • C40B50/08Liquid phase synthesis, i.e. wherein all library building blocks are in liquid phase or in solution during library creation; Particular methods of cleavage from the liquid support
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • Described herein is a method of SuFEx chemistry for the conversion of phenolic compounds to their respective arylfluorosulfate derivatives in situ in 96-well plates.
  • This method is compatible with automated synthesis and screening to quickly assess the biological activities of the in situ generated crude products.
  • late-stage functionalization of a panel of known anti-cancer drugs generated the corresponding arylfluorosulfates.
  • These in situ generated arylfluorosulfates were directly tested in a cancer-cell growth inhibition assay in parallel with their phenolic precursors.
  • Three arylfluorosulfates exhibited improved anti-cancer cell proliferation activities compared to their phenol precursors (ABT-751, fulvestrant, and combretastatin A4).
  • the fluorosulfate derivative of fulvestrant showed significantly enhanced activity to down-regulate estrogen receptor (ER) expression in ER + breast cancer cell line MCF-7; and the fluorosulfate derivative of combretastatin A4 exhibited a 70-fold increase in potency in the drug resistant colon cancer cell line HT-29.
  • a fluorosulfate compound which comprises a fluorosulfate derivative of a phenolic medicinal agent, e.g., an anticancer agent, wherein at least one phenolic OH group of the medicinal agent is replaced by —OSO 2 F.
  • the medicinal agent is selected from the group consisting of ATB-751 (1), apigenin (2), ezetimibe (3), topotecan (4), ganetespib (5), etoposide (6), G100713 (7), estrone (8), PI-103 (9), raloxifene (10), fulvestrant (11), mycophenolic acid (12), gimeracil (13), flavopiridol (14), serotonin (15), PAC-1 (16), SKI II (17), endoxifen (18), vanillin (19), A-769662 (20), xanthohumol (21), sirtinol (22), IOX1 (23), TWS119 (24), combretastatin A4 (25), VER-50589 (26), luminespib (27), TW-37 (28), estrol (29), SB415286 (30), rotigotine (31), estradiol (32), RKI-1447 (33), licochal
  • a compound of Formula 1F is provided, which is a fluorosulfate derivative of ABT-751:
  • ABT-751 which is a useful antimitotic agent for treating certain cancers, e.g., certain breast cancer.
  • the parent, underivatized phenolic compound, ABT-751 reportedly binds to the colchicine site on beta-tubulin, which leads to a block in the cell cycle at the G2M phase, resulting in cellular apoptosis.
  • ABT-751 has general activity against a variety of cancers and solid tumors, for example.
  • a method of treating a cancer in a patient e.g., a human
  • administering an effective dose of a compound of Formula 1F to the patient.
  • a compound of Formula 11F is provided, which is a fluorosulfate derivative of fulvestrant:
  • Fulvestrant is indicated for the treatment of hormone receptor positive metastatic breast cancer in postmenopausal women with disease progression following antiestrogen therapy.
  • this drug has poor bioavailability and the only administration method is through intramuscular injection at a dose of 250 mg/month. It takes over 3 months to reach a steady serum concentration due to its quick clearance, significantly limiting its clinical value.
  • a method of treating a cancer in a patient comprising administering an effective dose of a compound of Formula 11F to the patient.
  • a compound of Formula 25F is provided, which is a fluorosulfate derivative of combretastatin A4:
  • Combretastatin A-4 is a tubulin-binding chemotherapy drug that is structurally related to colchicine, and has general anti-cancer activity. Also disclosed herein is a method of treating a cancer in a patient (e.g., a human), comprising administering an effective dose of a compound of Formula 25F to the patient.
  • the foregoing fluorosulfate (O-fluorosulfonyl) derivatives were prepared by contacting the phenol compound with a solution of SO 2 F 2 in acetonitrile at room temperature.
  • the method is adapted for use with a compound library in a high throughput screening format, such as a multi-well plate, e.g., 32-, 64- or 96-well plates, each well containing a compound comprising a phenolic hydroxyl group, for conversion to the corresponding fluorosulfonyl derivative.
  • This library of fluorosulfonylated compounds was then tested in various bioactivity evaluations.
  • the forgoing fluorosulfate compounds can be formulated into a pharmaceutical composition comprising the compound in a pharmaceutically acceptable carrier.
  • the fluorosulfate compounds can be utilized in a method of treatment of cancer in a patient (e.g., a human, or non-human animal), comprising administering an effective dose of the compound to the patient.
  • a patient e.g., a human, or non-human animal
  • the foregoing fluorosulfate compounds can be used for preparing a medicament for treating cancer.
  • FIG. 1 provides a comparison of phenolic drug functionalization workflows via the standard SuFEx and in situ SuFEx reactions.
  • FIG. 2 depicts development and optimization in situ SuFEx reaction and processing conditions.
  • A comparison of gas-liquid based SuFEx and liquid based SuFEx protocols for reactions in 96 well plates;
  • B removal of fluoride ions in situ after reaction: (1) volatiles in the reaction mixture were removed in vacuo, 19 F NMR showed the presence of anionic fluorine ( ⁇ 151 ppm, in CDCl 3 ); (2) after treating with trimethylsilanol (TMSOH), volatiles in the reaction mixture were removed in vacuo and 19 F NMR showed no presence of anionic fluorine or frimethylsilyl fluoride (TMSF).
  • TMSOH trimethylsilanol
  • FIG. 3 shows cytotoxicity profiling of the in situ generated fluorosulfate derivatives 1F to 39F using a double-dose cell-viability assay
  • DMSO dimethylsulfoxide
  • FIG. 4 demonstrates that fluorosulfate derivative 11F is a new selective estrogen receptor degrader or downregulator (SERD) that binds to and down-regulates Estrogen Receptor;
  • SESD selective estrogen receptor degrader or downregulator
  • A fluorosulfate derivative 11F specifically inhibits the proliferation of ER + MCF-7 cells;
  • compounds 11 and 11F induce ER down-regulation in MCF-7 cells.
  • FIG. 8 depicts fluorosulfate derivatives prepared by in situ reaction of phenolic anticancer compounds with SO 2 F 2 using the methods described herein.
  • FIG. 9 shows a comparison of the PK profile of 11F and fulvestrant (11) following single P.O. or S.C. administrations to female C57BL/6 mice.
  • Late-stage functionalization a strategy for directly introducing functional groups onto a bioactive compound in the late stage of its synthesis, enables rapid diversification of drug candidates or drug-like molecules to improve their properties such as potency and metabolic properties.
  • Many innovative methods have been developed for this endeavor, including late stage C—H functionalization and nucleophilic aromatic substitution, just to name a few.
  • LSF requires a chemical reaction with high selectivity, high yield and mild reaction conditions. Converting a phenolic compound with known biological activities to the corresponding fluorosulfate via SuFEx is an excellent transformation for LSF, which is waiting to be explored further. In fact, the phenolic hydroxyl group is often employed in drug modification and diversification.
  • Described herein is a protocol of SuFEx click chemistry for the LSF of phenol-containing drugs or drug candidates for converting the phenolic hydroxyl groups thereof to their respective arylfluorosulfate derivatives in situ in multi-well (e.g. 96-well) plates ( FIG. 1 ).
  • the in situ generated crude products of arylfluorosulfates were directly tested in a cancer-cell growth inhibition assay together with their phenolic precursors.
  • Three arylfluorosulfates exhibited improved anti-cancer proliferation activities compared to their phenol precursors.
  • the fluorosulfate derivative of combretastatin A4 exhibited a 70-fold increase in potency on the combretastatin-resistant colon cancer cell line HT-29.
  • a cost-effective protocol for converting a commercial screening library into a new library should possess the following characteristics: (1) compatible with small reaction scales (e.g., microgram); (2) excellent chemoselectivity; and (3) directly transferable to biological assays.
  • organic solvents e.g., dichloromethane (DCM) or acetonitrile
  • DIPEA N,N-Diisopropylethylamine
  • fluoride ions generated in this transformation cannot be removed by vacuo ( ⁇ 151 ppm in CDCl 3 ; FIG. 2 , B). Fluoride ions may have a synergistic or detrimental effect on arylfluorosulfates in the downstream biological assay. To minimize such influence on the screening results, excess trimethylsilanol (TMSOH, 20 equiv., boiling point 99° C. at 1 atm) was used to convert fluoride ions into volatile trimethylsilyl fluoride (TMSF, ⁇ 158 ppm in CDCl 3 ) whose boiling point is 16° C. at 1 atm.
  • TMSOH trimethylsilanol
  • FIG. 8 all possess anti-cancer activities.
  • R is —SO 2 F for the fluorosulfate derivatives
  • R is H for the parent phenolic drugs.
  • the phenol compounds are numbered as 1 to 39 and the corresponding fluorosulfate derivatives are designated 1F to 39F.
  • estimated yields for the in situ fluorosulfation reactions are provided after derivative compound numbers, where “quant” means quantitative yield. Notably, some of the compounds bearing multiple phenolic hydroxyl groups (fluorosulfate derivatives 5, 10, 27 and 37) were fully transformed into the corresponding fluorosulfation products (i.e., all phenolic OH groups reacted).
  • reaction of compound 28 only one of the three phenolic OH groups reacted (as determined by LC-MS), thus in compound 28F, two of the R groups are H. In the case of compound 2, only two of the three phenolic OH groups reacted. Due to the small quantities involved, and subsequent biological testing, the identities of the unreacted OH groups in 2F and 28F were not determined. Similarly, reaction of compound 26 resulted in a mixture of mono and di-fluorosulfation. Compounds 18 and 34 (as well as 18F and 34F), were mixtures of E/Z isomers.
  • the common names of the underivatized drugs used to form fluorosulfate derivatives 1F-39F are: 1 (ATB-751), 2 (apigenin), 3 (ezetimibe), 4 (topotecan), 5 (ganetespib), 6 (etoposide), 7 (G100713), 8 (estrone), 9 (PI-103), 10 (raloxifene), 11 (fulvestrant), 12 (mycophenolic acid), 13 (gimeracil), 14 (flavopiridol), 15 (serotonin), 16 (PAC-1), 17 (SKI II), 18 (endoxifen), 19 (vanillin), 20 (A-769662), 21 (xanthohumol), 22 (sirtinol), 23 (IOX1), 24 (TWS119), 25 (combretastatin A4), 26 (VER-50589), 27 (luminespib), 28 (TW-37), 29 (estrol), 30 (SB415286), 31 (rotigotine), 32
  • Additional FDA approved anticancer drugs that are suitable for derivitization with sulfonyl fluoride in the same manner include lasofoxifene, diethylstilbestrol, LE-SN38, amrubicin, genistein, and phenoxodiol. Fluorosulfate derivatives of such compounds also can be utilized in the treatment of the same cancers as the parent compounds.
  • the fluorosulfate compounds described herein can be formulated as a pharmaceutical composition in combination with a pharmaceutically acceptable carrier (also referred to as a vehicle or diluent).
  • a pharmaceutically acceptable carrier also referred to as a vehicle or diluent.
  • Such carriers are well known in the pharmaceutical arts, and include, e.g., 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.
  • the phrase “effective dose” 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 described herein are administered in therapeutically effective dose to treat a disease.
  • an effective dose of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect.
  • the cytotoxicity of fluorosulfation products 1F to 39F and their phenol precursors was assessed in cancer cell lines using a double-dose cell viability test.
  • the adenocarcinomic human alveolar basal epithelial cell A549 and the breast cancer cell MCF-7 were treated with vehicle (0.2% DMSO), fluorosulfation products or their phenol precursors in a final concentration of 20 ⁇ M or 500 nM for 72 h before the cytotoxicity of each compound was assessed according to the viability of cancer cells relative to the vehicle control.
  • the difference between the cytotoxicity of a fluorosulfation product and that of its phenol precursor reflects the changes in compounds' anti-cancer activity after the SuFEx-based derivatization ( FIG. 3 , A).
  • a fluorosulfation product exhibiting strong cytotoxicity (cell viability relative to the vehicle control less than 50% at both 20 ⁇ M and 500 nM on MCF-7 or A549 after a three-day treatment) and significantly increased (>10%) cytotoxicity over the parent compound is considered as a “hit”. Based on these two selection criteria, three hits 1F, 11F and 25F, were identified. As shown in FIG. 3 , B, fluorosulfate derivative 1F exhibited significantly enhanced activity relative to phenol precursor 1 at 500 nM (6% on MCF-7 and 27% on A549).
  • the cytotoxicity of fluorosulfate derivative 11F was stronger than that of phenol 11 on MCF-7 cell line with an increase of 32% and 20%, respectively, at 20 ⁇ M and 500 nM. However, no significant differences were found on A549 cells. Although phenol 25 and fluorosulfate derivative 25F showed similar activities on MCF-7 cells at 20 ⁇ M, fluorosulfate derivative 25F indeed exhibited 8% enhanced cytotoxicity at 500 nM, and its activities are significantly stronger than those of 25 at both concentrations on A549 cells. Notably, the corresponding phenol precursors 1, 11 and 25 are either FDA-approved drugs or are currently under clinical trials.
  • 1 (ABT-751), which inhibits polymerization of microtubules, is under a phasel/2 clinic rial; 11 (fulvestrant) is the only selective estrogen receptor down regulator (SERD) for the treatment of ER + metastatic breast cancer in postmenopausal women whose disease has spread after anti-estrogen therapy; and 25 (combretastatin A4) is a naturally occurring stilbenoid found in plants with activity in a wide range of cancers.
  • the corresponding phosphate of combretastatin A4 is a tumor vascular targeting agent, and the water-soluble prodrug of combretastatin A4.
  • arylfluorosulfate 1F and its phenol precursor 1 were evaluated against lung cancer cell line A549, breast cancer cell line MCF-7, SKBR3 and colon cancer cell line HT-29.
  • compound 1 showed IC 50 of 632 nM, 266 nM, 13.5 nM and 236 nM on A549, MCF-7, SKBR3 and HT-29 cells, respectively.
  • 1F exhibited 2 to 5-fold lower IC 50 values on these three cancer cell lines, i.e., 89 nM on A549, 134 nM on MCF-7, 6.9 nM on SKBR3 and 82 nM on HT-29.
  • 11F exhibited strong inhibition of the proliferation of ER + breast cancer cell lines MCF-7 and T47D, with IC 50 values of 5.5 nM and 4.8 nM, respectively, which are at the same level with those of 11 (7.7 nM and 14.8 nM, respectively) (Table 1).
  • both 11 and 11F have no activity on ER ⁇ MCF-7 and lung cancer A549 cells, suggesting 11F may still target estrogen receptors.
  • IC50 (nM) SKBR3 Compound A549Cells MCF-7 Cells Cells HT-29 Cells 1 (ABT-751) 632 266 13.5 236 1F 89 134 6.9 82 11 (fulvestrant) NI 7.7 NI 14.8 11F NI 5.5 NI 4.8 25 (combretastatin A4) 28 6.6 0.48 4947 25F 113 15.6 1.8 70 NI: No Inhibition (i.e., less than 50% inhibition at 10000 nM) Fluorosulfate 11F is a New SERD with Strong Potency.
  • fluorosulfate derivative 11F has stronger anti-cancer potency than the commercial drug 11, the cellular target of this new compound was investigated to determine whether it shares the same cellular target with compound 11.
  • fulvestrant (11) is the only SERD on the market for the treatment of ER + breast cancer. Fulvestrant competitively binds to estrogen receptor, which is a crucial regulator of ER + breast cancer growth, inhibiting dimerization of ER and leading to its degradation.
  • FIG. 4 A, fluorosulfate 11F inhibited ER + MCF-7 proliferation, but had no effect on ER ⁇ MCF-7 cells, suggesting that the anti-cancer activity of 11F is still ER dependent.
  • a competitive displacement ER binding assay in which the binding affinity of 11 and 11F to ER ⁇ protein was compared in competition with a fluoromone ligand was performed to confirm that ER is the target of fluorosulfate 11F.
  • MCF-7 cells were treated with 11 or 11F at concentrations of 3 nM, 9 nM, or 27 nM for 5 days. The ER ⁇ expression levels of the treated cells were then determined by Western blot.
  • fluorosulfate 11F showed surprisingly good oral bioavailability in vivo in C57BL/6 mice, whereas fulvestrant (11) is not effectively orally absorbed ( FIG. 9 ).
  • fluorosulfate 11F can be synthesized from 11 using a one-step quantitative “click” procedure, making it readily accessible for the future pharmacokinetics evaluation in animal models.
  • Fluorosulfate 25F Overcomes the Drug Resistance of HT-29.
  • Combretastatin A4 (25) is a one of the most potent anti-vascular agents that targets the colchicine-binding site of ⁇ -tubulin and hence disrupts tubulin polymerization.
  • HT-29 cells were treated with 25F followed by staining with anti-tubulin-FITC antibody.
  • Confocal imaging revealed that HT-29 cells treated with a vehicle (0.2% DMSO) or combretastatin A4 (25) at various concentrations clearly showed the presence of the microtubule network.
  • FIGS. 5, 6, and 7 show results of proliferation inhibitions studies in cancer cell lines.
  • the data in the figures show improved activity for the fluorosulfate derivatives relative to the underivatized drug in at least one cell line and dosage level.
  • converting a phenol group into an arylfluorosulfate is unlikely to change the protein targeting specificity of the parent compound, but instead this transformation confers the resulting arylfluorosulfate new properties which may enact in distinct mechanisms.
  • an arylfluorosulfate forms a covalent bond with the target protein via S VI -F exchange given the presence of tyrosine residues in the binding pocket and the presence of residues to facilitate the F departure.
  • a fluorosulfate-containing molecule binds to its target via non-covalent interactions stronger than that of the parent phenolic compound, which in some cases blocks the phase II metabolism pathway of the phenol moiety, i.e. sulfation, glucuronidation, and oxidation, which are key routes that change the pharmacokinetic properties of a phenolic compound.
  • LC-MS Liquid chromatography-mass spectrometry
  • AgilentTM 1260 LC/MSD with an AgilentTM 6120 quadrupole mass spectrometer (electrospray ionization, ES) eluting with 0.05% trifluoroacetic acid in H 2 O and 0.05% trifluoroacetic acid in CH 3 CN.
  • Precoated MerckTM F-254 silica gel plates were used for thin layer analytical chromography (TLC) and visualized with short wave (254 nm) UV light or by potassium permanganate stain.
  • Column chromatography was performed using SilicycleTM Silica Gel 60 (40-63 ⁇ m). All phenol compounds were purchased from Selleck chemicals. Sulfuryl fluoride (SO 2 F 2 ) gas was a gift from Dow AgroSciences. Phenolic compound library (10 mM in DMSO) was purchased from Selleck Chemicals.
  • a glass vial of organic solvent (5 mL) was evacuated in vacuo and a balloon containing SO 2 F 2 gas was connected with the glass vial, filling the vial with gas. Then the organic solvent was vigorously stirred for 30 min to make the stock solution of sulfuryl fluoride.
  • Triethylamine (TEA) (27.2 mg, 0.27 mmol) was added in a solution of compound 1 (10 mg, 0.027 mmol) in CH 3 CN (0.5 mL). After evacuation, SO 2 F 2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 12 hours until TLC (hexane/ethyl acetate, 1:1) showed the full conversion of starting material 1 to 1F (R f 0.70). Then volatiles were removed in vacuo and the resulting crude product was purified by flash column chromatography (hexane/ethyl acetate, 2:1 to 1:2) to obtain fluorosulfate 1F (11.3 mg, 0.025 mmol, 93%) as a light red solid.
  • Triethylamine (TEA) (41.5 mg, 0.41 mmol) was added in a solution of compound 11 (25 mg, 0.041 mmol) in CH 3 CN (0.5 mL). After evacuation, SO 2 F 2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 1 hours until TLC (hexane/ethyl acetate, 1:1) showed the full conversion of starting material 11 to 11F (R f 0.40).
  • Triethylamine (TEA) (79.9 mg, 0.79 mmol) was added in a solution of compound 25 (25 mg, 0.079 mmol) in CH 3 CN (0.5 mL). After evacuation, SO 2 F 2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 2 hours until TLC (hexane/ethyl acetate, 1:1) showed the formation of product 25F (R f 0.86).
  • All culture media are purchased from GIBCO. Charcoal-stripped FBS, PolarScreenTM ER ⁇ competitor assay kit (Green) and CellTiter-GloTM assay kit are purchased from Life Technologies.
  • Mouse monoclonal anti-tublin-FITC antibody (DM1A) is from AbCam Inc.
  • Rabbit monoclonal UGT1 antibody is from Cell Signaling Technology.
  • Rabbit monoclonal ER ⁇ antibody is from AbCam Inc.
  • Experimental data were processed by Prism 7TM and western blot images were analyzed by ImageJ 1.50iTM software.
  • the MCF-7, A549, SKBR3 and HT-29 cells were originally purchased from ATCC.
  • the T47D and ER ⁇ MCF-7 cells were kind grifts from Christopher K. Glass group (UCSD). All cancer cell lines were routinely maintained in DMEM (+Gluta MAXTM) supplement with 10% FBS and 1% peneciline/streptomycin in 5% carbon dioxide at a temperature of 37° C.
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAXTM) supplement with 5% FBS.
  • MCF-7 and A549 cancer cells were inoculated into 96-well plates in 100 ⁇ L medium at a plating density of 5,000 cells/well. After cell inoculation, the plates were incubated at 37° C. and 5% CO 2 . After 24 hours, all wells were refreshed with medium containing DMSO (0.2%, as vehicle control), phenols (20 ⁇ M or 500 nM) or in situ generated fluorosulfation products (20 ⁇ M or 500 nM). Then cancer cells were maintained at 37° C.
  • V control , V phenol and V fluorosulfation product were evaluated by CellTiter-GloTM Assay following manual of protocol.
  • Cell viability percentage relative to vehicle control is defined as V phenol /V control ⁇ 100% or V fluorosulfation product /V control ⁇ 100%.
  • the cytotoxicity difference between a fluorosulfation product and its phenol precursor is quantified as (V phenol /V control ⁇ 100%) ⁇ (V fluorosulfation product /V control ⁇ 100%).
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAXTM) supplement with 5% FBS. Cancer cells were inoculated into a 96-well plate in 100 ⁇ L medium at a plating density of 5,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO 2 for 24 hours. Then the viabilities of cancer cells for time-0 were evaluated by CellTiter-GloTM assay following manual of protocol to obtain V 0 . And another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (1 or 1F) in seven different concentrations from 20 ⁇ M.
  • V test ⁇ V 0 the growth inhibition percentage
  • IC 50 values were obtained from dose-response curves. All tests included 3 repeats.
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAXTM) supplement with 5% charcoal-stripped FBS and 0.01 nM 17 ⁇ -estradiol. Cancer cells were inoculated into a 96-well plate in 100 ⁇ L medium at a plating density of 3,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO 2 for 24 hours. Then the viabilities of cancer cells for time 0 were evaluated by CellTiter-GloTM assay following manual of protocol to obtain V 0 .
  • Another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (11 or 11F) in 5 to 8 different concentrations from 20 ⁇ M (2 ⁇ M for MCF-7 and T47D cells). Then cells were incubated for 6 days with one change of medium containing test compounds on day 3, before cell viabilities were evaluated by CellTiter-GloTM Assay to obtain V control and V test .
  • the growth inhibition percentage is defined as (V test ⁇ V 0 )/(V control ⁇ V 0 ) ⁇ 100%.
  • IC 50 values were obtained from dose-response curves. All tests included 3 repeats.
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAXTM) supplement with 5% FBS. Cancer cells were inoculated into a 96-well plate in 100 ⁇ L medium at a plating density of 5,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO 2 for 24 hours. Then the viabilities of cancer cells for time 0 were evaluated by CellTiter-GloTM assay to obtain V 0 .
  • Another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (25 or 25F) in 7 to 9 different concentrations from 20 ⁇ M (2 ⁇ M for MCF-7 cells). Then cells were incubation for 72 hours, before cell viabilities were evaluated by CellTiter-GloTM assay to obtain V control and V test .
  • the growth inhibition percentage is defined as (V test ⁇ V 0 )/(V control ⁇ V 0 ) ⁇ 100%.
  • IC 50 values were obtained from dose-response curves. All tests included 3 repeats.
  • Estrogen receptor binding assays were preformed using a PolarScreenTM ER ⁇ competitor assay kit (green) from Life Technologies following manual of protocol. This method uses recombinant ER and competition with a fluoromone ligand. The experiment included seven concentrations of test compounds from 10 4 nM to 10 ⁇ 2 nM and three repeats for each concentration. Relative EC 50 values were obtained from dose-response curves.
  • MCF-7 cells were inoculated in a 6-well plate at a seeding density of 10,000 cells/well in DMEM (+Gluta MAXTM) supplement with 5% FBS. After incubation for 24 hours, cells were exposed to either 11 or 11F at the concentrations of 0 nM (0.2% DMSO), 3 nM, 9 nM and 27 nM for 5 days with daily changes of media containing test compounds. T hen cells were lysed and stored at ⁇ 80° C. until western blot for ER ⁇ . After cell lysis, denaturing and centrifuge at 20,000 rpm for 20 min, protein samples in equal volume were subjected to western protocol.
  • Membranes were blocked and then incubated with 1:400 dilution of ER ⁇ antibody at 4° C. overnight followed by 1:5000 dilution of secondary antibody for 1 h at room temperature. GAPDH was detected as loading control. They were then imaged on a ChemiDocTM imaging system (Bio-Rad).
  • A549, MCF-7 and HT-29 cells were inoculated in a 6-well plate at a seeding density of 100,000 cells/well in DMEM (+Gluta MAXTM) supplement with 5% FBS. After incubation for 24 hours, cells were incubated for 24 hours before cells were lysed and stored at ⁇ 80° C. until western blot for UGT1. After cell lysis, denaturing and centrifuge at 20,000 rpm for 20 min, protein samples in equal volume were subjected to western protocol. Membranes were blocked and then incubated with 1:500 dilution of UGT1 antibody at 4° C. overnight followed by 1:5000 dilution of secondary antibody for 1 h at room temperature. GAPDH was detected as loading control. They were then imaged on a ChemiDocTM imaging system (Bio-Rad).
  • HT-29 were inoculated in a 4-well chamber at a seeding density of 10,000 cells/well in DMEM (+Gluta MAXTM) supplement with 5% FBS. After incubation at 37° C. for 24 hours, cells were treated with media containing DMSO (0.2%), 25 (1 ⁇ M, 0.1 ⁇ M) or 25F (1 ⁇ M, 0.1 ⁇ M, 0.01 ⁇ M) for 24 hours. Then cells were gently washed in PBS, fixed for 20 min with 4% paraformaldehyde in PBS and permeabilised in 0.5% Triton X-100TM surfactant.
  • PK pharmacokinetic
  • mice were restrained manually at the designated time points, approximately 110 ⁇ L of blood sample was collected via retro-orbital puncture or cardiac puncture for terminal bleeding under the anesthesia with Isoflurane inhalation into K2EDTA tubes.
  • the blood samples were maintained in wet ice first and centrifuged to obtain plasma (2000g, 4° C., 5 min) within 15 minutes post sampling. Compound concentrations in plasma were determined by LC-MS.
  • PK profile of 11F and fulvestrant (11) is shown in FIG. 9 .
  • the PK parameters are shown in Table 2.
  • Fluorosulfate 11F showed surprisingly good oral bioavailability: Maximum plasma concentration (218 ng/mL) was reached in 3 hours post administration. In contrast, fulvestrant cannot be effectively absorbed orally with AUC 250 h ⁇ ng/mL. The half-life of 11F is around 4 h, which is lower than fulvestrant via S.C. (16.5 h). However, its Area Under Curve value (AUC) is 1380 h ⁇ ng/mL, which is higher than fulvestrant via S.C. (936 h ⁇ ng/mL). Fluorosulfate 11F showed lower bioavailability via S.C. administration.
  • PK 11F Fulvestrant Fulvestrant parameters Unit 11F P.O. S.C. P.O. S.C. T max h 3 1 1 3 C max ng/mL 218 48.7 48.6 61.6 T 1/2 h 3.95 9.51 3.2 16.5 AUC h ⁇ ng/mL 1380 455 250 936

Abstract

Arylfluorosulfate compounds derived from anticancer drugs are disclosed herein, which exhibit improved anti-cancer cell proliferation activities compared to their phenolic drug precursors. Among these compounds, the fluorosulfate derivative of fulvestrant showed significantly enhanced activity to down-regulate estrogen receptor (ER) expression in ER+ breast cancer cell line MCF-7, and oral availability in vivo; the fluorosulfate derivative of combretastatin A4 exhibited a 70-fold increase in potency in the drug resistant colon cancer cell line HT-29; and the fluorosulfate derivative of ABT-751 showed enhanced activity relative to ABT-751.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. application Ser. No. 16/158,608, filed on Oct. 12, 2018, which is a division of U.S. application Ser. No. 15/316,742, filed on Dec. 6, 2016, now U.S. Pat. No. 10,117,840, which is a 371 of PCT/US2015/034516, filed on Jun. 5, 2015, which claims the benefit of U.S. Provisional Application Ser. No. 62/008,925, filed on Jun. 6, 2014, each of which is incorporated herein by reference in its entirety. The application also claims the benefit of U.S. Provisional Application Ser. No. 62/615,328, filed on Jan. 9, 2018, which is incorporated herein by reference in its entirety.
  • STATEMENT OF GOVERNMENT SUPPORT
  • This invention was made with government support under R01GM117145 and R01GM111938 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • BACKGROUND
  • Click chemistry, inspired by Nature's powerful heteroatom-linkage creation strategy, has found broad applications in materials chemistry, chemical biology, and drug development since the concept was first introduced in 1998. The sulfur (VI) fluoride exchange (SuFEx), developed by the Sharpless lab in 2014, represents another set of ideal click chemistry transformations. In SuFEx manifestation, arylfluorosulfates (Ar—O—SO2—F) and iminosulfur oxydifluorides (R—N═SOF2) can be readily synthesized using two gases sulfuryl fluoride (SO2F2) and thionyl tetrafluoride (SOF4), respectively. Though rarely studied in the past, these two S(VI)-F motifs have since been successfully used as connective linkers in polymer synthesis and for the construction of various functional molecules.
  • However, due to the previous inaccessibility of these compounds and their assumed high reactivity toward biomolecules as is the case of other sulfur (VI) halides, the study and application of S(VI)-F in medicinal chemistry has largely been unexplored. In the Sharpless lab's pilot work with the Kelly group at the Scripps Research Institute, it was discovered that arylfluorosulfates are only reactive toward proximal nucleophilic residues, e.g. tyrosine, found within specific protein partners. These unique features make arylfluorosulfates distinct from other sulfur (VI) halides and herald an emerging invaluable tool in drug development.
  • There is an ongoing need to develop new drug candidates, particularly in the area of cancer treatment. The compounds and methods described herein address this need.
  • SUMMARY
  • Described herein is a method of SuFEx chemistry for the conversion of phenolic compounds to their respective arylfluorosulfate derivatives in situ in 96-well plates. This method is compatible with automated synthesis and screening to quickly assess the biological activities of the in situ generated crude products. Using this method, late-stage functionalization of a panel of known anti-cancer drugs generated the corresponding arylfluorosulfates. These in situ generated arylfluorosulfates were directly tested in a cancer-cell growth inhibition assay in parallel with their phenolic precursors. Three arylfluorosulfates exhibited improved anti-cancer cell proliferation activities compared to their phenol precursors (ABT-751, fulvestrant, and combretastatin A4). Among these compounds, the fluorosulfate derivative of fulvestrant showed significantly enhanced activity to down-regulate estrogen receptor (ER) expression in ER+ breast cancer cell line MCF-7; and the fluorosulfate derivative of combretastatin A4 exhibited a 70-fold increase in potency in the drug resistant colon cancer cell line HT-29.
  • In one aspect, a fluorosulfate compound is provided, which comprises a fluorosulfate derivative of a phenolic medicinal agent, e.g., an anticancer agent, wherein at least one phenolic OH group of the medicinal agent is replaced by —OSO2F. In some embodiments the medicinal agent is selected from the group consisting of ATB-751 (1), apigenin (2), ezetimibe (3), topotecan (4), ganetespib (5), etoposide (6), G100713 (7), estrone (8), PI-103 (9), raloxifene (10), fulvestrant (11), mycophenolic acid (12), gimeracil (13), flavopiridol (14), serotonin (15), PAC-1 (16), SKI II (17), endoxifen (18), vanillin (19), A-769662 (20), xanthohumol (21), sirtinol (22), IOX1 (23), TWS119 (24), combretastatin A4 (25), VER-50589 (26), luminespib (27), TW-37 (28), estrol (29), SB415286 (30), rotigotine (31), estradiol (32), RKI-1447 (33), licochalcone A (34), onalespib (35), 2-methoxyestrodiol (36), SB202190 (37), salidroside (38), 10-hydroxycamptothecin (39, 10-HCPT), lasofoxifene, lipid encapsulated 7-ethyl-10-hydroxycamptothecin, (LE-SN38), diethylstilbestrol, amrubicin, genistein, and phenoxodiol.
  • In one embodiment, a compound of Formula 1F is provided, which is a fluorosulfate derivative of ABT-751:
  • Figure US20190167604A1-20190606-C00001
  • which is a useful antimitotic agent for treating certain cancers, e.g., certain breast cancer. The parent, underivatized phenolic compound, ABT-751 reportedly binds to the colchicine site on beta-tubulin, which leads to a block in the cell cycle at the G2M phase, resulting in cellular apoptosis. Thus, ABT-751 has general activity against a variety of cancers and solid tumors, for example. Also disclosed herein is a method of treating a cancer in a patient (e.g., a human), comprising administering an effective dose of a compound of Formula 1F to the patient.
  • In another embodiment, a compound of Formula 11F is provided, which is a fluorosulfate derivative of fulvestrant:
  • Figure US20190167604A1-20190606-C00002
  • and which is useful for treatment of certain cancers, e.g., certain breast cancers. Fulvestrant is indicated for the treatment of hormone receptor positive metastatic breast cancer in postmenopausal women with disease progression following antiestrogen therapy. However, this drug has poor bioavailability and the only administration method is through intramuscular injection at a dose of 250 mg/month. It takes over 3 months to reach a steady serum concentration due to its quick clearance, significantly limiting its clinical value. Also disclosed herein is a method of treating a cancer in a patient (e.g., a human), comprising administering an effective dose of a compound of Formula 11F to the patient.
  • In another embodiment, a compound of Formula 25F is provided, which is a fluorosulfate derivative of combretastatin A4:
  • Figure US20190167604A1-20190606-C00003
  • and which is useful for treatment of certain cancers, e.g., certain breast cancers. Combretastatin A-4 is a tubulin-binding chemotherapy drug that is structurally related to colchicine, and has general anti-cancer activity. Also disclosed herein is a method of treating a cancer in a patient (e.g., a human), comprising administering an effective dose of a compound of Formula 25F to the patient.
  • The foregoing fluorosulfate (O-fluorosulfonyl) derivatives were prepared by contacting the phenol compound with a solution of SO2F2 in acetonitrile at room temperature. The method is adapted for use with a compound library in a high throughput screening format, such as a multi-well plate, e.g., 32-, 64- or 96-well plates, each well containing a compound comprising a phenolic hydroxyl group, for conversion to the corresponding fluorosulfonyl derivative. This library of fluorosulfonylated compounds was then tested in various bioactivity evaluations.
  • The forgoing fluorosulfate compounds can be formulated into a pharmaceutical composition comprising the compound in a pharmaceutically acceptable carrier.
  • In another aspect, the fluorosulfate compounds can be utilized in a method of treatment of cancer in a patient (e.g., a human, or non-human animal), comprising administering an effective dose of the compound to the patient.
  • In yet another aspect, the foregoing fluorosulfate compounds can be used for preparing a medicament for treating cancer.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 provides a comparison of phenolic drug functionalization workflows via the standard SuFEx and in situ SuFEx reactions.
  • FIG. 2 depicts development and optimization in situ SuFEx reaction and processing conditions. (A) comparison of gas-liquid based SuFEx and liquid based SuFEx protocols for reactions in 96 well plates; (B) removal of fluoride ions in situ after reaction: (1) volatiles in the reaction mixture were removed in vacuo, 19F NMR showed the presence of anionic fluorine (−151 ppm, in CDCl3); (2) after treating with trimethylsilanol (TMSOH), volatiles in the reaction mixture were removed in vacuo and 19F NMR showed no presence of anionic fluorine or frimethylsilyl fluoride (TMSF).
  • FIG. 3 shows cytotoxicity profiling of the in situ generated fluorosulfate derivatives 1F to 39F using a double-dose cell-viability assay; (A) thirty-nine in situ SuFEx generated fluorosulfation products 1F to 39F were tested in a cell-viability assay with MCF-7 and A549 cells at the final concentrations of 20 μM and 500 nM. After a 72-hour treatment, the cytotoxicity of fluorosulfate derivatives 1F to 39F and their phenol precursors 1 to 39 were evaluated according to the cancer cell viabilities relative to vehicle (DMSO) control. Bar graphs reflect the % increase or decrease of the cytotoxicity of the corresponding fluorosulfation product compared to its phenol precursor (n=3); (B) cancer cell viabilities under the treatment of in situ generated fluorosulfate derivatives 1F, 11F and 25F and their phenol parents at a final concentration of 20 μM and 500 nM for 72 hours. Cells treated with dimethylsulfoxide (DMSO; 0.2%) as the vehicle control. The structures of fluorosulfate derivatives 1F, 11F and 25F were confirmed by nuclear magnetic resonance after flash column chromatography purification. P values were calculated using two-way ANOVA (n=3); ns: P≥0.05; *: P≤0.1; **: P≤0.01; ***: P≤0.001; ****: P≤0.0001.
  • FIG. 4 demonstrates that fluorosulfate derivative 11F is a new selective estrogen receptor degrader or downregulator (SERD) that binds to and down-regulates Estrogen Receptor; (A) fluorosulfate derivative 11F specifically inhibits the proliferation of ER+ MCF-7 cells; (B) competitive binding curves of Estrogen, fulvestrant (11) and fluorosulfate derivative 11F to ERα evaluated by a PolarScreen™ ERα competitor assay kit from Life Technologies (n=3); (C) compounds 11 and 11F induce ER down-regulation in MCF-7 cells. P values were calculated using two-way ANOVA. n=4; ns: P≥0.05; *: P≤0.1; **: P≤0.01; ***: P≤0.001; ****: P≤0.0001.
  • FIG. 5 provides proliferation inhibition curves of 1 and 1F on different cancer cell lines (n=3).
  • FIG. 6 provides proliferation inhibition curves of 11 and 11F on different cancer cell lines (n=3)
  • FIG. 7 provides proliferation inhibition curves of 25 and 25F on different cancer cell lines (n=3).
  • FIG. 8 depicts fluorosulfate derivatives prepared by in situ reaction of phenolic anticancer compounds with SO2F2 using the methods described herein.
  • FIG. 9 shows a comparison of the PK profile of 11F and fulvestrant (11) following single P.O. or S.C. administrations to female C57BL/6 mice.
  • DETAILED DESCRIPTION
  • Late-stage functionalization (LSF), a strategy for directly introducing functional groups onto a bioactive compound in the late stage of its synthesis, enables rapid diversification of drug candidates or drug-like molecules to improve their properties such as potency and metabolic properties. Many innovative methods have been developed for this endeavor, including late stage C—H functionalization and nucleophilic aromatic substitution, just to name a few. LSF requires a chemical reaction with high selectivity, high yield and mild reaction conditions. Converting a phenolic compound with known biological activities to the corresponding fluorosulfate via SuFEx is an excellent transformation for LSF, which is waiting to be explored further. In fact, the phenolic hydroxyl group is often employed in drug modification and diversification. Converting a phenolic molecules with known biological activities to the corresponding fluorosulfate serves as a quick and cost-effective way to identify new hits with similar or even improved properties. Currently, there are approximately 120 phenolic compounds within the repertoire of FDA-approved drugs. In addition, hundreds of drug candidates bearing phenols are under investigation according to the Canadian online DrugBank database.
  • Current gas-liquid interface-based reaction protocols for the installation of arylfluorosulfates are hurdles for the direct transfer of SuFEx to the drug development pipeline.
  • In a drug discovery process, compounds are usually tested using automated screening to quickly assess their biological activities, which requires a protocol to achieve the chemo-selective and efficient functionalization of screening compounds in situ in multi-well plates with low substrate concentrations and small volumes. Ideally compounds produced in such ways should be subjected to biological assays directly without further purification. The very nature of click chemistry, including high selectivity and yields with minimal byproducts as well as facile purification, provides an excellent solution to meet such requirement.
  • Described herein is a protocol of SuFEx click chemistry for the LSF of phenol-containing drugs or drug candidates for converting the phenolic hydroxyl groups thereof to their respective arylfluorosulfate derivatives in situ in multi-well (e.g. 96-well) plates (FIG. 1). The in situ generated crude products of arylfluorosulfates were directly tested in a cancer-cell growth inhibition assay together with their phenolic precursors. Three arylfluorosulfates exhibited improved anti-cancer proliferation activities compared to their phenol precursors. Among these compounds, the fluorosulfate derivative of combretastatin A4 exhibited a 70-fold increase in potency on the combretastatin-resistant colon cancer cell line HT-29.
  • Development of a Liquid-Based In Situ SuFEx Protocol.
  • As aforementioned, a cost-effective protocol for converting a commercial screening library into a new library should possess the following characteristics: (1) compatible with small reaction scales (e.g., microgram); (2) excellent chemoselectivity; and (3) directly transferable to biological assays. Currently, the typical procedure for synthesizing arylfluorosulfates is performed at the gas-liquid interface: phenol compounds dissolved in organic solvents (e.g., dichloromethane (DCM) or acetonitrile) are subjected to the SO2F2 gas in a sealed reaction vessel in the presence of an organic base (e.g., N,N-Diisopropylethylamine (DIPEA)). This protocol is suitable for high concentration (>100 mM) reactions in which substrates are present in milligram or greater scales, but becomes impractical for low concentration samples (<10 mM) due to dramatically decreased reaction kinetics under such conditions.
  • To develop an in situ SuFEx procedure that can be easily coupled with LSF and quick biological assays, the feasibility of pre-dissolving SO2F2 in an organic solvent to form a saturated solution was explored for use in reacting phenolic compounds with SO2F2 in 96-well plates. Such a procedure is easily conducted by multichannel pipette or a robotic system. Indeed, SO2F2 is known to have good solubility in several organic solvents including carbon tetrachloride and toluene. Toward this end, saturated solutions of SO2F2 in various organic solvents (100 μL) were prepared and mixed with ezetimibe (3), a commercial phenol-containing drug (0.1 μmol in 10 μL dimethyl sulfoxide (DMSO)), in the presence of TEA or DIPEA (10 equiv., in 10 μL of the solvent) in a sealed Eppendorf™ tube at room temperature. The reaction progress was monitored by liquid chromatography-mass spectrometry (LC-MS). Reactions in CH3CN afforded significantly better yields after 3 hours (h) compared to those obtained by using DCM or THF as the solvent.
  • Next, the efficiency of the interfacial and the liquid-based method was compared in a 96-well plate. Phenol compounds 1, 2, 3, and 4 (0.1 μmol) were treated with either SO2F2 atmosphere over an open 96-well plate, or by addition of a dissolved SO2F2 solution (about 4 mg/mL in CH3CN) in a sealed 96-well plate for 12 h (a total volume of 120 μL in each well). All substrates achieved higher yields in the liquid-based system compared to the gas-liquid based method (>20% increase) (FIG. 2, A).
  • Unlike low boiling point solvents and organic bases, fluoride ions generated in this transformation cannot be removed by vacuo (−151 ppm in CDCl3; FIG. 2, B). Fluoride ions may have a synergistic or detrimental effect on arylfluorosulfates in the downstream biological assay. To minimize such influence on the screening results, excess trimethylsilanol (TMSOH, 20 equiv., boiling point 99° C. at 1 atm) was used to convert fluoride ions into volatile trimethylsilyl fluoride (TMSF, −158 ppm in CDCl3) whose boiling point is 16° C. at 1 atm. The subsequent in vacuo treatment could then remove nearly all low boiling point components (TMSF, TMSOH, and TEA), leaving behind only a small amount of unreacted phenol precursors and the arylfluorosulfate products, which could be directly used for the subsequent biological screening tests (FIG. 2, B).
  • Using this liquid-based protocol in a 96-well plate, thirty-nine fluorosulfate derivatives (0.1 μmol) were obtained in good to quantitative yields, whose parent phenolic compounds
  • (FIG. 8) all possess anti-cancer activities. In FIG. 8, R is —SO2F for the fluorosulfate derivatives, whereas R is H for the parent phenolic drugs. The phenol compounds are numbered as 1 to 39 and the corresponding fluorosulfate derivatives are designated 1F to 39F. In FIG. 8, estimated yields for the in situ fluorosulfation reactions are provided after derivative compound numbers, where “quant” means quantitative yield. Notably, some of the compounds bearing multiple phenolic hydroxyl groups ( fluorosulfate derivatives 5, 10, 27 and 37) were fully transformed into the corresponding fluorosulfation products (i.e., all phenolic OH groups reacted). In the case of reaction of compound 28, only one of the three phenolic OH groups reacted (as determined by LC-MS), thus in compound 28F, two of the R groups are H. In the case of compound 2, only two of the three phenolic OH groups reacted. Due to the small quantities involved, and subsequent biological testing, the identities of the unreacted OH groups in 2F and 28F were not determined. Similarly, reaction of compound 26 resulted in a mixture of mono and di-fluorosulfation. Compounds 18 and 34 (as well as 18F and 34F), were mixtures of E/Z isomers. This approach also is applicable to a wide range of substrates and high chemoselectivity: phenol rings bearing electron-withdrawing and electron-donating groups are both reactive with SO2F2. Finally, the crude products were dissolved in DMSO (10 μL; approximately 10 mM) for the direct evaluation of their anti-cancer activities. The common names of the underivatized drugs used to form fluorosulfate derivatives 1F-39F are: 1 (ATB-751), 2 (apigenin), 3 (ezetimibe), 4 (topotecan), 5 (ganetespib), 6 (etoposide), 7 (G100713), 8 (estrone), 9 (PI-103), 10 (raloxifene), 11 (fulvestrant), 12 (mycophenolic acid), 13 (gimeracil), 14 (flavopiridol), 15 (serotonin), 16 (PAC-1), 17 (SKI II), 18 (endoxifen), 19 (vanillin), 20 (A-769662), 21 (xanthohumol), 22 (sirtinol), 23 (IOX1), 24 (TWS119), 25 (combretastatin A4), 26 (VER-50589), 27 (luminespib), 28 (TW-37), 29 (estrol), 30 (SB415286), 31 (rotigotine), 32 (estradiol), 33 (RKI-1447), 34 (licochalcone A), 35 (onalespib), 36 (2-methoxyestrodiol), 37 (SB202190), 38 (salidroside), and 39 (10-HCPT). Additional FDA approved anticancer drugs that are suitable for derivitization with sulfonyl fluoride in the same manner include lasofoxifene, diethylstilbestrol, LE-SN38, amrubicin, genistein, and phenoxodiol. Fluorosulfate derivatives of such compounds also can be utilized in the treatment of the same cancers as the parent compounds.
  • The fluorosulfate compounds described herein can be formulated as a pharmaceutical composition in combination with a pharmaceutically acceptable carrier (also referred to as a vehicle or diluent). Such carriers are well known in the pharmaceutical arts, and include, e.g., 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.
  • In the present specification, the phrase “effective dose” 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 described herein are administered in therapeutically effective dose to treat a disease. Alternatively, an effective dose of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect.
  • Screening the Anti-Cancer Activity of the In Situ SuFEx Generated 1F to 39F.
  • The cytotoxicity of fluorosulfation products 1F to 39F and their phenol precursors was assessed in cancer cell lines using a double-dose cell viability test. The adenocarcinomic human alveolar basal epithelial cell A549 and the breast cancer cell MCF-7 were treated with vehicle (0.2% DMSO), fluorosulfation products or their phenol precursors in a final concentration of 20 μM or 500 nM for 72 h before the cytotoxicity of each compound was assessed according to the viability of cancer cells relative to the vehicle control. The difference between the cytotoxicity of a fluorosulfation product and that of its phenol precursor reflects the changes in compounds' anti-cancer activity after the SuFEx-based derivatization (FIG. 3, A). A fluorosulfation product exhibiting strong cytotoxicity (cell viability relative to the vehicle control less than 50% at both 20 μM and 500 nM on MCF-7 or A549 after a three-day treatment) and significantly increased (>10%) cytotoxicity over the parent compound is considered as a “hit”. Based on these two selection criteria, three hits 1F, 11F and 25F, were identified. As shown in FIG. 3, B, fluorosulfate derivative 1F exhibited significantly enhanced activity relative to phenol precursor 1 at 500 nM (6% on MCF-7 and 27% on A549). The cytotoxicity of fluorosulfate derivative 11F was stronger than that of phenol 11 on MCF-7 cell line with an increase of 32% and 20%, respectively, at 20 μM and 500 nM. However, no significant differences were found on A549 cells. Although phenol 25 and fluorosulfate derivative 25F showed similar activities on MCF-7 cells at 20 μM, fluorosulfate derivative 25F indeed exhibited 8% enhanced cytotoxicity at 500 nM, and its activities are significantly stronger than those of 25 at both concentrations on A549 cells. Notably, the corresponding phenol precursors 1, 11 and 25 are either FDA-approved drugs or are currently under clinical trials. Specifically, 1 (ABT-751), which inhibits polymerization of microtubules, is under a phasel/2 clinic rial; 11 (fulvestrant) is the only selective estrogen receptor down regulator (SERD) for the treatment of ER+ metastatic breast cancer in postmenopausal women whose disease has spread after anti-estrogen therapy; and 25 (combretastatin A4) is a naturally occurring stilbenoid found in plants with activity in a wide range of cancers. The corresponding phosphate of combretastatin A4 is a tumor vascular targeting agent, and the water-soluble prodrug of combretastatin A4.
  • IC50 Evaluation of 1F, 11F and 25F on Cancer Cell Lines.
  • To quantitatively compare the potency of 1F, 11F and 25F with their parent phenolic drugs, we synthesized and purified these compounds in gram quantities and determined their IC50 values on a panel of cancer cell lines.
  • The anti-tumor potencies of arylfluorosulfate 1F and its phenol precursor 1 were evaluated against lung cancer cell line A549, breast cancer cell line MCF-7, SKBR3 and colon cancer cell line HT-29. As shown in Table 1, compound 1 showed IC50 of 632 nM, 266 nM, 13.5 nM and 236 nM on A549, MCF-7, SKBR3 and HT-29 cells, respectively. In comparison, 1F exhibited 2 to 5-fold lower IC50 values on these three cancer cell lines, i.e., 89 nM on A549, 134 nM on MCF-7, 6.9 nM on SKBR3 and 82 nM on HT-29. Similarly, 11F exhibited strong inhibition of the proliferation of ER+ breast cancer cell lines MCF-7 and T47D, with IC50 values of 5.5 nM and 4.8 nM, respectively, which are at the same level with those of 11 (7.7 nM and 14.8 nM, respectively) (Table 1). In addition, both 11 and 11F have no activity on ER MCF-7 and lung cancer A549 cells, suggesting 11F may still target estrogen receptors. Although the measured IC50 values of arylfluorosulfate 25F on A549, MCF-7, and SKBR3 cells were slightly higher compared to those of its parent drug 25 (Table 1), compound 25F showed stronger activities to inhibit these three cells at high concentrations (20 μM and 2 μM) according to dose-response studies, which is consistent with the two-dose screening results (FIG. 3). Surprisingly, the IC50 value of 25F on HT-29 cells, a drug resistant colon cancer cell line, is 70 nM, which is 70-fold lower than the IC50 of compound 25 (4,947 nM). After confirming the anti-tumor potency of these three compounds, two of them (11F and 25F) were selected for further mechanistic studies.
  • TABLE 1
    IC50 values (nM) of arylfluorosulfate derivatives and their
    phenol precursors on different cancer cell lines.
    IC50 (nM)
    SKBR3
    Compound A549Cells MCF-7 Cells Cells HT-29 Cells
    1 (ABT-751) 632 266 13.5 236
    1F 89 134 6.9 82
    11 (fulvestrant) NI 7.7 NI 14.8
    11F NI 5.5 NI 4.8
    25 (combretastatin A4) 28 6.6 0.48 4947
    25F 113 15.6 1.8 70
    NI: No Inhibition (i.e., less than 50% inhibition at 10000 nM)

    Fluorosulfate 11F is a New SERD with Strong Potency.
  • Since fluorosulfate derivative 11F has stronger anti-cancer potency than the commercial drug 11, the cellular target of this new compound was investigated to determine whether it shares the same cellular target with compound 11. As described previously, fulvestrant (11) is the only SERD on the market for the treatment of ER+ breast cancer. Fulvestrant competitively binds to estrogen receptor, which is a crucial regulator of ER+ breast cancer growth, inhibiting dimerization of ER and leading to its degradation. As shown in FIG. 4, A, fluorosulfate 11F inhibited ER+ MCF-7 proliferation, but had no effect on ER MCF-7 cells, suggesting that the anti-cancer activity of 11F is still ER dependent.
  • A competitive displacement ER binding assay, in which the binding affinity of 11 and 11F to ERα protein was compared in competition with a fluoromone ligand was performed to confirm that ER is the target of fluorosulfate 11F. Notably, fluorosulfate 11F binds to ERα with a relative EC50 of 1.5 nM, which is 3-fold stronger than that of fulvestrant (EC50=4.8 nM) (FIG. 4, B). Finally, to determine whether 11F was capable of downregulating the ERα expression level, MCF-7 cells were treated with 11 or 11F at concentrations of 3 nM, 9 nM, or 27 nM for 5 days. The ERα expression levels of the treated cells were then determined by Western blot. Both 11 and 11F downregulated ERα in a dose dependent manner, and 11F showed significantly stronger ERα down-regulation activity (FIG. 4, C). These results confirm that 11F acts as a SERD with strong potency. New SERDs with improved bioavailability are needed. In a recent report, Wang et al. demonstrated that the phenol moiety in 11 could be converted into a borate in a 5-step synthesis. The resulting compound has excellent bioavailability regardless of slightly weaker potency. The cause of its improved bioavailability is believed to derive from blocked phase II metabolism in the blood stream, which is consistent with the finding that fluorosulfate 11F showed surprisingly good oral bioavailability in vivo in C57BL/6 mice, whereas fulvestrant (11) is not effectively orally absorbed (FIG. 9). Importantly, fluorosulfate 11F can be synthesized from 11 using a one-step quantitative “click” procedure, making it readily accessible for the future pharmacokinetics evaluation in animal models.
  • Fluorosulfate 25F Overcomes the Drug Resistance of HT-29.
  • Combretastatin A4 (25) is a one of the most potent anti-vascular agents that targets the colchicine-binding site of β-tubulin and hence disrupts tubulin polymerization. To assess the mechanism of fluorosulfate 25F, HT-29 cells were treated with 25F followed by staining with anti-tubulin-FITC antibody. Confocal imaging revealed that HT-29 cells treated with a vehicle (0.2% DMSO) or combretastatin A4 (25) at various concentrations clearly showed the presence of the microtubule network. Surprisingly, cells treated with fluorosulfate 25F (1 μM and 0.1 μM) for 24 h completely lost microtubule structure, consistent with tubulin depolymerization. Previous investigations of the in vitro and in vivo phase II metabolism revealed that 25 is rapidly converted into the corresponding glucuronide and sulfate metabolites via glucuronidation and sulfation, respectively. In fact, glucuronidation of 25 on the phenol group by uridine 5-diphosphoglucuronosyl transferases (UGTs) has been previously identified as a mechanism of resistance evolved by HT-29 colon cancer cells and hepatocellular cancer cells. Indeed, the expression level of UGT1 in HT-29 cells is significantly higher than those of non-drug-resistant A549 and MCF-7 cells. Combined together, the above observations indicate that 25F inhibits microtubule formation and surprisingly overcomes combretastatin A4 (25) resistance in HT-29 cells.
  • FIGS. 5, 6, and 7 show results of proliferation inhibitions studies in cancer cell lines. The data in the figures show improved activity for the fluorosulfate derivatives relative to the underivatized drug in at least one cell line and dosage level. Without being bound by theory, it is believed that converting a phenol group into an arylfluorosulfate is unlikely to change the protein targeting specificity of the parent compound, but instead this transformation confers the resulting arylfluorosulfate new properties which may enact in distinct mechanisms. In some cases an arylfluorosulfate forms a covalent bond with the target protein via SVI-F exchange given the presence of tyrosine residues in the binding pocket and the presence of residues to facilitate the F departure. In other cases, a fluorosulfate-containing molecule binds to its target via non-covalent interactions stronger than that of the parent phenolic compound, which in some cases blocks the phase II metabolism pathway of the phenol moiety, i.e. sulfation, glucuronidation, and oxidation, which are key routes that change the pharmacokinetic properties of a phenolic compound.
  • All patents and publications referred to herein are incorporated by reference herein to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety.
  • EXAMPLES General
  • 1H NMR spectra were recorded at 400 MHz on Bruker™ AV-400 NMR spectrometers; 13C NMR were recorded at 151 MHz on Bruker™ AV-600. 19F NMR were recorded at 376 MHz on Bruker™ AV-400. All chemical shifts (δ) are quoted in ppm; coupling constants (J) in hertz. Tetramethylsilane was used as international reference for 1H and 13C NMR. Trichlorofluoromethane was used as international reference for 19F NMR. Abbreviations are: s, singlet; d, doublet; t, triplet; q, quartet, p, pentet; br s, broad singlet; m, multiplet. Liquid chromatography-mass spectrometry (LC-MS) was performed on an Agilent™ 1260 LC/MSD with an Agilent™ 6120 quadrupole mass spectrometer (electrospray ionization, ES) eluting with 0.05% trifluoroacetic acid in H2O and 0.05% trifluoroacetic acid in CH3CN. Precoated Merck™ F-254 silica gel plates were used for thin layer analytical chromography (TLC) and visualized with short wave (254 nm) UV light or by potassium permanganate stain. Column chromatography was performed using Silicycle™ Silica Gel 60 (40-63 μm). All phenol compounds were purchased from Selleck chemicals. Sulfuryl fluoride (SO2F2) gas was a gift from Dow AgroSciences. Phenolic compound library (10 mM in DMSO) was purchased from Selleck Chemicals.
  • Example 1. Preparation of Sulfuryl Fluoride (SO2F2) Solution in Organic Solvent
  • A glass vial of organic solvent (5 mL) was evacuated in vacuo and a balloon containing SO2F2 gas was connected with the glass vial, filling the vial with gas. Then the organic solvent was vigorously stirred for 30 min to make the stock solution of sulfuryl fluoride.
  • Example 2. Condition Screening For in situ SuFEx
  • A solution of SO2F2 in organic solvent (CH3CN, DCM or THF, 100 μL) and base (triethylamine (TEA) or N,N-Diisopropylethylamine (DIPEA), 1 μmol in 10 μL corresponding organic solvent) were added in an Eppendorf™ tube containing compound 3 (0.1 μmol in 10 μL DMSO). The tube was sealed by parafilm and left at room temperature for 3 hours before the product and yield was evaluated by LC-MS.
  • Example 3. General Procedure I for the Gas/Liquid Based SuFEx Method on a 96-Well Plate
  • CH3CN (100 μL) and TEA (1 μmol in 10 μL CH3CN) were added in each well of a 96-well plate containing phenol compounds (0.1 μmol in 10 μL DMSO). The plate was left without lid in a vacuum desiccator containing SO2F2(˜1 atm) at room temperature overnight, before the products and yields were evaluated by LC-MS.
  • Example 4. General Procedure II for the Liquid Based SuFEx Method on a 96-Well Plate
  • A solution of SO2F2 in CH3CN (˜4 mg/mL, 100 μL) and TEA (1 μmol in 10 μL) were added in each well of a 96-well plate containing phenol compounds (0.1 μmol in 10 μL DMSO). The plate was left tightly covered by a solvent resistant sealing mat (Corning™ 96 well storage system) at room temperature overnight. Then trimethylsilanol (2 μmol in 10 μL CH3CN) was added to each well and left for 0.5 hours before the plate was left in vacuo overnight. The resulting crudes were dissolved in DMSO (10 μL, ˜10 mM) before biological assays. The products and yields were determined by LC-MS.
  • Example 5. Synthesis of 1F, 11F and 25F in Milligram Scale for IC50 Evaluation 5A. 4-((3-((4-Methoxyphenyl)sulfonamido)pyridin-2-yl)amino)phenyl sulfurofluoridate 1F
  • Figure US20190167604A1-20190606-C00004
  • Triethylamine (TEA) (27.2 mg, 0.27 mmol) was added in a solution of compound 1 (10 mg, 0.027 mmol) in CH3CN (0.5 mL). After evacuation, SO2F2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 12 hours until TLC (hexane/ethyl acetate, 1:1) showed the full conversion of starting material 1 to 1F (Rf 0.70). Then volatiles were removed in vacuo and the resulting crude product was purified by flash column chromatography (hexane/ethyl acetate, 2:1 to 1:2) to obtain fluorosulfate 1F (11.3 mg, 0.025 mmol, 93%) as a light red solid.
  • m.p. 158° C.-160° C.; 1H NMR (400 MHz, CDCl3): 8.15 (dd, 1H, J=4 Hz, 8 Hz), 7.67 (t, 4H, J=8 Hz), 7.26 (d, 2H, J=8 Hz), 6.94 (d, 2H, J=8 Hz), 6.81 (dd, 1H, J=4 Hz, 8 Hz), 6.63 (dd, 1H, J=4 Hz, 8 Hz), 3.85 (s, 3H); 13C NMR (151 MHz, CDCl3): 164.0, 153.0, 147.4, 144.6, 140.8, 136.8, 130.2 (2C), 129.5, 121.6 (2C), 120.5 (2C), 118.1, 115.8, 114.7 (2C), 56.0; 19F NMR (376 MHz, CDCl3): 36.1; ESI (m/z): 454 [M+H]+.
  • 5B. (7R,8R,9S,13S,14S,17S)-17-Hydroxy-13-methyl-7-(9-((4,4,5,5,5-pentafluoropentyl)sulfinyl)nonyl)-7,8,9,11,12,13,14,15,16,17-decahydro-6H-cyclopenta[a]phenanthren-3-yl sulfurofluoridate 11F
  • Figure US20190167604A1-20190606-C00005
  • Triethylamine (TEA) (41.5 mg, 0.41 mmol) was added in a solution of compound 11 (25 mg, 0.041 mmol) in CH3CN (0.5 mL). After evacuation, SO2F2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 1 hours until TLC (hexane/ethyl acetate, 1:1) showed the full conversion of starting material 11 to 11F (Rf 0.40). Then volatiles were removed in vacuo and the resulting crude product was purified by flash column chromatography (hexane/ethyl acetate, 5:1 to 1:2) to obtain fluorosulfate 11F (25.5 mg, 0.037 mmol, 90%) as a colorless foam.
  • 1H NMR (400 MHz, CDCl3): 7.37 (d, 1H, J=8 Hz), 7.09 (d, 1H, J=8 Hz), 7.03 (s, 1H), 3.76 (t, 1H, J=12 Hz), 2.93 (dd, 1H, J=8 Hz, 16 Hz), 2.82-2.59 (m, 5H), 2.40-2.13 (m, 7H), 1.95 (d, J=12 Hz), 1.80-1.72 (m, 3H), 1.65-1.59 (m, 4H), 1.53-1.19 (m, 20H), 0.98 (br s, 1H); 13C NMR (151 MHz, CDCl3): 148.4, 140.8, 139.0, 128.3, 121.9, 118.0, 82.2, 53.0, 51.3, 46.8, 43.6, 41.8, 38.6, 37.1, 34.9, 33.3, 30.9, 30.2, 30.0, 29.9, 29.8, 29.7, 29.5, 29.1, 27.4, 28.4, 26.0, 23.0, 22.92, 22.91, 15.0, 11.4; 19F NMR (376 MHz, CDCl3): 37.2, −85.7, −118.05 to −118.64 (m); ESI (m/z): 689 [M+H]+.
  • 5C. (Z)-2-Methoxy-5-(3,4,5-trimethoxystyryl)phenyl sulfurofluoridate 25F
  • Figure US20190167604A1-20190606-C00006
  • Triethylamine (TEA) (79.9 mg, 0.79 mmol) was added in a solution of compound 25 (25 mg, 0.079 mmol) in CH3CN (0.5 mL). After evacuation, SO2F2 gas was back-filled in the flask and the reaction mixture was left with stirring at room temperature for 2 hours until TLC (hexane/ethyl acetate, 1:1) showed the formation of product 25F (Rf 0.86). Then solvent was removed in vacuo and the resulting crude product was purified by flash column chromatography (hexane/ethyl acetate, 5:1 to 1:1) to obtain fluorosulfate 25F (28.7 mg, 0.072 mmol, 91%) as a light-yellow syrup.
  • 1H NMR (400 MHz, CDCl3): 7.26 (d, 1H, J=4 Hz), 7.24 (d, 1H, J=4 Hz), 7.19 (br s, 1H), 6.93 (d, 1H, J=8 Hz), 6.57 (d, 1H, J=12 Hz), 6.46 (d, 1H, J=12 Hz), 6.43 (s, 2H), 3.89 (s, 3H), 3.84 (s, 3H), 3.70 (s, 6H); 13C NMR (151 MHz, CDCl3): 153.6, 150.4, 150.3, 138.9, 138.8, 137.8, 132.3, 131.3, 130.8, 130.6, 127.8, 123.0, 113.3, 106.0 (2 x C), 61.3, 56.6, 56.2 (2 x C); 19F NMR (376 MHz, CDCl3): 39.5; EI (m/z): 399 [M+H]+.
  • Example 6. In Vitro Biology
  • 6A. Reagents and Software.
  • All culture media are purchased from GIBCO. Charcoal-stripped FBS, PolarScreen™ ERα competitor assay kit (Green) and CellTiter-Glo™ assay kit are purchased from Life Technologies. Mouse monoclonal anti-tublin-FITC antibody (DM1A) is from AbCam Inc. Rabbit monoclonal UGT1 antibody is from Cell Signaling Technology. Rabbit monoclonal ERα antibody is from AbCam Inc. Experimental data were processed by Prism 7™ and western blot images were analyzed by ImageJ 1.50i™ software.
  • 6B. Cell Culture.
  • The MCF-7, A549, SKBR3 and HT-29 cells were originally purchased from ATCC. The T47D and ER MCF-7 cells were kind grifts from Christopher K. Glass group (UCSD). All cancer cell lines were routinely maintained in DMEM (+Gluta MAX™) supplement with 10% FBS and 1% peneciline/streptomycin in 5% carbon dioxide at a temperature of 37° C.
  • 6C. Two-Dose Cell Viability Assay to Compare the Cytotoxicities of Fluorosulfation Products (1F to 39F) and Phenol Parents (1 to 39).
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAX™) supplement with 5% FBS. MCF-7 and A549 cancer cells were inoculated into 96-well plates in 100 μL medium at a plating density of 5,000 cells/well. After cell inoculation, the plates were incubated at 37° C. and 5% CO2. After 24 hours, all wells were refreshed with medium containing DMSO (0.2%, as vehicle control), phenols (20 μM or 500 nM) or in situ generated fluorosulfation products (20 μM or 500 nM). Then cancer cells were maintained at 37° C. and 5 CO2 for 72 hours before cell viability (Vcontrol, Vphenol and Vfluorosulfation product) were evaluated by CellTiter-Glo™ Assay following manual of protocol. Cell viability percentage relative to vehicle control is defined as Vphenol/Vcontrol×100% or Vfluorosulfation product/Vcontrol×100%. The cytotoxicity difference between a fluorosulfation product and its phenol precursor is quantified as (Vphenol/Vcontrol×100%)−(Vfluorosulfation product/Vcontrol×100%).
  • 6D. Cancer Cell Growth Inhibition Assay.
  • Experimental compounds were dissolved in DMSO at 500-fold of the desired final maximum test concentration. Then it was 10-fold serially diluted to other concentrations in DMSO. All aliquots were frozen prior to use. At the time of test, the aliquots of frozen concentrates are thawed and diluted to the desired final test concentrations with DMEM (+Gluta MAX™) supplement with 5% FBS.
  • 6D(i). Growth Inhibition of MCF-7, A549, SKBR3 and HT-29 by 1 and 1F
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAX™) supplement with 5% FBS. Cancer cells were inoculated into a 96-well plate in 100 μL medium at a plating density of 5,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO2 for 24 hours. Then the viabilities of cancer cells for time-0 were evaluated by CellTiter-Glo™ assay following manual of protocol to obtain V0. And another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (1 or 1F) in seven different concentrations from 20 μM. After incubation for 72 hours, cell viabilities were evaluated by CellTiter-Glo™ assay to obtain Vcontrol and Vtest. The growth inhibition percentage is defined as (Vtest−V0)/(Vcontrol−V0)×100%. IC50 values were obtained from dose-response curves. All tests included 3 repeats.
  • 6D(ii). Growth Inhibition of MCF-7, T47D, ER MCF-7 and A549 by 11 and 11F
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAX™) supplement with 5% charcoal-stripped FBS and 0.01 nM 17β-estradiol. Cancer cells were inoculated into a 96-well plate in 100 μL medium at a plating density of 3,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO2 for 24 hours. Then the viabilities of cancer cells for time 0 were evaluated by CellTiter-Glo™ assay following manual of protocol to obtain V0. Another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (11 or 11F) in 5 to 8 different concentrations from 20 μM (2 μM for MCF-7 and T47D cells). Then cells were incubated for 6 days with one change of medium containing test compounds on day 3, before cell viabilities were evaluated by CellTiter-Glo™ Assay to obtain Vcontrol and Vtest. The growth inhibition percentage is defined as (Vtest−V0)/(Vcontrol−V0)×100%. IC50 values were obtained from dose-response curves. All tests included 3 repeats.
  • 6D(iii). Growth Inhibition of MCF-7, A549, SKBR3 and HT-29 using 25 and 25F
  • All cancer cells for the anti-cancer assay were grown in DMEM (+Gluta MAX™) supplement with 5% FBS. Cancer cells were inoculated into a 96-well plate in 100 μL medium at a plating density of 5,000 cells/well. Another same plate of cells was inoculated for the evaluation of time 0. After cell inoculation, the plates were incubated at 37° C. and 5% CO2 for 24 hours. Then the viabilities of cancer cells for time 0 were evaluated by CellTiter-Glo™ assay to obtain V0. Another plate of cancer cells was refreshed with medium containing either DMSO (0.2%, as control) or test compounds (25 or 25F) in 7 to 9 different concentrations from 20 μM (2 μM for MCF-7 cells). Then cells were incubation for 72 hours, before cell viabilities were evaluated by CellTiter-Glo™ assay to obtain Vcontrol and Vtest. The growth inhibition percentage is defined as (Vtest−V0)/(Vcontrol−V0)×100%. IC50 values were obtained from dose-response curves. All tests included 3 repeats.
  • 6E. ER Binding Assay.
  • Estrogen receptor binding assays were preformed using a PolarScreen™ ERα competitor assay kit (green) from Life Technologies following manual of protocol. This method uses recombinant ER and competition with a fluoromone ligand. The experiment included seven concentrations of test compounds from 104 nM to 10−2 nM and three repeats for each concentration. Relative EC50 values were obtained from dose-response curves.
  • 6E(i). Western Blot for ERα Downregulation.
  • MCF-7 cells were inoculated in a 6-well plate at a seeding density of 10,000 cells/well in DMEM (+Gluta MAX™) supplement with 5% FBS. After incubation for 24 hours, cells were exposed to either 11 or 11F at the concentrations of 0 nM (0.2% DMSO), 3 nM, 9 nM and 27 nM for 5 days with daily changes of media containing test compounds. T hen cells were lysed and stored at −80° C. until western blot for ERα. After cell lysis, denaturing and centrifuge at 20,000 rpm for 20 min, protein samples in equal volume were subjected to western protocol. Membranes were blocked and then incubated with 1:400 dilution of ERα antibody at 4° C. overnight followed by 1:5000 dilution of secondary antibody for 1 h at room temperature. GAPDH was detected as loading control. They were then imaged on a ChemiDoc™ imaging system (Bio-Rad).
  • 6E(ii). Western Blot for UGT1 Expression on A549, MCF-7 and HT-29 Cells.
  • A549, MCF-7 and HT-29 cells were inoculated in a 6-well plate at a seeding density of 100,000 cells/well in DMEM (+Gluta MAX™) supplement with 5% FBS. After incubation for 24 hours, cells were incubated for 24 hours before cells were lysed and stored at −80° C. until western blot for UGT1. After cell lysis, denaturing and centrifuge at 20,000 rpm for 20 min, protein samples in equal volume were subjected to western protocol. Membranes were blocked and then incubated with 1:500 dilution of UGT1 antibody at 4° C. overnight followed by 1:5000 dilution of secondary antibody for 1 h at room temperature. GAPDH was detected as loading control. They were then imaged on a ChemiDoc™ imaging system (Bio-Rad).
  • 6E(iii). Immunofluorescence Microscopy of HT-29 Microtubule Disruption induced by 25 and 25F.
  • HT-29 were inoculated in a 4-well chamber at a seeding density of 10,000 cells/well in DMEM (+Gluta MAX™) supplement with 5% FBS. After incubation at 37° C. for 24 hours, cells were treated with media containing DMSO (0.2%), 25 (1 μM, 0.1 μM) or 25F (1 μM, 0.1 μM, 0.01 μM) for 24 hours. Then cells were gently washed in PBS, fixed for 20 min with 4% paraformaldehyde in PBS and permeabilised in 0.5% Triton X-100™ surfactant. Following washes in PBS containing 0.1% Tween™ (PBST) surfactant, cells were blocked in 5% bovine serum albumin diluted in PBST. Then cells were incubated with mouse monoclonal anti-tublin-FITC antibody (DM1A) (1:100 in PBS) for 3 hours. After washing with PBST, cells were incubated with DAPI (1:2000) for 30 min and mounted in PBS for confocal analysis. Images were captured by Nikon™ spinning disk confocal microscopy. All images in each experiment were collected on the same day using identical parameters.
  • Example 7. In Vivo Biology of Fulvestrant and Fluorosulfate 11F.
  • The pharmacokinetic (PK) profiles of fulvestrant (11) and fluorosulfate derivative 11F were evaluated in female C57BL/6 mice using single oral (P.O.) or subcutaneous (S.C.) administrations of the compounds in 5% corn oil +5% DMSO. Results are summarized in FIG. 9.
  • Administration: P.O.: 8.3 mg/kg (10 mL/kg) via oral gavage (N=3); S.C.: 8.3 mg/kg (10 mL/kg) via subcutaneous injection (N=3).
  • Blood Collection and LC-MS: The mice were restrained manually at the designated time points, approximately 110 μL of blood sample was collected via retro-orbital puncture or cardiac puncture for terminal bleeding under the anesthesia with Isoflurane inhalation into K2EDTA tubes. The blood samples were maintained in wet ice first and centrifuged to obtain plasma (2000g, 4° C., 5 min) within 15 minutes post sampling. Compound concentrations in plasma were determined by LC-MS.
  • Result analysis: A comparison of the PK profile of 11F and fulvestrant (11) is shown in FIG. 9. The PK parameters are shown in Table 2. Fluorosulfate 11F showed surprisingly good oral bioavailability: Maximum plasma concentration (218 ng/mL) was reached in 3 hours post administration. In contrast, fulvestrant cannot be effectively absorbed orally with AUC 250 h·ng/mL. The half-life of 11F is around 4 h, which is lower than fulvestrant via S.C. (16.5 h). However, its Area Under Curve value (AUC) is 1380 h·ng/mL, which is higher than fulvestrant via S.C. (936 h·ng/mL). Fluorosulfate 11F showed lower bioavailability via S.C. administration.
  • TABLE 2
    PK Parameters for fulvestrant and fluorosulfate derivative 11F.
    PK 11F Fulvestrant Fulvestrant
    parameters Unit
    11F P.O. S.C. P.O. S.C.
    Tmax h 3 1 1 3
    Cmax ng/mL 218 48.7 48.6 61.6
    T1/2 h 3.95 9.51 3.2 16.5
    AUC h · ng/mL 1380 455 250 936

Claims (16)

What is claimed is:
1. A chemical compound comprising a fluorosulfate derivative of a phenolic anticancer medicinal agent, wherein at least one phenolic OH group of the medicinal agent is replaced by —OSO2F.
2. The compound of claim 1, wherein the medicinal agent is selected from the group consisting of ATB-751 (1), apigenin (2), ezetimibe (3), topotecan (4), ganetespib (5), etoposide (6), G100713 (7), estrone (8), PI-103 (9), raloxifene (10), fulvestrant (11), mycophenolic acid (12), gimeracil (13), flavopiridol (14), serotonin (15), PAC-1 (16), SKI II (17), endoxifen (18), vanillin (19), A-769662 (20), xanthohumol (21), sirtinol (22), IOX1 (23), TWS119 (24), combretastatin A4 (25), VER-50589 (26), luminespib (27), TW-37 (28), estrol (29), SB415286 (30), rotigotine (31), estradiol (32), RKI-1447 (33), licochalcone A (34), onalespib (35), 2-methoxyestrodiol (36), SB202190 (37), salidroside (38), 10-hydroxycamptothecin (39, 10-HCPT), lasofoxifene, lipid encapsulated 7-ethyl-10-hydroxycamptothecin, (LE-SN38), diethylstilbestrol, amrubicin, genistein, and phenoxodiol.
3. The compound of claim 1, wherein the compound has a formula selected from the group consisting of Formula 1F, Formula 11F, and Formula 25F:
Figure US20190167604A1-20190606-C00007
4. A pharmaceutical composition comprising the compound of claim 1 in a pharmaceutically acceptable carrier.
5. A pharmaceutical composition comprising the compound of claim 2 in a pharmaceutically acceptable carrier.
6. A pharmaceutical composition comprising the compound of claim 3 in a pharmaceutically acceptable carrier.
7. A method of treatment of cancer in a patient, comprising administering an effective dose of the compound of claim 1 to the patient.
8. A method of treatment of cancer in a patient, comprising administering an effective dose of the compound of claim 2 to the patient.
9. A method of treatment of cancer in a patient, comprising administering an effective dose of the compound of claim 3 to the patient.
10. The method of claim 7, wherein the patient is a human.
11. The method of claim 8, wherein the patient is a human.
12. The method of claim 9, wherein the patient is a human.
13. The method of claim 9 wherein the compound is a compound of Formula 1F.
14. The method of claim 9, wherein the compound is a compound of Formula 11F.
15. The method of claim 14, wherein the cancer is hormone receptor positive metastatic breast cancer.
16. The method of claim 9, wherein the compound is a compound of Formula 25F.
US16/243,761 2014-06-06 2019-01-09 Arylfluorosulfate compounds and methods Abandoned US20190167604A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/243,761 US20190167604A1 (en) 2014-06-06 2019-01-09 Arylfluorosulfate compounds and methods
US16/871,902 US11779548B2 (en) 2014-06-06 2020-05-11 Arylfluorosulfate compounds and methods

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201462008925P 2014-06-06 2014-06-06
PCT/US2015/034516 WO2015188120A1 (en) 2014-06-06 2015-06-05 Sulfur(vi) fluoride compounds and methods for the preparation thereof
US201615316742A 2016-12-06 2016-12-06
US201862615328P 2018-01-09 2018-01-09
US16/158,608 US10765645B2 (en) 2014-06-06 2018-10-12 Sulfur(VI) fluoride compounds and methods for the preparation thereof
US16/243,761 US20190167604A1 (en) 2014-06-06 2019-01-09 Arylfluorosulfate compounds and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/158,608 Continuation-In-Part US10765645B2 (en) 2014-06-06 2018-10-12 Sulfur(VI) fluoride compounds and methods for the preparation thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/871,902 Continuation US11779548B2 (en) 2014-06-06 2020-05-11 Arylfluorosulfate compounds and methods

Publications (1)

Publication Number Publication Date
US20190167604A1 true US20190167604A1 (en) 2019-06-06

Family

ID=72141377

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/243,761 Abandoned US20190167604A1 (en) 2014-06-06 2019-01-09 Arylfluorosulfate compounds and methods
US16/871,902 Active 2037-04-22 US11779548B2 (en) 2014-06-06 2020-05-11 Arylfluorosulfate compounds and methods

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/871,902 Active 2037-04-22 US11779548B2 (en) 2014-06-06 2020-05-11 Arylfluorosulfate compounds and methods

Country Status (1)

Country Link
US (2) US20190167604A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113121401A (en) * 2020-01-16 2021-07-16 中国科学院上海有机化学研究所 N-substituted carbonyl fluorosulfonamide compound, preparation method and application thereof
CN113248444A (en) * 2021-05-31 2021-08-13 福州大学 Fluorosulfonyl radical reagent and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5112866A (en) * 1988-09-06 1992-05-12 Ortho Pharmaceutical Corporation Ethanesulfonamide derivatives

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5112866A (en) * 1988-09-06 1992-05-12 Ortho Pharmaceutical Corporation Ethanesulfonamide derivatives

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113121401A (en) * 2020-01-16 2021-07-16 中国科学院上海有机化学研究所 N-substituted carbonyl fluorosulfonamide compound, preparation method and application thereof
CN113248444A (en) * 2021-05-31 2021-08-13 福州大学 Fluorosulfonyl radical reagent and preparation method and application thereof

Also Published As

Publication number Publication date
US11779548B2 (en) 2023-10-10
US20200268683A1 (en) 2020-08-27

Similar Documents

Publication Publication Date Title
US9365510B2 (en) Aziridine bisphenol ethers and related compounds and methods for their use
US20160068466A1 (en) Ester derivatives of androgen receptor modulators and methods for their use
RU2767257C2 (en) Bisphenol derivatives and their use as activity modulators of androgen receptor
KR20190018408A (en) Cyclic di-nucleotide compounds and methods of use
US9375496B2 (en) Halogenated compounds for cancer imaging and treatment and methods for their use
US10384997B2 (en) Bio-stable cannabinoid compounds and methods for enhancing their physiological concentration
US11779548B2 (en) Arylfluorosulfate compounds and methods
EP3988541A1 (en) Heterocyclic compounds for cancer imaging and treatment and methods for their use
Zhang et al. Boronic prodrug of endoxifen as an effective hormone therapy for breast cancer
US20220089602A1 (en) Compounds that induce ferroptic cell death
WO2018045450A1 (en) Bisphenol a compounds and methods for treating drug-resistant androgen receptor mediated cancers
Ebrahim et al. (1S, 2E, 4S, 7E, 11E)-2, 7, 11-Cembratriene-4, 6-diol semisynthetic analogs as novel c-Met inhibitors for the control of c-Met-dependent breast malignancies
Liang et al. Design and synthesis of marine sesterterpene analogues as novel estrogen receptor α degraders for breast cancer treatment
US10487056B2 (en) Small molecule androgen receptor inhibitors and methods of use thereof
Song et al. Discovery of bazedoxifene analogues targeting glycoprotein 130
WO2007065869A1 (en) Labelled docetaxel
Li et al. Synthesis and biological evaluation of geldanamycin–ferulic acid conjugate as a potent Hsp90 inhibitor
WO2019139979A2 (en) Arylfluorosulfate compounds and methods
US6566341B1 (en) Derivative of isoindigo, indigo and indirubin for the treatment of cancer
das Neves et al. 2′-Hydroxychalcones as an alternative treatment for trichomoniasis in association with metronidazole
JP2019514843A (en) Uridines phosphoramide prodrugs, process for their preparation and their use in medicine
San Nicolás-Hernández et al. Multi-target withaferin-A analogues as promising anti-kinetoplastid agents through the programmed cell death
CN111511364A (en) Targeted therapeutic agents
Sighel et al. Streptogramin A derivatives as mitochondrial translation inhibitors to suppress glioblastoma stem cell growth
RU2818191C1 (en) Methyl-3-aryl-3a-nitro-4-(trifluoromethyl)-1,2,3,3a,4,9b-hexahydrochromeno[3,4-c]pyrrole-1-carboxylates, having cytotoxic activity on cell line human cervical carcinoma hela, and method for production thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: THE SCRIPPS RESEARCH INSTITUTE, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, JIE;LIU, ZILEI;LI, SUHUA;AND OTHERS;SIGNING DATES FROM 20190309 TO 20190510;REEL/FRAME:049442/0291

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE