WO2009067663A1 - Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds - Google Patents

Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds Download PDF

Info

Publication number
WO2009067663A1
WO2009067663A1 PCT/US2008/084345 US2008084345W WO2009067663A1 WO 2009067663 A1 WO2009067663 A1 WO 2009067663A1 US 2008084345 W US2008084345 W US 2008084345W WO 2009067663 A1 WO2009067663 A1 WO 2009067663A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyne
formula
compound
functional
Prior art date
Application number
PCT/US2008/084345
Other languages
French (fr)
Other versions
WO2009067663A8 (en
Inventor
Geert-Jan Boons
Jun Guo
Xinghai Ning
Margaretha Wolfert
Original Assignee
University Of Georgia Research Foundation, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Georgia Research Foundation, Inc. filed Critical University Of Georgia Research Foundation, Inc.
Priority to JP2010535090A priority Critical patent/JP5498952B2/en
Priority to CN200880125596.0A priority patent/CN101925366B/en
Priority to US12/743,632 priority patent/US8133515B2/en
Priority to DK08852196.8T priority patent/DK2222341T3/en
Priority to EP08852196.8A priority patent/EP2222341B1/en
Publication of WO2009067663A1 publication Critical patent/WO2009067663A1/en
Publication of WO2009067663A8 publication Critical patent/WO2009067663A8/en
Priority to US13/418,676 priority patent/US8940859B2/en
Priority to US14/591,290 priority patent/US9227943B2/en
Priority to US14/967,896 priority patent/US9725405B2/en
Priority to US15/657,601 priority patent/US9932297B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C251/00Compounds containing nitrogen atoms doubly-bound to a carbon skeleton
    • C07C251/32Oximes
    • C07C251/50Oximes having oxygen atoms of oxyimino groups bound to carbon atoms of substituted hydrocarbon radicals
    • C07C251/58Oximes having oxygen atoms of oxyimino groups bound to carbon atoms of substituted hydrocarbon radicals of hydrocarbon radicals substituted by nitrogen atoms not being part of nitro or nitroso groups
    • 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
    • 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
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • 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
    • A61K47/69Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0041Xanthene dyes, used in vivo, e.g. administered to a mice, e.g. rhodamines, rose Bengal
    • A61K49/0043Fluorescein, used in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0052Small organic molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/33Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C211/39Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton
    • C07C211/41Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton containing condensed ring systems
    • C07C211/42Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton containing condensed ring systems with six-membered aromatic rings being part of the condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C251/00Compounds containing nitrogen atoms doubly-bound to a carbon skeleton
    • C07C251/32Oximes
    • C07C251/34Oximes with oxygen atoms of oxyimino groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • C07C251/44Oximes with oxygen atoms of oxyimino groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals with the carbon atom of at least one of the oxyimino groups being part of a ring other than a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/32Esters of carbamic acids having oxygen atoms of carbamate groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C271/34Esters of carbamic acids having oxygen atoms of carbamate groups bound to carbon atoms of rings other than six-membered aromatic rings with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C35/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a ring other than a six-membered aromatic ring
    • C07C35/22Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a ring other than a six-membered aromatic ring polycyclic, at least one hydroxy group bound to a condensed ring system
    • C07C35/37Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a ring other than a six-membered aromatic ring polycyclic, at least one hydroxy group bound to a condensed ring system with a hydroxy group on a condensed system having three rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/657Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings
    • C07C49/683Unsaturated compounds containing a keto groups being part of a ring containing six-membered aromatic rings having unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/16Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/20Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/12Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/94Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom spiro-condensed with carbocyclic rings or ring systems, e.g. griseofulvins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/78Ring systems having three or more relevant rings
    • C07D311/80Dibenzopyrans; Hydrogenated dibenzopyrans
    • C07D311/82Xanthenes
    • C07D311/90Xanthenes with hydrocarbon radicals, substituted by amino radicals, directly attached in position 9
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • 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
    • C07C2603/36Ortho- or ortho- and peri-condensed systems containing three rings containing eight-membered rings

Definitions

  • Bioorthogonal reactions are reactions of materials with each other, wherein each material has limited or substantially no reactivity with functional groups found in vivo.
  • the efficient reaction between an azide and a terminal alkyne i.e., the most widely studied example of "click" chemistry, is known as a useful example of a bioorthogonal reaction.
  • the Cu(I) catalyzed 1,3-dipolar cyclization of azides with terminal alkynes to give stable triazoles e.g., Binder et al., Macromol. Rapid Commun. 2008, 29:952-981
  • Binder et al. Macromol. Rapid Commun. 2008, 29:952-981
  • the cycloaddition has also been used for activity-based protein profiling, monitoring of enzyme activity, and the chemical synthesis of microarrays and small molecule libraries.
  • An attractive approach for installing azides into biomolecules is based on metabolic labeling whereby an azide containing biosynthetic precursor is incorporated into biomolecules using the cells' biosynthetic machinery. This approach has been employed for tagging proteins, glycans, and lipids of living systems with a variety of reactive probes. These probes can facilitate the mapping of saccharide-selective glycoproteins and identify glycosylation sites.
  • Alkyne probes have also been used for cell surface imaging of azide-modif ⁇ ed bio-molecules and a particularly attractive approach involves the generation of a fluorescent probe from a non-fluorescent precursor by a [3+2] cycloaddition.
  • the present invention provides an alkyne, and methods of making an alkyne.
  • the alkyne is of the formula:
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 3 and R 4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker).
  • blends of certain alkynes with a polymer or a copolymer that can optionally form a copolymer micelle which can be useful, for example, for controlling the delivery of drugs as described herein.
  • the alkyne includes: a cleavable linker fragment including at least two ends; an alkyne fragment attached to a first end of the cleavable linker fragment; and a biotinylated fragment attached to a second end of the cleavable linker fragment.
  • the alkyne fragment includes a strained, cyclic alkyne fragment.
  • the alkyne further includes at least one heavy mass isotope.
  • the alkyne further includes at least one detectable label such as a fluorescent label.
  • Alkynes such as those described herein above can be reacted with at least one 1,3-dipole-functional compound (e.g., an azide-functional compound, a nitrile oxide-functional compound, a nitrone-functional compound, an azoxy- functional compound, and/or an acyl diazo-functional compound) in a cyclization reaction to form a heterocyclic compound, preferably in the substantial absence of added catalyst (e.g., Cu(I)).
  • the reaction can take place within or on the surface of a living cell.
  • the at least one 1,3-dipole-functional compound includes a 1 ,3-dipole-functional biomolecule such as a peptide, protein, glycoprotein, nucleic acid, lipid, saccharide, oligosaccharide, and/or polysaccharide.
  • the 1 ,3-dipole- functional biomolecule includes a detectable label such as an affinity label.
  • the heterocyclic compounds formed by the alkyne with the at least one 1,3-dipole- functional compound are also disclosed herein.
  • the reaction between the alkyne and the at least one 1,3-dipole-functional compound can take place within or on the surface of a living cell.
  • the heterocyclic compound includes a biotinylated fragment
  • the heterocyclic compound can be bound to a compound that binds biotin, such as avidin and/or streptavidin.
  • the present invention provides a substrate having an alkyne as described herein on the surface thereof.
  • the substrate can be in the form of a resin, a gel, nanoparticles, or combinations thereof.
  • the substrate is a three-dimensional matrix.
  • the X group of an alkyne of Formula I represents a point of attachment to the surface of the substrate.
  • Such substrates can be useful for immobilizing biomolecules such as peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, and/or polysaccharides.
  • Articles including an immobilized biomolecule, such as a protein immobilized on a three-dimensional matrix, are also disclosed herein.
  • compositions and methods disclosed herein can offer advantages over bioorthogonal reactions known in the art. See, for example, Baskin et al., QSAR Comb. ScL 2007, 26: 1211-1219.
  • X represents CH?
  • Codelli et al., J. Am. Chem.
  • alkynes of Formula I have the capability of reacting not only with azides, but also a variety of other 1,3-dipole-functional compounds. Definitions:
  • Figure 1 illustrates exemplary reagents for labeling azide-functional biomolecules.
  • Figure 2 illustrates Scheme 1 : exemplary reagents and conditions: a) TBSCl, pyridine; b) Br 2 , CHCl 3 ; c) LDA, tetrahydrofuran; d) 4-nitrophenyl chloroformate, pyridine, CH 2 Cl?; e) N,N-dimethylformamide (DMF), triethylamine (TEA).
  • LDA lithium diisopropylamide
  • TBS tert- butyldimethylsilyl.
  • Figure 3 illustrates Scheme 2: exemplary reagents and conditions: a) compound 3 in methanol.
  • Figure 4 illustrates exemplary metal-free cycloadditions of compound 3 with azide-functional amino acid and saccharides.
  • Boc tert-butoxycarbonyl
  • TDS thexyldimethylsilyl.
  • Figure 5 illustrates Scheme 3: exemplary reaction conditions: a) 1, triethylamine, DMF, room temperature, 78%; b) 20% trifluoroacetic acid (TFA), room temperature, 95%; c) 2, TEA, DMF, room temperature, 68%.
  • Figure 6 illustrates embodiments of cell-surface labeling with compounds
  • Jurkat cells grown for three days in the absence or presence of Ac 4 ManNAz 25 micromolar were incubated a) with compounds 2 and 9 (30 micromolar) for 0-180 minutes or b) with compounds 2 and 9 (0-100 micromolar) for 1 hour at room temperature. Next, the cells were incubated with avidin-FITC for 15 minutes at 4°C, after which cell lysates were assessed for fluorescence intensity. Samples are indicated as follows: blank cells incubated with 2 (o) or 9 ( ⁇ ), and Ac,ManNAz cells incubated with 2 (•) or 9 ( ⁇ ).
  • Figure 7 illustrates an embodiment of toxicity assessment of cell labeling procedure and cycloaddition reaction with compound 9.
  • Jurkat cells grown for 3 days in the absence (a) or presence (b) of Ac 4 ManNAz (25 micromolar) were incubated with compound 9 (0 - 100 micromolar) for 1 hour at room temperature.
  • the cells were washed three times and then incubated with avidin conjugated with fluorescein for 15 minutes at 4°C, after which cells were washed three times.
  • Cell viability was assessed at different points during the procedure with trypan blue exclusion; after incubation with 9 (black), after avidin-FITC incubation (grey), and after complete procedure (white).
  • Treatment with Cu 1 Cl (ImM) under the same conditions led to approximately 98% cell death for both the blank and the Ac 4 MaIiNAz treated cells.
  • Figure 8 illustrates fluorescence images for embodiments of cells labeled with compound 9 and avidin-Alexa Fluor 488.
  • CHO cells grown for 3 days in the absence (d-f) or presence (a-c) of Ac 4 ManNAz (100 micromolar) were incubated with compound 9 (30 micromolar) for 1 hour at 4°C (a, d) or room temperature (b, c, e, f).
  • Figure 9 illustrates exemplary compounds comprising an alkyne fragment, a cleavable linker fragment, and a biotinylated fragment.
  • Figure 10 illustrates Scheme 4: exemplary reaction conditions: a) DMF, 80 0 C, 70%; b) potassium thioacetate (KSAc), DMF, 60 0 C, 90%; c) NH 2 NH 2 , ethanol (EtOH), refluxing, 95%; d) N,N-diisopropylethylamine (DIPEA), DMF, 0 0 C, 56%; e) DIPEA, DMF, room temperature, 85%.
  • KSAc potassium thioacetate
  • DIPEA N,N-diisopropylethylamine
  • Figure 1 1 illustrates Scheme 5: exemplary reaction conditions: a) p- toluenesulfonic acid (TsOH), room temperature, 81%; b) DMF, 80 0 C, 86%; c) 0. IN HCl, EtOH, room temperature, 90%; d) 9, NaH, DMF, 0 0 C, 88%; e) 0.
  • TsOH p- toluenesulfonic acid
  • DMF 80 0 C, 86%
  • c 0.
  • Figure 12 illustrates exemplary cleavable linkers.
  • Figure 13 illustrates exemplary alkynes and a reactive diene.
  • Figure 14 illustrates scheme 6: exemplary reaction conditions: a) TMSCH 2 N 2 , BF 3 OEt 2 , DCM, -1O 0 C, 3 hours, 71%; b) NaBH 4 , 1 : 1 EtOH/THF, room temperature, 7 hours, 100%; C) Br 2 , CHCl 3 , room temperature, 0.5 hour, 58%; d) LDA, THF, 0.5 hour, 57%; e) Dess-Martin reagent, DCM, 0.5 h; f) 4- nitrophenyl chloroformate, pyridine, DCM, 18 hours, 92%; g) tris(ethylene glycol)- 1 ,8-diamine, TEA, DCM, room temperature, 3 hours, 80%; h) bromoacetic acid, NaH, THF, 22%; i) tris(ethylene glycol)- 1,8-diamine, HATU coupling reagent, DIPEA, DMF, room temperature, 2 hours, 7
  • Figure 15 illustrates compounds 61-68 and second order constants.
  • Figure 16 illsutrates scheme 7: exemplary reagents and conditions: a) LiAlH 4 , AlCl 3 , Et 2 O, O 0 C, 61%; b) Br 2 , CHCl 3 , O 0 C, 58%; c) potassium t- butoxide (Y-BuOK), THF, room temperature, 25%.
  • Figure 17 illsutrates scheme 8: exemplary reagents and conditions: a) TEA, CH 2 Cl?, room temperature, 77%.
  • Figure 18 illustrates scheme 9: exemplary reagents and conditions: a) NaH, benzyl bromide, DMF, room temperature, 59%; b) acetic anhydride (Ac 2 O), pyridine, room temperature, 81%.
  • Figure 19 illustrates scheme 10: exemplary reagents and conditions: a) LDA, Et 3 SiCl, THF, room temperature, 85%; b) SELECTFLUOR fluorinating reagent, DMF, room temperature, 66% c) LDA, Et 3 SiCl, THF, room temperature, 79%; d) SELECTFLUOR fluorinating reagent, DMF, room temperature, 51%; e) NaBH 4 , EtOH, room temperature, 78%; f) Br 2 , CHCl 3 , O 0 C, 46%; g) c) r-BuOK, THF, room temperature, 49%.
  • Figure 20 illustrates the use of copolymer micelles for drug delivery.
  • Figure 21 illustrates the preparation of macromolecules with 4- dibenzocyclooctyne functionality.
  • Figure 22 illustrates cycloadditions of 4-dibenzocyclooctynol with various nitrones. Compounds were mixed at 1 : 1 molar ratio at a final concentration of 6mM and reacted for a time indicated.
  • Alkynes such as those described herein can be reacted with at least one 1,3-dipole-functional compound in a cyclization reaction to form a heterocyclic compound.
  • the reaction can be earned out in the substantial absence of added catalyst (e.g., Cu(I)).
  • exemplary 1,3-dipole- functional compounds include, but are not limited to, azide-functional compounds, nitrile oxide-functional compounds, nitrone-functional compounds, azoxy-functional compounds, and/or acyl diazo-functional compounds.
  • Exemplary alkynes include alkynes of the formula:
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group (and preferably a Cl-Cl O organic moiety); each R ⁇ is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group (and preferably a Cl-ClO organic moiety);
  • each R 1 represents hydrogen and/or each R 2 represents hydrogen.
  • R 3 includes a covalently bound organic dye (e.g., a fluorescent dye).
  • organic group is used for the purpose of this invention to mean a hydrocarbon group that is classified as an aliphatic group, cyclic group, or combination of aliphatic and cyclic groups (e.g., alkaryl and aralkyl groups).
  • suitable organic groups for compounds of this invention are those that do not interfere with the reaction of an alkyne with a 1,3-dipole-functional compound to form a heterocyclic compound.
  • aliphatic group means a saturated or unsaturated linear or branched hydrocarbon group. This term is used to encompass alkyl, alkenyl, and alkynyl groups, for example.
  • alkyl group means a saturated linear or branched monovalent hydrocarbon group including, for example, methyl, ethyl, ⁇ -propyl, isopropyl, tert-butyl, amyl, heptyl, and the like.
  • alkenyl group means an unsaturated, linear or branched monovalent hydrocarbon group with one or more olefmically unsaturated groups (i.e., carbon-carbon double bonds), such as a vinyl group.
  • alkynyl group means an unsaturated, linear or branched monovalent hydrocarbon group with one or more carbon-carbon triple bonds.
  • cyclic group means a closed ring hydrocarbon group that is classified as an alicyclic group, aromatic group, or heterocyclic group.
  • alicyclic group means a cyclic hydrocarbon group having properties resembling those of aliphatic groups.
  • aromatic group or “aryl group” means a mono- or polyiiuclear aromatic hydrocarbon group.
  • heterocyclic group means a closed ring hydrocarbon in which one or more of the atoms in the ring is an element other than carbon (e.g., nitrogen, oxygen, sulfur, etc.).
  • group and “moiety” are used to differentiate between chemical species that allow for substitution or that may be substituted and those that do not so allow for substitution or may not be so substituted.
  • group when the term “group” is used to describe a chemical substituent, the described chemical material includes the unsubstituted group and that group with nonperoxidic O, N, S, Si, or F atoms, for example, in the chain as well as carbonyl groups or other conventional substituents.
  • moiety is used to describe a chemical compound or substituent, only an unsubstituted chemical material is intended to be included.
  • alkyl group is intended to include not only pure open chain saturated hydrocarbon alkyl substituents, such as methyl, ethyl, propyl, tert-buty ⁇ , and the like, but also alkyl substituents bearing further substituents known in the art, such as hydroxy, alkoxy, alkylsulfonyl, halogen atoms, cyano, nitro, amino, carboxyl, etc.
  • alkyl group includes ether groups, haloalkyls, nitroalkyls, carboxyalkyls, hydroxyalkyls, sulfoalkyls, etc.
  • the phrase “alkyl moiety” is limited to the inclusion of only pure open chain saturated hydrocarbon alkyl substituents, such as methyl, ethyl, propyl, tert-butyl, and the like.
  • R 3 can have the formula -(CH 2 ) a C(O)Y, wherein: a is 1-3; Y represents OH or NHR 5 ; and R 5 represents hydrogen or a biotinylation product of a primary amine-containing organic group.
  • the primary amine-containing group can, for example, be of the formula
  • X can represent CHOR 3 , wherein R 3 is selected from the group consisting of an alkyl group, an aryl group, an alkaryl group, and an aralkyl group.
  • R 3 can have the formula -C(O)Z, wherein: Z represents an alkyl group, OR 6 , or NHR 7 ; and R 6 and R 7 are each independently selected from the group consisting of an alkyl group, an aryl group, an alkaryl group, and an aralkyl group.
  • R 7 can be a biotinylation product of a primary amine-containing organic group.
  • Another exemplary alkyne of Formula I is the species in which X represents CHOH, an alkyne of the formula:
  • Another exemplary alkyne of Formula I is the species in which X represents CHNH 2 , an alkyne of the formula:
  • Additional exemplary alkynes include alkynes that have: a cleavable linker fragment including at least two ends; an alkyne fragment attached to a first end of the cleavable linker fragment; and a biotinylated fragment attached to a second end of the cleavable linker fragment.
  • the alkyne fragment includes a strained, cyclic alkyne fragment.
  • the alkyne further includes at least one heavy mass isotope.
  • the alkyne further includes at least one detectable label (e.g., a fluorescent label).
  • X can represent a polymeric or a copolymeric group.
  • the copolymeric group can include a hydrophilic segment and a hydrophobic segment.
  • the method includes: brominating an alkene of the formula:
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R " is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R and R 4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker).
  • 1,3-dipole-functional compounds can be used to react with the alkynes disclosed herein.
  • a "1,3-dipole-functional compound” is meant to include compounds having at least one 1,3-dipole group attached thereto.
  • a "1,3-dipole group” is intended to refer to a group having a three-atom pi-electron system containing 4 electrons delocalized over the three atoms.
  • Exemplary 1,3-dipole groups include, but are not limited to, azides, nitrile oxides, nitrones, azoxy groups, and acyl diazo groups.
  • the 1 ,3-dipole-functional compound can be a biomolecule having at least one 1,3-dipole group attached thereto.
  • the at least one 1,3-dipole-functional compound can include a detectable label (e.g., an immunoassay or affinity label).
  • One or more 1,3-dipole-functional compounds can be combined with an alkyne as described herein under conditions effective to react in a cyclization reaction and form a heterocyclic compound.
  • conditions effective to form the heterocyclic compound can include the substantial absence of added catalyst.
  • Conditions effective to form the heterocyclic compound can also include the presence or absence of a wide variety of solvents including, but not limited to, aqueous (e.g., water) and non- aqueous solvents; protic and aprotic solvents; polar and non-polar solvents; and combinations thereof.
  • the heterocyclic compound can be formed over a wide temperature range, with a temperature range of 0 0 C to 40 0 C (and in some embodiments 23 0 C to 37°C) being particularly useful when biomolecules are involved. Conveniently, reaction times can be less than one day, and sometimes one hour or even less.
  • the cyclization reaction between the one or more 1,3-dipole-functional compounds and the alkyne can take place within or on the surface of a living cell. Such reactions can take place in vivo or ex vivo.
  • in vivo refers to a reaction that is within the body of a subject.
  • ex vivo refers to a reaction in tissue (e.g., cells) that has been removed, for example, isolated, from the body of a subject.
  • Tissue that can be removed includes, for example, primary cells (e.g., cells that have recently been removed from a subject and are capable of limited growth or maintenance in tissue culture medium), cultured cells (e.g., cells that are capable of extended growth or maintenance in tissue culture medium), and combinations thereof.
  • primary cells e.g., cells that have recently been removed from a subject and are capable of limited growth or maintenance in tissue culture medium
  • cultured cells e.g., cells that are capable of extended growth or maintenance in tissue culture medium
  • R 8 can include a detectable label (e.g., an affinity label).
  • each R is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 3 and R 4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker); and R represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
  • R 8 can include a detectable label (e.g., an affinity label).
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-Cl O organic group
  • each R " is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 3 and R 4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker); and R represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
  • at least one R 10 can include a detectable label (e.g., an affinity label).
  • the cyclization reaction of a nitrone- functional compound of the formula (R 10 ) 2 CN(R 10 )O with an exemplary alkyne of Formula I can result in one or more heterocyclic compounds of the formulas:
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R " is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 3 , R 4 , and R 10 independently represents hydrogen or an organic group, with the proviso that at least one R 10 represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
  • at least one R 1 can include a detectable label (e.g., an affinity label).
  • each R 1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group
  • each R 3 , R 4 , and R 10 independently represents hydrogen or an organic group, with the proviso that at least one R 10 represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
  • the heterocyclic compound formed from the cyclization reaction between the alkyne and the one or more 1 ,3-dipole- functional compounds includes a detectable label
  • the heterocyclic compound can be detected using the detectable label.
  • the detectable label is an affinity label
  • affinity binding e.g., affinity chromatography
  • the heterocyclic compound formed from the cyclization reaction between the alkyne and the one or more 1 ,3 -dipole- functional compounds includes a biotinylated fragment
  • the heterocyclic compound can be bound by contacting the heterocyclic compound with a compound that binds biotin (e.g., avidin and/or streptavidin). Further, the bound heterocyclic compound can be detected by methods described herein.
  • Cyclization reactions between alkynes as disclosed herein and 1,3-dipole- functional compounds can be used for a wide variety of applications.
  • an alkyne as disclosed herein can be attached to the surface of a substrate.
  • the X group of the alkyne represents a point of attachment to the surface of the substrate.
  • the X group can advantageously be selected to include functionality (e.g., biotin, activated esters, activated carbonates, and the like) to enable attachment of the alkyne to a functional substrate (e.g., amine functionality, thiol functionality, and the like) through a wide variety of reactions.
  • Substrates having an alkyne attached to the surface thereof can be reacted with 1,3-dipole-functional compounds to form heterocyclic compounds, effectively chemically bonding the 1 ,3-dipole-functional compounds to the substrate.
  • substrates can be, for example, in the form of resins, gels, nanoparticles (e.g., including magnetic nanoparticles), or combinations thereof.
  • such substrates can be in the form of microarrays or even three-dimensional matrices or scaffolds.
  • Exemplary three-dimensional matrices include, but are not limited to, those available under the trade designations ALGIMATRIX 3D Culture system, GELTRIX matrix, and GIBCO three-dimensional scaffolds, all available from Invitrogen (Carlsbad, CA). Such three- dimensional matrices can be particularly useful for applications including cell cultures.
  • 1 ,3-Dipole-functional biomolecules e.g., 1 ,3-dipole-functional peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, and/or polysaccharides
  • 1 ,3-Dipole-functional biomolecules can be immobilized on, and preferably covalently attached to, a substrate surface by contacting the 1 ,3-dipole-functional biomolecules with a substrate having an alkyne attached to the surface thereof under conditions effective for a cyclization reaction to form a heterocyclic compound.
  • conditions effective to form the heterocyclic compound can include the substantial absence of added catalyst.
  • Conditions effective to form the heterocyclic compound can also include the presence or absence of a wide variety of solvents including, but not limited to, aqueous (e.g., water and other biological fluids) and non-aqueous solvents; protic and aprotic solvents; polar and non-polar solvents; and combinations thereof.
  • the heterocyclic compound can be formed over a wide temperature range, with a temperature range of 0 0 C to 40 0 C (and in some embodiments 23 0 C to 37°C) being particularly useful. Conveniently, reaction times can be less than one day, and sometimes one hour or even less.
  • the cyclization reaction can result in an article having a protein immobilized on a three-dimensional matrix.
  • matrices can have a wide variety of uses including, but not limited to, separating and/or immobilizing cell lines.
  • Particularly useful proteins for these applications include, but are not limited to, collagen, fibronectin, gelatin, laminin, vitronectin, and/or other proteins commonly used for cell plating.
  • cyclization reactions between 1,3-dipole-functional compounds and alkynes of Formula I in which X represents a polymeric or a copolymeric group can be used, for example, for controlling the delivery of drugs as described herein below.
  • alkynes of Formula I in which X represents a copolymeric group including a hydrophilic segment and a hydrophobic segment can be blended with a polymer or a copolymer.
  • a copolymer micelle can be formed when an alkyne of Formula I in which X represents a copolymeric group including a hydrophilic segment and a hydrophobic segment is blended with a copolymer having a hydrophilic segment and a hydrophobic segment, a copolymer micelle can be formed.
  • copolymer micelles that include an alkyne of Formula 1 as described herein above can advantageously be used to control the delivery of drugs.
  • a copolymer micelle that includes an alkyne of Formula 1 can be combined with at least one 1,3-dipole-functional drug and allowed to react under conditions effective to form a heterocyclic compound and attach the drug to the copolymer micelle.
  • Nishiyama et al., Adv. Polym. Sci. 2006, 193:67-101 Gaucher et al., J. Control. Release 2005, 109:169-188; Choi et al., J. Dispersion Sci. Tech. 2003, 24:475-487; Lavasanifar et al., Adv. Drug Delivery Rev. 2002, 54: 169-190; and Rosier et al., Adv. Drug Delivery Rev. 2001 , 53:95-108.
  • the novel cycloaddition reaction provided by the invention can be used for labeling of living cells.
  • cells can first be metabolically labeled with an azide- functional precursor to produce azide-functional biomolecules (also referred to as bioconjugates) such as azide-functional glycoproteins (also referred to as glycoconjugates).
  • the cells can then be contacted with an alkyne of Formula I, either in solution or on a substrate as discussed above, under conditions to permit labeling (via the cycloaddition reaction) of the azide- functional biomolecules at the surface of the cell.
  • the resulting triazole conjugate can be detected at the cell surface, or it can be endocytosed by the cell and detected inside the cell.
  • Alkynes of Formula I can also have utility for imaging applications including, for example, as reagents for magnetic resonance imaging (MRI).
  • alkynes of Formula I can contain a fluorescent tag.
  • Alkynes of Formula I can also be useful in qualitative or quantitative proteomics and glycomics applications utilizing mass spectrometry.
  • the alkyne of Formula I can be selected to contain one or more heavy mass isotopes, such as deuterium, 13 C, 15 N, 35 S and the like, and then can be used to label and/or immobilize azide- functional biomolecules as described herein.
  • Alkynes of Formula I can also have utility for applications such as vaccines.
  • alkynes of Formula I can be reacted with an azide- functional protein (e.g., an azide-functional carbohydrate, an azide-functional peptide, and/or an azide-functional glycopeptide), and the resulting triazole conjugate can be used as a carrier protein for the vaccine.
  • an azide- functional protein e.g., an azide-functional carbohydrate, an azide-functional peptide, and/or an azide-functional glycopeptide
  • Alkyne probes have also been used for cell- surface imaging of azide-modified biomolecules, and a particularly attractive approach involves the generation of a fluorescent probe from a nonfluorescent precursor by a [3+2] cycloaddition (Sivakumar et al., Org. Lett. 2004, 6:4603- 4606).
  • alkynes can be activated by ring strain, and, for example, constraining an alkyne within an eight-membered ring creates 18 kcalmol "1 of strain, much of which is released in the transition state upon [3+2] cycloaddition with an azide (Turner et al., J. Am. Chem. Soc. 1973, 95:790 -792; Agard et al., J. Am. Chem. Soc. 2004, 126:15046-15047).
  • cyclooctynes such as 1 react with azides at room temperature without the need for a catalyst ( Figure 1 ).
  • strain-promoted cycloaddition has been used to label biomolecules without observable cytotoxicity (Agard et al., J. Am. Chem. Soc. 2004, 126: 15046-15047).
  • the scope of the approach has, however, been limited because of the slow rate of reaction (Agard et al., ACS Chem. Biol. 2006, 1 :644-648). Appending electron-withdrawing groups to the octyne ring can increase the rate of strain-promoted cycloadditions; however, currently
  • Staudinger ligation with phosphine 2 offers the most attractive reagent for cell- surface labeling with azides.
  • 4-dibenzocyclooctynols such as compound 3 would be ideal for labeling living cells with azides because the aromatic rings are expected to impose additional ring strain and conjugate with the alkyne, thereby increasing the reactivity of the alkyne in metal-free [2+3] cycloadditions with azides.
  • the compound should, however, have excellent stability because the ortho hydrogen atoms of the aromatic rings shield the alkyne from nucleophilic attack.
  • the hydroxy group of 3 provides a handle for the incorporation of tags such as fluorescent probes and biotin.
  • Compound 3 could be prepared easily from known (Jung et al., J. Org. Chem. 1978, 43:3698-3701 ; Jung and Miller, J. Am. Chem. Soc. 1981 , 103:1984- 1992) 3-hydroxy-l ,2:5,6-dibenzocycloocta-l,5,7-triene (4 ) by protection of the hydroxy group as a TBS ether to give 5, which was brominated to provide dibromide 6 in a yield of 60% (Scheme 1 ; Figure T). The TBS protecting group was lost during the latter transformation, but the bromination was low yielding when performed on alcohol 4.
  • Compound 3 has an excellent, long shelf life and after treatment did not react with nucleophiles such as thiols and amines. However, upon exposure to azides a fast reaction took place and gave the corresponding triazoles in high yield. For example, triazoles 10-13 were obtained in quantitative yields as mixtures of regioisomers by reaction of the corresponding azido-containing sugar and amino acid derivatives with 3 in methanol for 30 minutes (Scheme 2; Figure 3 and Figure 4).
  • Compound 9 could easily be prepared by a two-step reaction involving treatment of 3 with 4-nitrophenyl chloroformate to give activated intermediate 7, followed by immediate reaction with 8.
  • 4-dibenzocyclooctynol (9) may also be functionalized with a fluorescent tag to yield a fluorescent derivative (Scheme 3; Figure 5).
  • Jurkat cells were employed that were grown in the absence of Ac 4 MaIiNAz. The cells were exposed to a 30 micromolar solution of compound 9 for various time periods, and after washing, the cells were stained with avidin- fluorescein isothiocyanate (FITC) for 15 minutes at 4°C. The efficiency of the two-step cell-surface labeling was determined by measuring the fluorescence intensity of the cell lysates. For comparison, the cell-surface azido moieties were also labeled by Staudinger ligation with biotin-modified phosphine 2 followed by treatment with avidin-FITC. The labeling with 9 was almost complete after an incubation time of 60 minutes ( Figure 6a).
  • FITC avidin- fluorescein isothiocyanate
  • CHO cells adherent Chinese hamster ovary (CHO) cells were cultured in the presence of Ac 4 ManNAz (100 micromolar) for three days.
  • the resulting cell-surface azido moieties were treated with 9 (30 micromolar) for 1 hour and then with avidin-AlexaFluor488 for 15 minutes at 4°C.
  • staining was observed only at the surface ( Figure 8), and the labeling procedure was equally efficient when performed at either ambient temperature or 4°C.
  • blank cells exhibited very low fluorescence staining, confirming that background labeling is negligible.
  • DIFO difluorinated cyclooctyne
  • 4-Dibenzocyclooctynols such as 3 and 9 have several advantageous features for researchers such as ease of chemical synthesis and the possibility to further enhance the rate of cycloaddition by functionalization of the aromatic moieties. Modifying the aromatic rings may also offer an exciting opportunity to obtain reagents that become fluorescent upon [3+2] cycloaddition with azido- containing compounds, which will make it possible to monitor in real time the trafficking of glycoproteins and other biomolecules in living cells.
  • CDCI3 and chemical shifts ( ⁇ ) are given in ppm relative to solvent peaks ( 1 H, ⁇ 7.24; 13 C, ⁇ 77.0) as internal standard for protected compounds.
  • Negative ion matrix assisted laser desorption ionization time of flight (MALDI-TOF) were recorded on a VOYAGER-DE Applied Biosystems using dihydrobenzoic acid as a matrix.
  • High-resolution mass spectra were obtained using a VOYAGER-DE Applied Biosystems in the positive mode by using 2,5-dihydroxyl-benzoic acid in THF as matrix.
  • tert-Butyl dimethyl silyl chloride (3.0 g, 20 mmol) was added to a stirred solution of 4 (2.2 g, 10 mmol) in a mixture Of CH 2 Cl 2 (20 mL) and pyridine (5 mL). After stirring for 6 hours at room temperature, the reaction mixture was diluted with water and extracted with CH 2 Cl 2 (40 mL). The combined organic extracts were washed with water and brine and then dried (MgSO 4 ).
  • reaction mixture was stirred for 1 hour at room temperature, after which the DMF was removed in vacuo to give an oily residue, which was purified by flash silica gel column chromatography (CH 2 C1 2 /CH 3 OH, 25/1, v/v) to afford 7V-Boc-iV'-biotinyl-3,6-dioxaoctane-l, 8- diamine (2.0 g, 90%).
  • N-Boc-I ⁇ -biotinyl-3, 6-dioxaoctane-l , 8-diamine (1.9 g, 4 mmol) was dissolved in 50% TFA in CH 2 Cl 2 (20 mL) and stirred for 1 hour at room temperature. The solvents were evaporated under reduced pressure to give an oily residue, which was purified by flash silica gel column chromatography (CH 2 C1 2 /CH 3 OH, 10/1 , v/v) to afford 7 (1.3 g, 92%).
  • Synthetic compounds 2 and 9 were reconstituted in DMF and stored at 80 0 C. Final concentrations of DMF never exceeded 0.56% to avoid toxic effects.
  • Human Jurkat cells (Clone E6-1; ATCC) were cultured in RPMI 1640 medium (ATCC) with L-glutamine (2 mM), adjusted to contain sodium bicarbonate (1.5 g L “1 ), glucose (4.5 g L “1 ), HEPES (10 mM), and sodium pyruvate (1.0 mM) and supplemented with penicillin (100 u mL ⁇ ')/streptomycin (100 micrograms mL "1 ; Mediatech) and fetal bovine serum (FBS, 10%;
  • Hyclone Cells were maintained in a humid 5% CO 2 atmosphere at 37°C.
  • Jurkat cells were grown in the presence of peracetylated ./V-azidoacetylmannosamine (Ac4ManNaz; 25 micromolar final concentration) for 3 days, leading to the metabolic incorporation of the corresponding /V-azidoacetyl sialic acid (SiaNAz) into their cell surface glycoproteins.
  • Jurkat cells bearing azides and untreated control cells were incubated with the biotinylated compounds 2 and 9 (0-100 micromolar) in labeling buffer (DPBS, supplemented with FBS (1%)) for 0-180 minutes at room temperature.
  • DPBS labeling buffer
  • the cells were washed three times with labeling buffer and then incubated with avidin conjugated with fluorescein (Molecular Probes) for 15 minutes at 4°C. Following three washes and cell lysis, cell lysates were analysed for fluorescence intensity (485 ex / 520 em) using a microplate reader (BMG Labtech). Data points were collected in triplicate and are representative of three separate experiments. Cell viability was assessed at different points in the procedure with exclusion of trypan blue. Cell labeling and detection by fluorescence microscopy
  • CHO cells Chinese hamster ovary (CHO) cells (Clone Kl ; ATCC) were cultured in Kaighn's modification of Ham's F-12 medium (F- 12K) with L-glutamine (2 mM), adjusted to contain sodium bicarbonate (1.5 g L "1 ) and supplemented with penicillin (100 u mL "1 ) / streptomycin (100 micrograms mL ⁇ ' and FBS (10%). Cells were maintained in a humid 5% CO? atmosphere at 37°C. CHO cells were grown in the presence of Ac4ManNaz (100 micromolar final concentration) for 3 days to metabolically incorporate SiaNAz into their cell surface glycoproteins.
  • CHO cells bearing azides and untreated control cells were then transferred to a glass coverslip and cultured for 36 hours in their original medium.
  • Live CHO cells were treated with the biotinylated compound 9 (30 micromolar) in labeling buffer (DPBS, supplemented with FBS (1%)) for 1 hour at 4°C or at room temperature, followed by incubation with avidin conjugated with Alexa Fluor 488 (Molecular Probes) for 15 minutes at 4°C.
  • Cells were washed 3 times with labeling buffer and fixed with formaldehyde (3.7% in PBS) or incubated for 1 hour at 37°C before fixation.
  • the nucleus was labeled with the far red fluorescent TO-PRO-3 dye (Molecular Probes).
  • the cells were mounted with PermaFluor (Thermo Electron Corporation) before imaging. Initial analysis was performed on a Zeiss Axioplan2 fluorescent microscope. Confocal images were acquired using a 6OX (NAl .42) oil objective. Stacks of optical sections were collected in the z dimensions. The step size, based on the calculated optimum for each objective, was between 0.25 and 0.5 micrometers. Subsequently, each stack was collapsed into a single image (..--projection). Analysis was perfo ⁇ ned offline using ImageJ 1.39f software (National Institutes of Health, USA) and Adobe Photoshop CS3 Extended Version 10.0 (Adobe Systems Incorporated), whereby all images were treated equally.
  • Example 2 Alkyne Reagents Containing Biotin and a Cleavable Linker
  • Azides which are extremely rare in biological systems, are emerging as attractive chemical handles for bioconjugation (Dedola et al., Org. Biomol. Chem. 2007, 5, 1006; KoIb and Sharpless, Drug D is. Today 2003, 8, 1 128; Moses and Moorhouse, Chem. Soc. Rev. 2007, 36, 1249; Nandivada et al., Adv. Mater. 2007, 19, 2197; Wu and Fokin, Aldrichimica ACTA 2007, 40, 7; Agard et al., ACS Chem. Biol. 2006, 1, 644).
  • the Cu(I) catalyzed 1,3-dipolar cyclization of azides with terminal alkynes to give stable triazoles has been employed for tagging a variety of biomolecules including proteins, nucleic acids, lipids, and saccharides (Chin et al., Science 2003, 301, 964; Gierlich et al., Org. Lett. 2006, 8, 3639; Kho et al., Proc. Natl. Acad. Sci. 2004, 101, 12479; Link et al., Proc. Natl. Acad. Sci. 2006, 103, 10180; Wang et al., J. Am. Chem. Soc. 2003, 125, 3192).
  • the cycloaddition has also been used for activity-based protein profiling (Speers et al., J. Am. Chem. Soc. 2003, 125, 4686), monitoring of enzyme activity, and the chemical synthesis of microarrays and small molecule libraries (Sun et al., Bioconjugate Chem. 2006, 17, 52).
  • Alkyne probes have also been used for cell surface imaging of azide-modified bio-molecules and a particularly attractive approach involves the generation of a fluorescent probe from a non-fluorescent precursor by a [3+2] cycloaddition (Sivakumar et al., Org. Lett. 2004, 6, 4603).
  • reagents including an alkyne fragment, a cleavable linker fragment, and biotin.
  • the alkyne fragment of the reagent can react with various biomolecules containing an azide fragment to give stable triazole adducts.
  • the biotin fragment gives an opportunity to retrieve the tagged compounds by affinity chromatography using immobilized avidin.
  • the cleavable linker allows the release of tagged and captured biomolecules for analysis. For example, released proteins or glycoproteins can be characterized by standard proteomics or glycomics analysis (Too, Expert Rev. Proteomics 2007, 4, 603; Bantscheff et al., Anal. Bioanal. Chem.
  • Compound 21 is an example of the new class of reagent ( Figure 9). It contains a 4-dibenzocyclooctynol fragment for reaction with azides, a disulfide, which can be cleaved with reducing reagents such as dithiothreitol (DTT), and biotin.
  • DTT dithiothreitol
  • Reagents composed of an alkyne, a cleavable linker and biotin can be employed to introduce mass tags into proteins, glycoproteins and other biomolecules containing an azide fragment.
  • reagents such as 21 and 22
  • different mass tags can be introduced to quantify proteins, glycoproteins, glycopeptides, peptides and carbohydrates.
  • the chemical synthesis of 21 and 22 is depicted in Schemes 4 and 5, respectively ( Figures 10 and 11).
  • Various alkyne moieties, cleavable linkers and biotin derivatives are depicted in Figure 12 and alkyne and reactive diene derivatives are depicted in Figure 13.
  • 4-Dibenzocyclooctynol 45 could be prepared by an alternative synthetic route (Scheme 6; Figure 14).
  • known of dibenzosuberenone (41) was treated trimethylsilyl diazomethane in the presence Of BF 3 OEt 2 in CH 2 Cl 2 (20 ml) at -10 0 C to give ⁇ T-Z-Dibenzof ⁇ ejcyclooctatrien-S-one (42) in good yield.
  • the ketone of 42 was reduced with sodium borohydride in a mixture of ethanol and THF to give alcohol 43, which could be converted into 4- dibenzocyclooctynol 45 by bromination of the double bond followed by elimination of the resulting compound 44 by treatment LDA in THF.
  • Compound 45 could be oxidized to the corresponding ketone 46 by employing Dess-Martin reagent.
  • Compounds 45 and 46 were converted into amine containing derivatives
  • Compound 50 was obtained by reaction of 46 with bromoacetic acid in the presence of lithium diisopropylamide in tetrahydrofuran followed by condensation of the resulting acid 48 with tris(ethylene glycol)- 1,8- diamine in DMF in the presence of the coupling reagent HATU and the base DIPEA. Finally, derivative 51 was prepared by oxime formation be reaction of ketone 51 with 7V- ⁇ 2-[2-(2-amino-ethoxy)-ethoxy]-ethyl ⁇ -2-aminooxy-acetamide (84 mg, 0.251 mmol) in the presence of acetic acid and in a mixture of methanol and dichloromethane.
  • a feature of 51 is that the oxime linkage can be cleaved by treatment with aqueous acid to detach the captured compound from the click reagent.
  • the Cu(I) catalyzed 1 ,3-dipolar cycloaddition of azides with terminal alkynes to give stable triazoles has been employed for tagging a variety of biomolecules including proteins, nucleic acids, lipids, and saccharides. This reaction has also been used to modify polymers and nanoscale materials. Potential difficulties to remove Cu(I), which is highly cytotoxic, complicates the use of the 1,3-dipolar cycloaddition for conjugation of compounds or material for biological or medical application.
  • the use of 4-dibenzocyclooctynol instead of a terminal alkyne for cycloadditions with azides should overcome this problem.
  • co- block polymers 83 and 84 were prepared. These materials were employed to form organomicelles in water and it was shown that 4-dibenzocyclooctyne fragment of these materials can be reacted was with azido containing molecules (Figure 2OA, B). It is well known that co-block polymers composed of a polyester and polyethyleneglycol fragment self-assemble in water to form organomicelles These nano-mate ⁇ als have attracted attention as drug delivery devises. De ⁇ vatization of organomicelles with, for example, tissue or tumor targeting moieties may lead to smart drug delivery devises. In addition, modification of organomicelles with fluorescent tags or MRI reagents, such as biotin, will be valuable for imaging purposes (Figure 20C)
  • Copolyme ⁇ zation of polyethylene glycol methyl ether (81) or azide (82) (MW -2000 Da) with caprolactone in the presence of a catalytic amount of SnOct gave copolymers 83 and 84, respectively (Scheme 1 1 , Figure 21 ).
  • the azido fragment of 84 was reduced with t ⁇ phenylphosphine and the amine of the resulting polymer 85 was reacted with 86 and 87 to give dibenzocyclooctyl derivatives 88 and 89, respectively.
  • a mixture of 83 and 88 or 89 (9/1, w/w) dissolved in a small amount of THF were added to water Cryo-TEM showed that organomicelles that have a diameter of approximately 4OA were formed
  • the resulting micelles were incubated with azido-contaimng saccharide 90 and after a reaction time of 24 hours, unreacted saccharide was lemoved by dialysis.
  • the micelles were analyzed for sugar content by hydrolysis with TFA followed by quantification by high pH anion exchange chromatography.
  • Azide-PEG-Z?-PCL was synthesized by a one-pot catiomc ⁇ ng opening polymerization at 130 0 C under a stream of argon adopting a previously reported method for the preparation of PEG- ⁇ -PCL with some modifications. Briefly, a predetermined volume (3.3 mL) of ⁇ -caprolactone monomer was placed in a flask containing a preweighed amount (2.5 g) of azide-PEG-OH 82 under a nitrogen atmosphere. Then a drop of SnOct was added.
  • DIDO-PEO-PCL copolymer (88) To a stirred solution of carbonic acid 5,6-dihydro-l l ,12-didehydro-dibenzo[a,e]cycloocten-5-yl ester 4-nitro-phenyl ester 86 (1 1.6 mg, 0.03 mmol) and copolymer 85 (98 mg, 0.02 mmol) in CH 2 Cl 2 (10 ml) at room temperature was added Et 3 N (0.014 ml, 0.1 mmol). The reaction mixture was stirred overnight at room temperature, after which the solvent was removed under reduced pressure. The residue was purified by size exclusion chromatography (SEC) on LH-20 column (1 : 1 v/v MeOH/CH 2 Cl 2 ) to give the product as yellowish solid (101 mg, 97%).
  • SEC size exclusion chromatography
  • DIDO-PEO-PCL copolymer (89).
  • Nitrones were prepared by a modification of the procedures disclosed in Dicken et al, J. Org. Chem. 1982, 47, 2047-2051 ; and Inouye et al., Bull. Chem. Soc. Jpn. 1983, 56, 3541-3542.
  • N-alkylhydroxylamine hydrochloride (10.0 mmol)
  • glyoxylic acid (0.92 g, 10.0 mmol)
  • sodium bicarbonate (1.68 g, 20.0 mmol) in toluene (20 ml) were stirred at room temperature overnight.
  • the solid was filtered and the filtrate was concentrated to afford the nitrone. This nitrone was then used directly without any purification.
  • nitrones 91-95 were mixed with 4-dibenzocyclooctynol and after a reaction time of 3 minutes to 3.5 hours the corresponding 2,3-dihydro-issoxazole cycloaddition products were isolated in almost a quantitative yield. It can be seen in figure 18 that the chemical nature of the nitrone has a dramatic impact of the reaction rate. In particular electron poor nitrones 93 and 94 react at much faster rates than corresponding azides.

Abstract

1,3-Dipole-functional compounds (e.g., azide functional compounds) can be reacted with certain alkynes in a cyclization reaction to form heterocyclic compounds. Useful alkynes (e.g., strained, cyclic alkynes) and methods of making such alkynes are also disclosed. The reaction of 1,3-dipole-functional compounds with alkynes can be used for a wide variety of applications including the immobilization of biomolecules on a substrate.

Description

ALKYNES AND METHODS OF REACTING ALKYNES WITH 1 ,3-DIPOLE-FUNCTIONAL COMPOUNDS
This application claims the benefit of U.S. Provisional Application Nos. 61/004,021, filed November 21, 2007; 61/007,674, filed December 14, 2007; and 61/137,061, filed July 25, 2008, all of which are hereby incorporated by reference in their entireties.
GOVERNMENT FUNDING
The present invention was made with government support under a grant from the Research Resource Center for Biomedical Complex Carbohydrates of the National Institutes of Health (Grant No. P41-RR-5351). The Government has certain rights in this invention.
BACKGROUND
Bioorthogonal reactions are reactions of materials with each other, wherein each material has limited or substantially no reactivity with functional groups found in vivo. The efficient reaction between an azide and a terminal alkyne, i.e., the most widely studied example of "click" chemistry, is known as a useful example of a bioorthogonal reaction. In particular, the Cu(I) catalyzed 1,3-dipolar cyclization of azides with terminal alkynes to give stable triazoles (e.g., Binder et al., Macromol. Rapid Commun. 2008, 29:952-981) has been employed for tagging a variety of biomolecules including proteins, nucleic acids, lipids, and saccharides. The cycloaddition has also been used for activity-based protein profiling, monitoring of enzyme activity, and the chemical synthesis of microarrays and small molecule libraries. An attractive approach for installing azides into biomolecules is based on metabolic labeling whereby an azide containing biosynthetic precursor is incorporated into biomolecules using the cells' biosynthetic machinery. This approach has been employed for tagging proteins, glycans, and lipids of living systems with a variety of reactive probes. These probes can facilitate the mapping of saccharide-selective glycoproteins and identify glycosylation sites. Alkyne probes have also been used for cell surface imaging of azide-modifϊed bio-molecules and a particularly attractive approach involves the generation of a fluorescent probe from a non-fluorescent precursor by a [3+2] cycloaddition.
Despite the apparent utility of reacting an azide with a terminal alkyne, applications in biological systems using this reaction have been practically limited by factors including the undesirable presence of a copper catalyst. Thus, there is a continuing, unmet need for new bioorthogonal reactions.
SUMMARY
In one aspect, the present invention provides an alkyne, and methods of making an alkyne. In one embodiment, the alkyne is of the formula:
Figure imgf000004_0001
Formula I, wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker). Also provided are blends of certain alkynes with a polymer or a copolymer that can optionally form a copolymer micelle, which can be useful, for example, for controlling the delivery of drugs as described herein.
In another embodiment, the alkyne includes: a cleavable linker fragment including at least two ends; an alkyne fragment attached to a first end of the cleavable linker fragment; and a biotinylated fragment attached to a second end of the cleavable linker fragment. In preferred embodiments, the alkyne fragment includes a strained, cyclic alkyne fragment. In certain embodiments, the alkyne further includes at least one heavy mass isotope. Optionally, the alkyne further includes at least one detectable label such as a fluorescent label.
Alkynes such as those described herein above can be reacted with at least one 1,3-dipole-functional compound (e.g., an azide-functional compound, a nitrile oxide-functional compound, a nitrone-functional compound, an azoxy- functional compound, and/or an acyl diazo-functional compound) in a cyclization reaction to form a heterocyclic compound, preferably in the substantial absence of added catalyst (e.g., Cu(I)). Optionally, the reaction can take place within or on the surface of a living cell. In certain embodiments, the at least one 1,3-dipole-functional compound includes a 1 ,3-dipole-functional biomolecule such as a peptide, protein, glycoprotein, nucleic acid, lipid, saccharide, oligosaccharide, and/or polysaccharide. Optionally, the 1 ,3-dipole- functional biomolecule includes a detectable label such as an affinity label. The heterocyclic compounds formed by the alkyne with the at least one 1,3-dipole- functional compound are also disclosed herein. In certain embodiments, the reaction between the alkyne and the at least one 1,3-dipole-functional compound can take place within or on the surface of a living cell.
For embodiments in which the heterocyclic compound includes a biotinylated fragment, the heterocyclic compound can be bound to a compound that binds biotin, such as avidin and/or streptavidin.
In another aspect, the present invention provides a substrate having an alkyne as described herein on the surface thereof. The substrate can be in the form of a resin, a gel, nanoparticles, or combinations thereof. Optionally, the substrate is a three-dimensional matrix. In preferred embodiments, the X group of an alkyne of Formula I represents a point of attachment to the surface of the substrate. Such substrates can be useful for immobilizing biomolecules such as peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, and/or polysaccharides. Articles including an immobilized biomolecule, such as a protein immobilized on a three-dimensional matrix, are also disclosed herein.
The compositions and methods disclosed herein can offer advantages over bioorthogonal reactions known in the art. See, for example, Baskin et al., QSAR Comb. ScL 2007, 26: 1211-1219. For example, alkynes of Formula I as described herein (e.g., wherein X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group) surprisingly have been found to have higher reactivity towards 1,3-dipole-functional compounds than other strained, cyclic alkynes (e.g., wherein X represents CH?). See, for example, Codelli, et al., J. Am. Chem. Soc. 2008, 130:1 1486-1 1493; Johnson et al., Chem. Commun. 2008, 3064-3066; Sletten et al., Organic Letters 2008, 10:3097-3099; and Laughlin et al., Science 2008, 320:664-667. Further, convenient methods having the flexibility to prepare a wide variety of alkynes of Formula I are disclosed herein. In addition, alkynes of Formula I have the capability of reacting not only with azides, but also a variety of other 1,3-dipole-functional compounds. Definitions:
The term "comprises" and variations thereof do not have a limiting meaning where these terms appear in the description and claims. As used herein, "a," "an," "the," "at least one," and "one or more" are used interchangeably.
As used herein, the term "or'" is generally employed in the sense as including "and/or" unless the context of the usage clearly indicates otherwise.
Also herein, the recitations of numerical ranges by endpoints include all numbers subsumed within that range (e.g., 1 to 5 includes 1, 1.5, 2, 2.75, 3, 3.80, 4, 5, etc.).
The above summary is not intended to describe each disclosed embodiment or every implementation of the present invention. The description that follows more particularly exemplifies illustrative embodiments. In several places throughout the application, guidance is provided through lists of examples, which examples can be used in various combinations. In each instance, the recited list serves only as a representative group and should not be interpreted as an exclusive list.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 illustrates exemplary reagents for labeling azide-functional biomolecules.
Figure 2 illustrates Scheme 1 : exemplary reagents and conditions: a) TBSCl, pyridine; b) Br2, CHCl3; c) LDA, tetrahydrofuran; d) 4-nitrophenyl chloroformate, pyridine, CH2Cl?; e) N,N-dimethylformamide (DMF), triethylamine (TEA). LDA = lithium diisopropylamide, TBS = tert- butyldimethylsilyl.
Figure 3 illustrates Scheme 2: exemplary reagents and conditions: a) compound 3 in methanol.
Figure 4 illustrates exemplary metal-free cycloadditions of compound 3 with azide-functional amino acid and saccharides. Boc = tert-butoxycarbonyl, TDS = thexyldimethylsilyl.
Figure 5 illustrates Scheme 3: exemplary reaction conditions: a) 1, triethylamine, DMF, room temperature, 78%; b) 20% trifluoroacetic acid (TFA), room temperature, 95%; c) 2, TEA, DMF, room temperature, 68%. Figure 6 illustrates embodiments of cell-surface labeling with compounds
2 and 9. Jurkat cells grown for three days in the absence or presence of Ac4ManNAz (25 micromolar) were incubated a) with compounds 2 and 9 (30 micromolar) for 0-180 minutes or b) with compounds 2 and 9 (0-100 micromolar) for 1 hour at room temperature. Next, the cells were incubated with avidin-FITC for 15 minutes at 4°C, after which cell lysates were assessed for fluorescence intensity. Samples are indicated as follows: blank cells incubated with 2 (o) or 9 (□), and Ac,ManNAz cells incubated with 2 (•) or 9 (■).
Figure 7 illustrates an embodiment of toxicity assessment of cell labeling procedure and cycloaddition reaction with compound 9. Jurkat cells grown for 3 days in the absence (a) or presence (b) of Ac4ManNAz (25 micromolar) were incubated with compound 9 (0 - 100 micromolar) for 1 hour at room temperature. The cells were washed three times and then incubated with avidin conjugated with fluorescein for 15 minutes at 4°C, after which cells were washed three times. Cell viability was assessed at different points during the procedure with trypan blue exclusion; after incubation with 9 (black), after avidin-FITC incubation (grey), and after complete procedure (white). Treatment with Cu1Cl (ImM) under the same conditions led to approximately 98% cell death for both the blank and the Ac4MaIiNAz treated cells.
Figure 8 illustrates fluorescence images for embodiments of cells labeled with compound 9 and avidin-Alexa Fluor 488. CHO cells grown for 3 days in the absence (d-f) or presence (a-c) of Ac4ManNAz (100 micromolar) were incubated with compound 9 (30 micromolar) for 1 hour at 4°C (a, d) or room temperature (b, c, e, f). Next, cells were incubated with avidin-Alexa Fluor 488 for 15 minutes at 4°C and, after washing, fixing, and staining for the nucleus with far-red-fluorescent dye TO-PRO, imaged (a, b, d, e) or after washing incubated for 1 hour at 37°C before fixing, nucleus staining, and imaging (c, f). Merged indicates that the images of cells labeled with Alexa Fluor (488 nanometers (nm)) and TO-PRO-3 iodide (633 nm) are merged.
Figure 9 illustrates exemplary compounds comprising an alkyne fragment, a cleavable linker fragment, and a biotinylated fragment.
Figure 10 illustrates Scheme 4: exemplary reaction conditions: a) DMF, 800C, 70%; b) potassium thioacetate (KSAc), DMF, 600C, 90%; c) NH2NH2, ethanol (EtOH), refluxing, 95%; d) N,N-diisopropylethylamine (DIPEA), DMF, 00C, 56%; e) DIPEA, DMF, room temperature, 85%.
Figure 1 1 illustrates Scheme 5: exemplary reaction conditions: a) p- toluenesulfonic acid (TsOH), room temperature, 81%; b) DMF, 800C, 86%; c) 0. IN HCl, EtOH, room temperature, 90%; d) 9, NaH, DMF, 00C, 88%; e) 0. IN HCl, EtOH, room temperature, 88%; f) CCl4, PPh3, dichloromethane (DCM), room temperature, 96%; g) KSAc, DMF, 600C, 90%; h) NH2NH2, EtOH, refluxing, 95%; then (Boc)2O, TEA, EtOH, 91%; i) 20% TFA, DCM, room temperature, 95%; j) DIPEA, DMF, 00C; then (Boc)2O, TEA, EtOH, 60% over two steps; k) 20% TFA, DCM, room temperature, then 8 DIPEA, DMF, room temperature, 80% over two steps.
Figure 12 illustrates exemplary cleavable linkers. Figure 13 illustrates exemplary alkynes and a reactive diene.
Figure 14 illustrates scheme 6: exemplary reaction conditions: a) TMSCH2N2, BF3OEt2, DCM, -1O0C, 3 hours, 71%; b) NaBH4, 1 : 1 EtOH/THF, room temperature, 7 hours, 100%; C) Br2, CHCl3, room temperature, 0.5 hour, 58%; d) LDA, THF, 0.5 hour, 57%; e) Dess-Martin reagent, DCM, 0.5 h; f) 4- nitrophenyl chloroformate, pyridine, DCM, 18 hours, 92%; g) tris(ethylene glycol)- 1 ,8-diamine, TEA, DCM, room temperature, 3 hours, 80%; h) bromoacetic acid, NaH, THF, 22%; i) tris(ethylene glycol)- 1,8-diamine, HATU coupling reagent, DIPEA, DMF, room temperature, 2 hours, 75%; j) 7V-{2-[2-(2- amino-ethoxy)-ethoxy]-ethyl}-2-aminooxy-acetamide, AcOH, 1 :1 DCM/MeOH, 63%.
Figure 15 illustrates compounds 61-68 and second order constants.
Figure 16 illsutrates scheme 7: exemplary reagents and conditions: a) LiAlH4, AlCl3, Et2O, O0C, 61%; b) Br2, CHCl3, O0C, 58%; c) potassium t- butoxide (Y-BuOK), THF, room temperature, 25%.
Figure 17 illsutrates scheme 8: exemplary reagents and conditions: a) TEA, CH2Cl?, room temperature, 77%.
Figure 18 illustrates scheme 9: exemplary reagents and conditions: a) NaH, benzyl bromide, DMF, room temperature, 59%; b) acetic anhydride (Ac2O), pyridine, room temperature, 81%.
Figure 19 illustrates scheme 10: exemplary reagents and conditions: a) LDA, Et3SiCl, THF, room temperature, 85%; b) SELECTFLUOR fluorinating reagent, DMF, room temperature, 66% c) LDA, Et3SiCl, THF, room temperature, 79%; d) SELECTFLUOR fluorinating reagent, DMF, room temperature, 51%; e) NaBH4, EtOH, room temperature, 78%; f) Br2, CHCl3, O0C, 46%; g) c) r-BuOK, THF, room temperature, 49%.
Figure 20 illustrates the use of copolymer micelles for drug delivery. A) Polyester and polyethyleneglycol groups self-assembled in water. B) Functionalized copolymer micelles as drug delivery devices. C) Oxime- modified alkyne derivatives of copolymer micelles.
Figure 21 illustrates the preparation of macromolecules with 4- dibenzocyclooctyne functionality. Figure 22 illustrates cycloadditions of 4-dibenzocyclooctynol with various nitrones. Compounds were mixed at 1 : 1 molar ratio at a final concentration of 6mM and reacted for a time indicated.
DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
Alkynes such as those described herein can be reacted with at least one 1,3-dipole-functional compound in a cyclization reaction to form a heterocyclic compound. In preferred embodiments, the reaction can be earned out in the substantial absence of added catalyst (e.g., Cu(I)). Exemplary 1,3-dipole- functional compounds include, but are not limited to, azide-functional compounds, nitrile oxide-functional compounds, nitrone-functional compounds, azoxy-functional compounds, and/or acyl diazo-functional compounds.
Exemplary alkynes include alkynes of the formula:
Figure imgf000010_0001
Formula I,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group (and preferably a Cl-Cl O organic moiety); each R~ is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group (and preferably a Cl-ClO organic moiety); X represents C=O, C=N-OR3, C=N- NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group (and in some embodiments an organic moiety). In preferred embodiments, each R1 represents hydrogen and/or each R2 represents hydrogen. Optionally, R3 includes a covalently bound organic dye (e.g., a fluorescent dye).
As used herein, the term "organic group" is used for the purpose of this invention to mean a hydrocarbon group that is classified as an aliphatic group, cyclic group, or combination of aliphatic and cyclic groups (e.g., alkaryl and aralkyl groups). In the context of the present invention, suitable organic groups for compounds of this invention are those that do not interfere with the reaction of an alkyne with a 1,3-dipole-functional compound to form a heterocyclic compound. In the context of the present invention, the term "aliphatic group" means a saturated or unsaturated linear or branched hydrocarbon group. This term is used to encompass alkyl, alkenyl, and alkynyl groups, for example. The term "alkyl group" means a saturated linear or branched monovalent hydrocarbon group including, for example, methyl, ethyl, π-propyl, isopropyl, tert-butyl, amyl, heptyl, and the like. The term "alkenyl group" means an unsaturated, linear or branched monovalent hydrocarbon group with one or more olefmically unsaturated groups (i.e., carbon-carbon double bonds), such as a vinyl group. The term "alkynyl group" means an unsaturated, linear or branched monovalent hydrocarbon group with one or more carbon-carbon triple bonds. The term "cyclic group" means a closed ring hydrocarbon group that is classified as an alicyclic group, aromatic group, or heterocyclic group. The term "alicyclic group" means a cyclic hydrocarbon group having properties resembling those of aliphatic groups. The term "aromatic group" or "aryl group" means a mono- or polyiiuclear aromatic hydrocarbon group. The term "heterocyclic group" means a closed ring hydrocarbon in which one or more of the atoms in the ring is an element other than carbon (e.g., nitrogen, oxygen, sulfur, etc.).
As a means of simplifying the discussion and the recitation of certain terminology used throughout this application, the terms "group" and "moiety" are used to differentiate between chemical species that allow for substitution or that may be substituted and those that do not so allow for substitution or may not be so substituted. Thus, when the term "group" is used to describe a chemical substituent, the described chemical material includes the unsubstituted group and that group with nonperoxidic O, N, S, Si, or F atoms, for example, in the chain as well as carbonyl groups or other conventional substituents. Where the term "moiety" is used to describe a chemical compound or substituent, only an unsubstituted chemical material is intended to be included. For example, the phrase "alkyl group" is intended to include not only pure open chain saturated hydrocarbon alkyl substituents, such as methyl, ethyl, propyl, tert-buty\, and the like, but also alkyl substituents bearing further substituents known in the art, such as hydroxy, alkoxy, alkylsulfonyl, halogen atoms, cyano, nitro, amino, carboxyl, etc. Thus, "alkyl group" includes ether groups, haloalkyls, nitroalkyls, carboxyalkyls, hydroxyalkyls, sulfoalkyls, etc. On the other hand, the phrase "alkyl moiety" is limited to the inclusion of only pure open chain saturated hydrocarbon alkyl substituents, such as methyl, ethyl, propyl, tert-butyl, and the like.
Alkynes of Foπnula I are typically strained, cyclic alkynes. Surprisingly it has been found that alkynes of Formula I as described herein (e.g., wherein X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group) have been found to have higher reactivity towards 1,3-dipole-functional compounds than other strained, cyclic alkynes (e.g., wherein X represents CH2).
In certain embodiments of alkynes of Formula I, X can represent C=N- OR3 wherein R3 is an organic group. For example, R3 can have the formula -(CH2)aC(O)Y, wherein: a is 1-3; Y represents OH or NHR5; and R5 represents hydrogen or a biotinylation product of a primary amine-containing organic group. The primary amine-containing group can, for example, be of the formula
-(CH2CH2θ)b(CH2)c-Ld-(CH2CH2θ)e(CH2)ιNH2 and/or -(CD2CD2θ)b(CD2)c-Ld- (CD2CD2O)6(CD2VNH2, wherein b = O to 1 OO (e.g., 10 to 100); c = O to 1 OO (and preferably 1 to 10); d = 0 to 100 (and preferably 1 to 10); e = 0 to 100 (e.g., 10 to 100); f = 0 to 100 (and preferably 1 to 10); and L is an optional cleavable linker (e.g., a disulfide).
In certain embodiments of alkynes of Formula I, X can represent CHOR3, wherein R3 is selected from the group consisting of an alkyl group, an aryl group, an alkaryl group, and an aralkyl group. For example, R3 can have the formula -C(O)Z, wherein: Z represents an alkyl group, OR6, or NHR7; and R6 and R7 are each independently selected from the group consisting of an alkyl group, an aryl group, an alkaryl group, and an aralkyl group. In certain embodiments, R7 can be a biotinylation product of a primary amine-containing organic group. The primary amine-containing group can, for example, be of the formula -(CH2CH2OMCH2)C-Ld-(CH2CH2COe(CH2)(NH2 and/or -(CD2CD2O)b(CD2)c-Ld-(CD2CD2O)e(CD2)fNH2, wherein b = 0 to 100 (e.g., 10 to 100); c = 0 to 100 (and preferably 1 to 10); d = 0 to 100 (and preferably 1 to 10); e = 0 to 100 (e.g., 10 to 100); f = 0 to 100 (and preferably 1 to 10); and L is an optional cleavable linker (e.g., a disulfide).
An exemplary alkyne of Formula I is the species in which X represents C=O, an alkyne of the formula:
Figure imgf000013_0001
Formula IV.
Another exemplary alkyne of Formula I is the species in which X represents CHOH, an alkyne of the formula:
Figure imgf000013_0002
Another exemplary alkyne of Formula I is the species in which X represents CHNH2, an alkyne of the formula:
Figure imgf000014_0001
Formula VI.
Another exemplary alkyne of Formula I is the species in which X represents C=N-OR3, an alkyne of the formula:
Figure imgf000014_0002
Formula VII, wherein R3 represents hydrogen or an organic group (and in some embodiments an organic moiety). Additional exemplary alkynes include alkynes that have: a cleavable linker fragment including at least two ends; an alkyne fragment attached to a first end of the cleavable linker fragment; and a biotinylated fragment attached to a second end of the cleavable linker fragment. In certain embodiments, the alkyne fragment includes a strained, cyclic alkyne fragment. In certain embodiments, the alkyne further includes at least one heavy mass isotope. Optionally, the alkyne further includes at least one detectable label (e.g., a fluorescent label).
In certain embodiments of alkynes of Formula I, X can represent a polymeric or a copolymeric group. For embodiments in which X represents a copolymeric group, the copolymeric group can include a hydrophilic segment and a hydrophobic segment. For example, the copolymeric group can include a fragment of the formula -[CH2CH2O]n-[C(O)(CH2)5O]m-H, wherein n = 0 to 100 (e.g., 10 to 100) and m = 0 to 100 (e.g., 10 to 100). Surfaces on which drops of water or aqueous solutions exhibit a contact angle of less than 90 degrees are commonly referred to as "hydrophilic." The contact angle of a hydrophobic material with water is typically greater than 90 degrees. Exemplary methods of making alkynes of Formula I are also disclosed herein. In one embodiment, the method includes: brominating an alkene of the formula:
Figure imgf000015_0001
Formula XIV
to provide a dibromide of the formula:
Figure imgf000015_0002
Formula XV;
and dehydrobrominating the dibromide of Formula XV to provide the alkyne of the formula:
Figure imgf000015_0003
Formula I,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R" is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR ; and each R and R4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker). A wide variety of 1,3-dipole-functional compounds can be used to react with the alkynes disclosed herein. As used herein, a "1,3-dipole-functional compound" is meant to include compounds having at least one 1,3-dipole group attached thereto. As used herein, a "1,3-dipole group" is intended to refer to a group having a three-atom pi-electron system containing 4 electrons delocalized over the three atoms. Exemplary 1,3-dipole groups include, but are not limited to, azides, nitrile oxides, nitrones, azoxy groups, and acyl diazo groups. In certain embodiments, the 1 ,3-dipole-functional compound can be a biomolecule having at least one 1,3-dipole group attached thereto. Optionally, the at least one 1,3-dipole-functional compound can include a detectable label (e.g., an immunoassay or affinity label).
One or more 1,3-dipole-functional compounds (e.g., azide-functional compounds, nitrile oxide-functional compounds, nitrone-functional compounds, azoxy-functional compounds, and/or acyl diazo-functional compounds) can be combined with an alkyne as described herein under conditions effective to react in a cyclization reaction and form a heterocyclic compound. Preferably, conditions effective to form the heterocyclic compound can include the substantial absence of added catalyst. Conditions effective to form the heterocyclic compound can also include the presence or absence of a wide variety of solvents including, but not limited to, aqueous (e.g., water) and non- aqueous solvents; protic and aprotic solvents; polar and non-polar solvents; and combinations thereof. The heterocyclic compound can be formed over a wide temperature range, with a temperature range of 00C to 400C (and in some embodiments 230C to 37°C) being particularly useful when biomolecules are involved. Conveniently, reaction times can be less than one day, and sometimes one hour or even less.
In certain embodiments, the cyclization reaction between the one or more 1,3-dipole-functional compounds and the alkyne can take place within or on the surface of a living cell. Such reactions can take place in vivo or ex vivo. As used herein, the term "in vivo" refers to a reaction that is within the body of a subject. As used herein, the term "ex vivo" refers to a reaction in tissue (e.g., cells) that has been removed, for example, isolated, from the body of a subject. Tissue that can be removed includes, for example, primary cells (e.g., cells that have recently been removed from a subject and are capable of limited growth or maintenance in tissue culture medium), cultured cells (e.g., cells that are capable of extended growth or maintenance in tissue culture medium), and combinations thereof.
An exemplary embodiment of a 1, 3 -dipole- functional compound is an azide-functional compound of the formula R8-N3 (e.g., represented by the valence structure R8-~N-N=N+), wherein R8 represents and organic group (e.g., a biomolecule). Optionally, R8 can include a detectable label (e.g., an affinity label).
The cyclization reaction of an azide-functional compound of the formula R -N3 with an exemplary alkyne of Formula I can result in one or more heterocyclic compounds of the formulas:
Figure imgf000017_0001
wherein: each R is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; each R3 and R4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker); and R represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
Another exemplary embodiment of a 1, 3 -dipole- functional compound is a nitrile oxide-functional compound of the formula R -CNO (e.g., represented by the valence structure R - C=N-O"), wherein R represents and organic group (e.g., a biomolecule). Optionally, R8 can include a detectable label (e.g., an affinity label).
The cyclization reaction of a nitrile oxide-functional compound of the formula R8-CNO with an exemplary alkyne of Formula I can result in one or more heterocyclic compounds of the formulas:
Figure imgf000018_0001
Formula VIII Formula IX,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-Cl O organic group; each R" is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; each R3 and R4 independently represents hydrogen or an organic group (e.g., which can include a cleavable linker); and R represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker). Another exemplary embodiment of a 1,3 -dipole- functional compound is a nitrone-functional compound of the formula (R l(k )2CN(R , I Ox )O (e.g., represented by the valence structure (Rl0)2C=+N(Rl 0)-O"), wherein each Ri 0 independently represents hydrogen or an organic group, with the proviso that at least one R 10 represents an organic group (e.g., a biomolecule). Optionally, at least one R 10 can include a detectable label (e.g., an affinity label). The cyclization reaction of a nitrone- functional compound of the formula (R10)2CN(R10)O with an exemplary alkyne of Formula I can result in one or more heterocyclic compounds of the formulas:
Figure imgf000019_0001
Formula X Formula XI,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R" is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3, R4, and R10 independently represents hydrogen or an organic group, with the proviso that at least one R10 represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
Another exemplary embodiment of a 1, 3 -dipole- functional compound is an azoxy-functional compound of the formula Rl0-NN(Rl 0)O (e.g., represented by the valence structure R IO -N= N(R , 10 )-O"), wherein each R IO independently represents hydrogen or an organic group, with the proviso that at least one R IO represents an organic group (e.g., a biomolecule). Optionally, at least one R1 can include a detectable label (e.g., an affinity label).
The cyclization reaction of an azoxy-functional compound of the formula R10-NN(R10)O with an exemplary alkyne of Formula I can result in one or more heterocyclic compounds of the formulas:
Figure imgf000020_0001
Formula XII Formula XIII,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3, R4, and R10 independently represents hydrogen or an organic group, with the proviso that at least one R10 represents an organic group (e.g., which can include a biomolecule and optionally a cleavable linker).
For embodiments in which the heterocyclic compound formed from the cyclization reaction between the alkyne and the one or more 1 ,3-dipole- functional compounds includes a detectable label, the heterocyclic compound can be detected using the detectable label. For example, for embodiments in which the detectable label is an affinity label, affinity binding (e.g., affinity chromatography) can be used to detect the heterocyclic compound.
In addition, for embodiments in which the heterocyclic compound formed from the cyclization reaction between the alkyne and the one or more 1 ,3 -dipole- functional compounds includes a biotinylated fragment, the heterocyclic compound can be bound by contacting the heterocyclic compound with a compound that binds biotin (e.g., avidin and/or streptavidin). Further, the bound heterocyclic compound can be detected by methods described herein.
Cyclization reactions between alkynes as disclosed herein and 1,3-dipole- functional compounds can be used for a wide variety of applications. For example, an alkyne as disclosed herein can be attached to the surface of a substrate. In certain embodiments, the X group of the alkyne represents a point of attachment to the surface of the substrate. One of skill in the art will recognize that the X group can advantageously be selected to include functionality (e.g., biotin, activated esters, activated carbonates, and the like) to enable attachment of the alkyne to a functional substrate (e.g., amine functionality, thiol functionality, and the like) through a wide variety of reactions.
Substrates having an alkyne attached to the surface thereof can be reacted with 1,3-dipole-functional compounds to form heterocyclic compounds, effectively chemically bonding the 1 ,3-dipole-functional compounds to the substrate. Such substrates can be, for example, in the form of resins, gels, nanoparticles (e.g., including magnetic nanoparticles), or combinations thereof. In certain embodiments, such substrates can be in the form of microarrays or even three-dimensional matrices or scaffolds. Exemplary three-dimensional matrices include, but are not limited to, those available under the trade designations ALGIMATRIX 3D Culture system, GELTRIX matrix, and GIBCO three-dimensional scaffolds, all available from Invitrogen (Carlsbad, CA). Such three- dimensional matrices can be particularly useful for applications including cell cultures.
1 ,3-Dipole-functional biomolecules (e.g., 1 ,3-dipole-functional peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, and/or polysaccharides) can be immobilized on, and preferably covalently attached to, a substrate surface by contacting the 1 ,3-dipole-functional biomolecules with a substrate having an alkyne attached to the surface thereof under conditions effective for a cyclization reaction to form a heterocyclic compound. Preferably, conditions effective to form the heterocyclic compound can include the substantial absence of added catalyst. Conditions effective to form the heterocyclic compound can also include the presence or absence of a wide variety of solvents including, but not limited to, aqueous (e.g., water and other biological fluids) and non-aqueous solvents; protic and aprotic solvents; polar and non-polar solvents; and combinations thereof. The heterocyclic compound can be formed over a wide temperature range, with a temperature range of 00C to 400C (and in some embodiments 230C to 37°C) being particularly useful. Conveniently, reaction times can be less than one day, and sometimes one hour or even less.
For example, when the substrate is in the form of a three-dimensional matrix and the 1 ,3-dipole-functional biomolecule is a 1 ,3-dipole-functional protein (e.g., an azide-functional protein), the cyclization reaction can result in an article having a protein immobilized on a three-dimensional matrix. Such matrices can have a wide variety of uses including, but not limited to, separating and/or immobilizing cell lines. Particularly useful proteins for these applications include, but are not limited to, collagen, fibronectin, gelatin, laminin, vitronectin, and/or other proteins commonly used for cell plating.
For another example, cyclization reactions between 1,3-dipole-functional compounds and alkynes of Formula I in which X represents a polymeric or a copolymeric group can be used, for example, for controlling the delivery of drugs as described herein below. For example, alkynes of Formula I in which X represents a copolymeric group including a hydrophilic segment and a hydrophobic segment can be blended with a polymer or a copolymer. Further, when an alkyne of Formula I in which X represents a copolymeric group including a hydrophilic segment and a hydrophobic segment is blended with a copolymer having a hydrophilic segment and a hydrophobic segment, a copolymer micelle can be formed. Particularly useful copolymers having a hydrophilic segment and a hydrophobic segment include those of the formula R9O-[CH2CH2O]p-[C(O)(CH2)5θ]o-H, wherein R9 represents an alkyl group (e.g., methyl), p = 0 to 100 (e.g., 1 to 100), and o = 0 to 100 (e.g., 1 to 100).
The copolymer micelles that include an alkyne of Formula 1 as described herein above can advantageously be used to control the delivery of drugs. For example, a copolymer micelle that includes an alkyne of Formula 1 can be combined with at least one 1,3-dipole-functional drug and allowed to react under conditions effective to form a heterocyclic compound and attach the drug to the copolymer micelle. See, for example, Nishiyama et al., Adv. Polym. Sci. 2006, 193:67-101 ; Gaucher et al., J. Control. Release 2005, 109:169-188; Choi et al., J. Dispersion Sci. Tech. 2003, 24:475-487; Lavasanifar et al., Adv. Drug Delivery Rev. 2002, 54: 169-190; and Rosier et al., Adv. Drug Delivery Rev. 2001 , 53:95-108.
Further, because it does not require a toxic catalyst such as copper, the novel cycloaddition reaction provided by the invention can be used for labeling of living cells. For example, cells can first be metabolically labeled with an azide- functional precursor to produce azide-functional biomolecules (also referred to as bioconjugates) such as azide-functional glycoproteins (also referred to as glycoconjugates). The cells can then be contacted with an alkyne of Formula I, either in solution or on a substrate as discussed above, under conditions to permit labeling (via the cycloaddition reaction) of the azide- functional biomolecules at the surface of the cell. The resulting triazole conjugate can be detected at the cell surface, or it can be endocytosed by the cell and detected inside the cell.
Alkynes of Formula I can also have utility for imaging applications including, for example, as reagents for magnetic resonance imaging (MRI). For another example, alkynes of Formula I can contain a fluorescent tag. Alkynes of Formula I can also be useful in qualitative or quantitative proteomics and glycomics applications utilizing mass spectrometry. The alkyne of Formula I can be selected to contain one or more heavy mass isotopes, such as deuterium, 13C, 15N, 35S and the like, and then can be used to label and/or immobilize azide- functional biomolecules as described herein.
Alkynes of Formula I can also have utility for applications such as vaccines. For example, alkynes of Formula I can be reacted with an azide- functional protein (e.g., an azide-functional carbohydrate, an azide-functional peptide, and/or an azide-functional glycopeptide), and the resulting triazole conjugate can be used as a carrier protein for the vaccine.
The present invention is illustrated by the following examples. It is to be understood that the particular examples, materials, amounts, and procedures are to be interpreted broadly in accordance with the scope and spirit of the invention as set forth herein. EXAMPLES
Example 1
Visualizing Metabolically Labeled Glycoconjugates of Living Cells by
Copper-Free and Fast Huisgen Cycloadditions
Azides, which are extremely rare in biological systems, are emerging as attractive chemical handles for bioconjugation (KoIb and Sharpless, Drug Discovery Today 2003, 8:1128-1 137; Dedola et al., Org. Biomol. Chem. 2007 5:1006-1017; Moses and Moorhouse, Chem. Soc. Rev. 2007, 36: 1249-1262; Nandivada et al., Adv. Mater. 2007, 19:2197-2208; Wu and Fokin, Aldrichimica Acta 2007, 40:7-17). In particular, the Cu'-catalyzed 1 ,3-dipolar cycloaddition of azides with teπninal alkynes to give stable triazoles (Rostovtsev et al., Angew. Chem. 2002, 1 14:2708-271 1 ; Rostovtsev et al., Angew. Chem. Int. Ed. 2002, 41 :2596-2599; Tornoe et al., J. Org. Chem. 2002, 67:3057-3064) has been employed for the tagging of a variety of biomolecules, (Chin et al., Science 2003, 301 :964 -967; Wang et al., J. Am. Chem. Soc. 2003, 125:3192-3193; Kho et al., Proc. Natl. Acad. ScL USA 2004, 101 :12479-12484; Gierlich et al., Org. Lett. 2006, 8:3639-3642; Link et al., Proc. Natl. Acad. ScL USA 2006,
103:10180- 10185) activity-based protein profiling (Speers et al., J. Am. Chem. Soc. 2003, 125:4686-4687), and the chemical synthesis of microarrays and small-molecule libraries (Sun et al., Bioconjugate Chem. 2006, 17:52-57).
An attractive approach for installing azides into biomolecules is based on metabolic labeling, whereby an azide-containing biosynthetic precursor is incorporated into biomolecules by using the cells' biosynthetic machinery (Prescher and Bertozzi, Nat. Chem. Biol. 2005, 1 :13-21). This approach has been employed for tagging proteins, glycans, and lipids of living systems with a variety of reactive probes. These probes can facilitate the mapping of saccharide- selective glycoproteins and identify glycosylation sites (Hanson et al., J. Am.
Chem. Soc. 2007, 129:7266-7267). Alkyne probes have also been used for cell- surface imaging of azide-modified biomolecules, and a particularly attractive approach involves the generation of a fluorescent probe from a nonfluorescent precursor by a [3+2] cycloaddition (Sivakumar et al., Org. Lett. 2004, 6:4603- 4606).
The cellular toxicity of the Cu1 catalyst has precluded applications wherein cells must remain viable (Link and Tirrel, J. Am. Chem. Soc. 2003, 125: 1 1 164- 1 1 165), and hence there is a great need for the development of Cu1- free [3+2] cycloadditions (Turner et al., J. Am. Chem. Soc. 1973, 95:790 -792; Agard et al., J. Am. Chem. Soc. 2004, 126: 15046-15047; vanBerkel et al., Chem- BioChem 2007, 8:1504-1508). In this respect, alkynes can be activated by ring strain, and, for example, constraining an alkyne within an eight-membered ring creates 18 kcalmol"1 of strain, much of which is released in the transition state upon [3+2] cycloaddition with an azide (Turner et al., J. Am. Chem. Soc. 1973, 95:790 -792; Agard et al., J. Am. Chem. Soc. 2004, 126:15046-15047). As a result, cyclooctynes such as 1 react with azides at room temperature without the need for a catalyst (Figure 1 ). The strain-promoted cycloaddition has been used to label biomolecules without observable cytotoxicity (Agard et al., J. Am. Chem. Soc. 2004, 126: 15046-15047). The scope of the approach has, however, been limited because of the slow rate of reaction (Agard et al., ACS Chem. Biol. 2006, 1 :644-648). Appending electron-withdrawing groups to the octyne ring can increase the rate of strain-promoted cycloadditions; however, currently
Staudinger ligation with phosphine 2 offers the most attractive reagent for cell- surface labeling with azides.
It was envisaged that 4-dibenzocyclooctynols such as compound 3 would be ideal for labeling living cells with azides because the aromatic rings are expected to impose additional ring strain and conjugate with the alkyne, thereby increasing the reactivity of the alkyne in metal-free [2+3] cycloadditions with azides. The compound should, however, have excellent stability because the ortho hydrogen atoms of the aromatic rings shield the alkyne from nucleophilic attack. Furthermore, the hydroxy group of 3 provides a handle for the incorporation of tags such as fluorescent probes and biotin.
Compound 3 could be prepared easily from known (Jung et al., J. Org. Chem. 1978, 43:3698-3701 ; Jung and Miller, J. Am. Chem. Soc. 1981 , 103:1984- 1992) 3-hydroxy-l ,2:5,6-dibenzocycloocta-l,5,7-triene (4 ) by protection of the hydroxy group as a TBS ether to give 5, which was brominated to provide dibromide 6 in a yield of 60% (Scheme 1 ; Figure T). The TBS protecting group was lost during the latter transformation, but the bromination was low yielding when performed on alcohol 4. Dehydrobromination of 6 by treatment with LDA in THF at 00C (Seitz et al, Angew. Chem. 1969, 81 :427-428; Seitz et al., Angew. Chem. Int. Ed. Engl. 1969, 8:447-448) gave the target cyclooctyne 3 in a yield of 45 %.
Compound 3 has an excellent, long shelf life and after treatment did not react with nucleophiles such as thiols and amines. However, upon exposure to azides a fast reaction took place and gave the corresponding triazoles in high yield. For example, triazoles 10-13 were obtained in quantitative yields as mixtures of regioisomers by reaction of the corresponding azido-containing sugar and amino acid derivatives with 3 in methanol for 30 minutes (Scheme 2; Figure 3 and Figure 4). The progress of the reaction of 3 with benzyl azide in methanol and in a mixture of water/acetonitrile (1 :4 v/v) was monitored by 1H NMR spectroscopy by integration of the benzylic proton signals, and second- order rate constants of 0.17 and 2.3 m"'s"', respectively, were determined. The rate constant of the reaction with 3 in acetonitrile/water is approximately three orders of magnitude greater than that with cyclooctyne 1.
Having established the superior reactivity of 3, we focused our attention on the preparation of a derivative of 4-dibenzocyclooctynol (9; Scheme 1 ; Figure 2), which is modified with biotin. Such a reagent should make it possible to visualize biomolecules after metabolically labeling cells with an azido- containing biosynthetic precursor, followed by cycloaddition with 9 and treatment with avidin modified with a fluorescence probe. Alternatively, biotinylation of glycoconjugates with 9 should make it possible to isolate these derivatives for glycocomics studies using avidin immobilized on a solid support. Compound 9 could easily be prepared by a two-step reaction involving treatment of 3 with 4-nitrophenyl chloroformate to give activated intermediate 7, followed by immediate reaction with 8. 4-dibenzocyclooctynol (9) may also be functionalized with a fluorescent tag to yield a fluorescent derivative (Scheme 3; Figure 5).
Next, Jurkat cells were cultured in the presence of 25 micromolar N- azidoacetylmannosamine (Ac4ManNAz) for three days to metabolically introduce /V-azidoacetylsialic acid (SiaNAz) moieties into glycoproteins
(Luchansky and Bertozzi, Chem-BioChem 2004, 5: 1706-1709). As a negative control, Jurkat cells were employed that were grown in the absence of Ac4MaIiNAz. The cells were exposed to a 30 micromolar solution of compound 9 for various time periods, and after washing, the cells were stained with avidin- fluorescein isothiocyanate (FITC) for 15 minutes at 4°C. The efficiency of the two-step cell-surface labeling was determined by measuring the fluorescence intensity of the cell lysates. For comparison, the cell-surface azido moieties were also labeled by Staudinger ligation with biotin-modified phosphine 2 followed by treatment with avidin-FITC. The labeling with 9 was almost complete after an incubation time of 60 minutes (Figure 6a).
Interestingly, under identical conditions phosphine 2 (Agard et al., ACS Chem. Biol. 2006, 1 :644-648) gave significantly lower fluorescent intensities, indicating that cell surface labeling by Staudinger ligation is slower and less efficient. In each case, the control cells exhibited very low fluorescence intensities, demonstrating that background labeling is negligible. It was found that the two-step labeling approach with 9 had no effect on cell viability, as determined by morphology and exclusion of trypan blue (data not shown; Figure
7)-
The concentration dependence of the cell-surface labeling was studied by incubation of cells with various concentrations of 2 and 9 followed by staining with avidin-FTIC (Figure 6b). As expected, cells displaying azido moieties showed a dose-dependent increase in fluorescence intensity. Reliable fluorescent labeling was achieved at a 3 micromolar concentration of 9; however, optimal results were obtained at concentrations ranging from 30 to 100 micromolar. No increase in labeling was observed at concentrations higher than 100 micromolar owing to the limited solubility of 9. Next, attention was focused on visualizing azido-containing glyco conjugates of living cells by confocal microscopy. Thus, adherent Chinese hamster ovary (CHO) cells were cultured in the presence of Ac4ManNAz (100 micromolar) for three days. The resulting cell-surface azido moieties were treated with 9 (30 micromolar) for 1 hour and then with avidin-AlexaFluor488 for 15 minutes at 4°C. As expected, staining was observed only at the surface (Figure 8), and the labeling procedure was equally efficient when performed at either ambient temperature or 4°C. Furthermore, blank cells exhibited very low fluorescence staining, confirming that background labeling is negligible. Cell-surface glycoconjugates are constantly recycled by endocytosis, and to monitor this process, metabolically labeled cells were reacted with 9 and avidin-AlexaFluor488 according to the standard protocol and incubated at 37°C for 1 hour before examination by confocal microscopy. We observed that a significant quantity of labeled glycoproteins had been internalized into vesicular compartments.
At the completion of these studies, Bertozzi and co- workers reported a difluorinated cyclooctyne (DIFO) that reacts with azides at almost the same reaction rate as compound 3 (Baskin et al., Proc. Natl. Acad. Sci. USA 2007, 104:16793-16797). DIFO linked to AlexaFluor was employed to investigate the dynamics of glycan trafficking. It was found that after incubation for 1 hour, labeled glycans colocalized with markers for endosomes and Golgi.
4-Dibenzocyclooctynols such as 3 and 9 have several advantageous features for researchers such as ease of chemical synthesis and the possibility to further enhance the rate of cycloaddition by functionalization of the aromatic moieties. Modifying the aromatic rings may also offer an exciting opportunity to obtain reagents that become fluorescent upon [3+2] cycloaddition with azido- containing compounds, which will make it possible to monitor in real time the trafficking of glycoproteins and other biomolecules in living cells.
General methods and materials
Chemicals were purchased from Aldrich and Fluka and used without further purification. Dichloromethane was distilled from CaH2 and stored over molecular sieves 4 A. Pyridine was distilled from P2O5 and stored over molecular sieves 4 A. THF was distilled form sodium. All reactions were performed under anhydrous conditions under an atmosphere of Argon. Reactions were monitored by thin layer chromatography (TLC) on Kieselgel 60 F254 (Merck). Detection was by examination under ultraviolet (UV) light (254 nm) or by charring with 5% sulfuric acid in methanol. Flash chromatography was performed on silica gel (Merck, 70-230 mesh). Iatrobeads (60 micrometers) were purchased from Bioscan. 1H NMR (ID, 2D) and 13C NMR were recorded on a Varian Merc 300 spectrometer and on Varian 500 and 600 MHz spectrometers equipped with Sun workstations. 1H and 13C NMR spectra were recorded in
CDCI3, and chemical shifts (δ) are given in ppm relative to solvent peaks (1H, δ 7.24; 13C, δ 77.0) as internal standard for protected compounds. Negative ion matrix assisted laser desorption ionization time of flight (MALDI-TOF) were recorded on a VOYAGER-DE Applied Biosystems using dihydrobenzoic acid as a matrix. High-resolution mass spectra were obtained using a VOYAGER-DE Applied Biosystems in the positive mode by using 2,5-dihydroxyl-benzoic acid in THF as matrix.
3-tert-Butyl-dimethylsilyl-oxy-l ,2:5,6-dibenzocycloocta-l ,5,7-triene (5) tert-Butyl dimethyl silyl chloride (3.0 g, 20 mmol) was added to a stirred solution of 4 (2.2 g, 10 mmol) in a mixture Of CH2Cl2 (20 mL) and pyridine (5 mL). After stirring for 6 hours at room temperature, the reaction mixture was diluted with water and extracted with CH2Cl2 (40 mL). The combined organic extracts were washed with water and brine and then dried (MgSO4). The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/ethyl acetate, 7/1, v/v) to afford 5 (2.9 g, 87%). 1H NMR (300 MHz, CDCl3): δ 7.60 (1 H, aromatics), 7.32-7.11 (7 H, aromatics), 6.93 (1 H, d, J= 7.5 Hz, CH=CH), 6.85 (1 H, d, J= 7.5 Hz, CH=CH), 5.51 (1 H, dd, J= 6.3, 9.6 Hz, CHOSi), 3.54 (1 Η, dd, J= 6.3, 9.6 Hz, CH2), 3.21(1 Η, dd, J= 6.3, 9.6 Hz, CH), 0.96 (3 Η, s, CH5), 0.95 (3 Η, s, CH5), 0.94 (3 Η, s, CH5), 0.10 (3 Η, s, CH5), 0.07 (3 Η, s, CH5); 13C NMR (75 MHz, CDCl3): δ 148.0, 141.6, 140.8, 139.2, 138.3, 135.5, 135.0, 134.9, 133.0, 131.9, 131.6, 130.9, 130.8, 130.2, 77.0, 52.1, 34.5, 30.7, 30.5, 23.1, 5.8, 0.1; MALDI HRMS: m/z 359.181 1 [M + Na+]. Calcd for C22H28NaOSi 359.1807.
3-Hydroxy-7,8-dibromo-l,2:5,6-dibenzocyclooctene (6) A solution of bromine (0.8 g, 5 mmol) in CHCl3 was added dropwise to a solution of 5 (1.7g, 5 mmol) in CHCl3 (30 mL) at 00C. The reaction mixture was stirred at room temperature for 12 hours until the reaction was complete (monitored by TLC). The resulting mixture was washed with aqueous saturated sodium thiosulfate solution (15 mL), and dried (MgSO4). The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/CH2Cl2, 7/1, v/v) to afford 6 (1.2 g, 60%). 1H NMR (300 MHz, CDCl3): δ 7.54-7.47 (2 H, aromatics), 7.31 -6.72 (6 H, aromatics), 5.77 (1 H, d, J= 5.4 Hz, CHBr), 5.22 (1 H, dd, J= 3.6, 15.9 Hz, CHOH), 5.19 (1 H, d, J= 5.4 Hz, CHBr), 3.50 (1 Η, dd, J= 3.6, 15.9 Hz, CH2), 2.75(1 Η, dd, J = 3.6, 15.9 Hz5 CH2); 13C NMR (75 MHz, CDCl3): δ 141.3,
140.0, 137.2, 134.0, 133.4, 131.5, 131.3, 130.9, 127.8, 126.2, 123.7, 121.3, 76.5, 70.0, 62.3, 32.2; MALDI HRMS: m/z 402.9313 [M + Na+]. Calcd for
Figure imgf000030_0001
3-Hydroxy-7,8- didehydro-l,2:5,6-dibenzocyclooctene (3)
To a solution of 6 (1.1 g, 3 mmol) in tetrahydrofuran (50 mL) was added dropwise lithium diisopropylamide in tetrahydrofuran (2.0 M), (5 mL) under an atmosphere of Argon at room temperature. The reaction mixture was stirred for 2 hours at room temperature, after which it was poured into ice water (50 mL) and the resulting mixture was extracted with CH2Cl2 (2 x 100 mL). The combined extracts were washed with water and brine and then dried (MgSO4). The solvents were evaporated under reduced pressure and the residue purified by silica gel column chromatography (hexane/ethyl acetate, 5/1, v/v) to afford 3 (0.30 g, 45%). 1H NMR (300 MHz, CDCl3): δ 7.67 (1 H, aromatics), 7.37-7.18 (7 H, aromatics), 4.57 (1 H, dd, J= 2.1, 14.7 Hz, CHOH), 3.04 (1 H, dd, J= 2.1 , 14.7 Hz, CH2), 2.86(1 Η, dd, J= 2.1, 14.7 Hz5 CH2); 13C NMR (75 MHz5 CDCl3): δ 154.5, 150.6, 128.6, 127.1 , 1 127.0, 126.0, 125.8, 125.1, 124.7, 123.0, 122.7, 121.7, 1 1 1.9, 109.6, 74.2, 47.7.
Carbonic acid 7, 8-didehydro-l ,2:5,6-dibenzocyclooctene-3-yl ester 4- nitrophenyl ester (7)
To a solution of 3 (0.22 g, 1 mmol) in CH2Cl2 (30 mL) was added A- nitro-phenyl chloroformate (0.4 g, 2 mmol) and pyridine (0.4 ml, 5 mmol). After stirring 4 hours at ambient temperature, the reacting mixture was washed with brine (2 xlO mL), and the organic layer was dried (MgSO4). The solvents were evaporated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate, 10/1, v/v) to afford 7 (0.34 g, 89%). 1H NMR (300 MHz, CDCl3): δ 8.23-8.18 (2 H, aromatics), 7.56-7.54 (2 H, aromatics), 7.46-7.18 (8 H, aromatics), 5.52 (1 H, dd, J= 3.9, 15.3 Hz, CHOH), 3.26 (1 H, dd, J = 3.9, 15.3 Hz, CH2), 2.97 (1 H, dd, J= 3.9, 15.3 Hz, CH2); 13C NMR (75 MHz, CDCl3): δ 154.5, 150.7, 149.1, 148.7, 129.0, 127.4, 127.3, 126.7, 126.5, 125.5, 125.2, 124.3, 124.0, 122.6, 122.4, 120.8, 120.6, 120.2, 1 12.2, 108.5, 80.6, 44.8; MALDI HRMS: m/z 408.0852 [M + Na+]. Calcd for C23Hi5NNaO5 408.0848.
Carbonic acid 7,8-didehydro-l ,2:5,6-dibenzocyclooctene-3-yl ester, 8'- biotinylamine- 3',6"-dioxaoctane 1 ' -amide (9)
To a solution of 8 (37 mg, 0.1 mmol) and NEt3 (30 mg, 0.3 mmol) in DMF (10 mL) was added 7 (39 mg, 0.1 mmol) under an atmosphere of Argon. After stirring the reaction mixture overnight at ambient temperature, the solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography (CH2C12/CH3OH, 20/1, v/v) to afford 9 (44 mg, 71%). 1H NMR (500 MHz, CD3OD): δ 7.59 (1 H, aromatics), 7.42-7.33 (7 H, aromatics), 5.44, (1 H, dd, J= 5.0, 14.1 Hz, CHOH), 4.60, 4.46 (m, 2H, CHNH), 4.24 (s, 4H, OCH2CH2O), 3.72 (m, 4Η, OCH2), 3.64 (m, 2Η, CH2NH), 3.55 (m, IH, CHS), 3.33 (dd, 1Η, Jl = 12.0 Hz, J2 = 4.8 Hz, IH, CHHexoS), 3.23 (t, 2H, J= 6 Hz, CH2-NH2), 3.22, (1 H, dd, J= 5.0, 14.1 Hz, CH2), 2.88, (1 Η, dd, J = 5.0, 14.1 Hz, CH2), 2.68 (d, 1Η, J= 12.45 Hz, CHHendoS), 2.20 (t, 2H, J= 7.5 Hz, CH2CO), δ 1.4 (m, 6H, biotin-CH2). 13C NMR (75 MHz, CD3OD): δ 175.0, 164.9, 156.9, 152.5, 151.3, 129.9, 128.2, 128.1 , 127.2, 127.1 , 126.0, 125.7, 123.8, 121.2, 1 12.7, 109.8, 76.8, 70.2, 70.1, 69.8, 69.4, 62.1 , 60.4, 55.8, 54.6, 46.0, 42.6, 40.6, 39.9, 39.1 , 35.5, 28.6, 28.3, 25.6, 17.5, 16.1, 12.0; MALDI HRMS: m/z 643.2575 [M + Na+]. Calcd for C33H40N4NaO6S 643.2566.
General procedure for click reactions with carbohydrates and peptides
3-Hydroxy-7,8-didehydro-l,2:5,6-dibenzocyclooctene (2.2 mg, 0.01 mmol) was dissolved in CH3OH (1 mL) and an azide (3-azidopropyl 2,3,4,6- tetra-O-acetate-α-D-mannopyranoside, I -O-[dimethyl( 1 ,1 ,2- trimethylpropyl)silyl]- 4,6-O-isopropylidene-2-azido-2-deoxy-β- Dglucopyranose, 4,7,8-tri-O-acetyl-5-acetamido-9-azido-2,3-anhydro-3,5,9-tri- deoxy-D-glycero-D-galacto-non-2-enonic methyl ester, and 4-azido-7V-[(l ,l- dimethylethoxy)carbonyl]-Lphenylalanine, 1.0 equivalents) was added. The reaction was monitored by TLC, and after stirring the reaction mixture for 30 minutes at room temperature, the reaction had gone to completion. The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography to afford the desired products 10-13 respectively in quantitative yields. Compound 10; 1H NMR (500 MHz, CDCl3.): δ 7.83 ( 1 H, m, aromatics),
7.58-6.99 (7H, m, aromatics), 5.33-4.98 (4H, m, 2-H, 3-H, 4-H, CHOH), 4.90- 4.61 (IH, m, 1-H), 4.26,4.10 ( 2 Η, m, 6-H), 3.93 (1 Η, m, 5 -H), 3.70-3.60 (2 Η, m, OCH2CH2), 3.58-3.41 (2H, m, CH2N), 3.31,3.20,3.06, 2.91 (2 Η, m, CHOHCH2), 2.35-1.94 (12Η, m, CH3CO), 1.38-1.14 (2Η, m, CH2CH2N); 13C NMR (75 MHz, CDCl3): δ 170.9, 170.2, 148.5, 146.9, 145.5, 144.9, 141.2, 139.3, 138.0, 136.7, 135.5, 133.8, 133.0, 132.3, 131.6, 130.3, 129.5, 129.0, 128.3, 127.7, 127.2, 126.5, 125.0, 124.2, 98.0, 97.4, 70.1 , 69.5, 68.8, 66.2, 65.4, 64.9, 64.4, 62.6, 47.0, 45.1, 40.5, 32.1, 31.1, 30.6, 29.9, 22.9, 20.9, 14.3; MALDI HRMS: m/z 674.2330 [M + Na+]. Calcd for C33H37N3NaOn 674.2326. Compound 11; 1H NMR (500 MHz, CDCl3): δ 7.80, 7.65 (IH, d, J=7.5
Hz, aromatics), 7.48-7.06 (7 H, aromatics), 5.82, 5.72, 5.60, 5.48 (1 H, d, J = 7.09 Hz, 1-H), , 5.13-4.60 (1Η, m, CHOΗ ), 4.40-4.20 (2 Η, m, 2-H 3-H), 4.10- 3.90 (2H, m, 5-H, 6-H ), 3.89-3.63 (IH, m, 6-H), 3.54-3.40 (2 H, m, A-H, HC//CHOH), 3.07,2.66 (IH, m, HCHCHOH), 1.54-1.20 (6 H, m, CH(CH3)C(CH3)2), 0.98-0.60 (13 H, m, 2 CH3, CH(CH3 )2), 0.35-0.19 (6Η, m, Si(CH3)2); 13C NMR (75 MHz, CDCl3): δ 151.0, 149.2, 148.5, 148.0, 146.1 , 145.3, 142.4, 141.6, 140.8, 139.4, 138.1 , 136.6, 135.7, 134.9, 133.4, 132.4,
131.6, 130.6, 129.5, 128.9, 127.3, 103.5, 100.4, 99.8, 80.6, 73.3, 70.9, 69.3, 68.8, 65.5, 50.1, 46.6, 45.4, 44.4, 37.3, 33.3, 32.5, 28.4, 23.4, 22.6, 21.9, 4.6, 1.4, 0.7, 0.0; MALDI HRMS: m/z 630.2980 [M + Na+]. Calcd for C33H45N3NaO6Si 630.2975. Compound 12; 1H NMR (500 MHz, CDCl3): δ 7.95-7.69 (1 H, m, aromatics), 7.60-7.03 (7 H, m, aromatics), 6.77-6.26 (1 H, m), 5.98-8.81(1 H, m), 5.80-5.61 (1 H, m), 5.58-5.33 (1 H, m), 5.32-5.16 (2 H, m), 5.16-4.94 (1 H, m), 4.93-4.80 (1 H, m), 4.69-4.34 (1 H, m), 4.24-4.06 (1 H, m), 3.95-3.60 (3H, m), 3.53-2.90 (2 H, m), 2.32-1.57 (12 H, m); 13C NMR (75 MHz, CDCl3): δ 169.6, 160.5, 147.8, 145.5, 145.1, 144.6, 143.9, 140.5, 138.7, 138.2, 137.1 , 135.7, 134.5, 133.5, 132.7, 132.2, 131.8, 131.2, 130.8, 129.5, 129.0, 128.3, 127.7, 127.2, 126.5, 125.9, 124.8, 122.7, 108.0, 107.5, 75.1 , 69.6, 68.8, 67.1, 66.6, 52.8, 51.7, 47.3, 46.4, 45.3, 28.7, 28.3, 22.0, 19.8; MALDI HRMS: m/z 699.2282 [M + Na+]. Calcd for C34H36N4NaO, , 699.2278. Compound 13; 1H NMR (300 MHz, CD3OD): δ 7.8-6.8 (12H, m, aromatics), 5.33, 5.17 (IH, dd, J= 5.1, 10.5 Hz CHOH), 4.37 (IH, m, CHCOOH), 3.8, 3.23 3.77, 3.20 (2H, m, CH2CHOH), 3.21, 2.93 (2H, m, CH2CHNH), 1.35(9H, m, C(CH3)3); 13C NMR (75 MHz, CD3OD): δ 156.6, 145.1, 414.3, 139.6, 139.4, 138.0, 137.3, 136.1 , 135.3, 135.0, 133.7, 133.4, 132.1, 131.7, 130.6, 130.3, 130.0, 129.5, 129.2, 128.8, 128.3, 128.0, 127.6,
126.9, 126.6, 126.6, 126.2, 125.3, 125.1 , 124.8, 79.4, 76.8, 76.2, 68.6, 58.5, 54.9, 46.2, 40.5, 37.1 , 29.6, 29.3, 27.5; MALDI HRMS: m/z 549.21 18 [M + Na+]. Calcd for C30H30N4NaO5 549.21 14.
N-Boc-3, 6-dioxaoctane-l ,8-diamine
A solution of di-/er?-butyl dicarbonate (di-Boc) (6 g, 28 mmol, 0.5 equiv) in CH2Cl2 (100 mL) was added dropwise to a mixture of tris(ethylene glycol)- 1,8-diamine (7.6 g, 56 mmol) and diisopropylethylamine (10 niL, 57 mmol) at room temperature over a period of 2 hours. The reaction mixture was stirred for 6 hours, after which it was concentrated in vacuo. Purification by flash silica gel column chromatography (CH2C12/CH3OH, 10/1 , v/v) afforded #-Boc-3,6- dioxaoctane- 1,8-diamine (4.1 g, 58%). 1H NMR (300 MHz, CD3OD): δ 3.6 (s, 4H), 3.54 (t, 2H), 3.53 (t, 2H), 3.24 (t, 2H), 2.8 (t, 2H), 1.4 (s, 9H); MALDI HRMS: m/z 271.1641 [M + Na+]. Calcd for C1 1H24N2NaO4 271.1634.
N-Boc-N '-biotinyl-3, 6-dioxaoctane-l, 8-diamine A solution of vitamin H (Biotin) (2.2 g, 9 mmol), O-benzotriazol-1-yl-
N,N,]Ϋ ,/V-tetramethyluronium hexafluorophosphate (HBTU) (3 g, 8 mmol), and DIPEA (1.8 mL, 10 mmol) in DMF (100 mL) was stirred for 10 minutes at room temperature before being adding dropwise to a solution of 7V-Boc-3,6- dioxaoctane- 1 ,8-diamine (1.5 g, 6 mmol, 1 ). The reaction mixture was stirred for 1 hour at room temperature, after which the DMF was removed in vacuo to give an oily residue, which was purified by flash silica gel column chromatography (CH2C12/CH3OH, 25/1, v/v) to afford 7V-Boc-iV'-biotinyl-3,6-dioxaoctane-l, 8- diamine (2.0 g, 90%). 1H NMR (300 MHz, CD3OD): δ 4.5 (m, IH), 4.3 (m, IH), 3.6 (s, 4H), 3.54 (tt, 4H), 3.39 (t, 2H), 3.26 (t, 2H), 2.9 (dd, IH), 2.7 (d, IH), 2.2 (t, 2H), 1.7-1.5 (m, 8H), 1.4 (s, 9H); MALDI HRMS: m/z 497.2416 [M + Na+]. Calcd for C2]H38N4NaO6S 497.2410.
N -Biotinyl-3, 6-dioxaoctane-l , 8-diamine (8)
N-Boc-IΨ -biotinyl-3, 6-dioxaoctane-l , 8-diamine (1.9 g, 4 mmol) was dissolved in 50% TFA in CH2Cl2 (20 mL) and stirred for 1 hour at room temperature. The solvents were evaporated under reduced pressure to give an oily residue, which was purified by flash silica gel column chromatography (CH2C12/CH3OH, 10/1 , v/v) to afford 7 (1.3 g, 92%). 1H NMR (300MHz, DMSO-d6): δ 7.85 (t, IH, J= 5.7 Hz, NHCO), 6.42, 6.35 (s, 2H, NH), 4.29, 4.1 1 (m, 2Η, CHNH), 3.5 (s, 4H, OCH2CH2O), 3.3 (m, 4Η, OCH2), 3.16 (m, 2Η, CH2NH), 3.10 (m, IH, CHS), 2.81 (dd, 1Η, Jl = 12.0 Hz, J2 = 4.8 Hz, IH, CHHexoS), 2.64 (t, 2Η, J = 6 Hz, CH2-NH2), 2.52 (d, IH, J= 12.45 Hz, CUHendoS), 2.06 (t, 2H, J= 7.5 Hz, CH2CO), 1.6 (s, 2H, NH2), δ 1.4 (m, 6Η, biotin-CH2); 13C NMR (75 MHz, DMSO-d6): δ 171.9, 160.6, 71.7, 71.6, 69.5, 69.1 , 64.4, 59.2, 55.0, 54.2, 40.7, 38.4, 35.1 , 28.4, 28.1, 25.2; MALDI HRMS: m/z 397.1892 [M + Na+]. CaIcO fOr Ci6H30N4NaO4S 397.1885.
Reagents for biological experiments
Synthetic compounds 2 and 9 were reconstituted in DMF and stored at 800C. Final concentrations of DMF never exceeded 0.56% to avoid toxic effects.
Cell surface azide labeling and detection by fluorescence intensity
Human Jurkat cells (Clone E6-1; ATCC) were cultured in RPMI 1640 medium (ATCC) with L-glutamine (2 mM), adjusted to contain sodium bicarbonate (1.5 g L"1), glucose (4.5 g L"1), HEPES (10 mM), and sodium pyruvate (1.0 mM) and supplemented with penicillin (100 u mL~')/streptomycin (100 micrograms mL"1; Mediatech) and fetal bovine serum (FBS, 10%;
Hyclone). Cells were maintained in a humid 5% CO2 atmosphere at 37°C. Jurkat cells were grown in the presence of peracetylated ./V-azidoacetylmannosamine (Ac4ManNaz; 25 micromolar final concentration) for 3 days, leading to the metabolic incorporation of the corresponding /V-azidoacetyl sialic acid (SiaNAz) into their cell surface glycoproteins. Jurkat cells bearing azides and untreated control cells were incubated with the biotinylated compounds 2 and 9 (0-100 micromolar) in labeling buffer (DPBS, supplemented with FBS (1%)) for 0-180 minutes at room temperature. The cells were washed three times with labeling buffer and then incubated with avidin conjugated with fluorescein (Molecular Probes) for 15 minutes at 4°C. Following three washes and cell lysis, cell lysates were analysed for fluorescence intensity (485 ex / 520 em) using a microplate reader (BMG Labtech). Data points were collected in triplicate and are representative of three separate experiments. Cell viability was assessed at different points in the procedure with exclusion of trypan blue. Cell labeling and detection by fluorescence microscopy
Chinese hamster ovary (CHO) cells (Clone Kl ; ATCC) were cultured in Kaighn's modification of Ham's F-12 medium (F- 12K) with L-glutamine (2 mM), adjusted to contain sodium bicarbonate (1.5 g L"1) and supplemented with penicillin (100 u mL"1) / streptomycin (100 micrograms mL~' and FBS (10%). Cells were maintained in a humid 5% CO? atmosphere at 37°C. CHO cells were grown in the presence of Ac4ManNaz (100 micromolar final concentration) for 3 days to metabolically incorporate SiaNAz into their cell surface glycoproteins. CHO cells bearing azides and untreated control cells were then transferred to a glass coverslip and cultured for 36 hours in their original medium. Live CHO cells were treated with the biotinylated compound 9 (30 micromolar) in labeling buffer (DPBS, supplemented with FBS (1%)) for 1 hour at 4°C or at room temperature, followed by incubation with avidin conjugated with Alexa Fluor 488 (Molecular Probes) for 15 minutes at 4°C. Cells were washed 3 times with labeling buffer and fixed with formaldehyde (3.7% in PBS) or incubated for 1 hour at 37°C before fixation. The nucleus was labeled with the far red fluorescent TO-PRO-3 dye (Molecular Probes). The cells were mounted with PermaFluor (Thermo Electron Corporation) before imaging. Initial analysis was performed on a Zeiss Axioplan2 fluorescent microscope. Confocal images were acquired using a 6OX (NAl .42) oil objective. Stacks of optical sections were collected in the z dimensions. The step size, based on the calculated optimum for each objective, was between 0.25 and 0.5 micrometers. Subsequently, each stack was collapsed into a single image (..--projection). Analysis was perfoπned offline using ImageJ 1.39f software (National Institutes of Health, USA) and Adobe Photoshop CS3 Extended Version 10.0 (Adobe Systems Incorporated), whereby all images were treated equally. Example 2 Alkyne Reagents Containing Biotin and a Cleavable Linker
Azides, which are extremely rare in biological systems, are emerging as attractive chemical handles for bioconjugation (Dedola et al., Org. Biomol. Chem. 2007, 5, 1006; KoIb and Sharpless, Drug D is. Today 2003, 8, 1 128; Moses and Moorhouse, Chem. Soc. Rev. 2007, 36, 1249; Nandivada et al., Adv. Mater. 2007, 19, 2197; Wu and Fokin, Aldrichimica ACTA 2007, 40, 7; Agard et al., ACS Chem. Biol. 2006, 1, 644). In particular, the Cu(I) catalyzed 1,3-dipolar cyclization of azides with terminal alkynes to give stable triazoles has been employed for tagging a variety of biomolecules including proteins, nucleic acids, lipids, and saccharides (Chin et al., Science 2003, 301, 964; Gierlich et al., Org. Lett. 2006, 8, 3639; Kho et al., Proc. Natl. Acad. Sci. 2004, 101, 12479; Link et al., Proc. Natl. Acad. Sci. 2006, 103, 10180; Wang et al., J. Am. Chem. Soc. 2003, 125, 3192). The cycloaddition has also been used for activity-based protein profiling (Speers et al., J. Am. Chem. Soc. 2003, 125, 4686), monitoring of enzyme activity, and the chemical synthesis of microarrays and small molecule libraries (Sun et al., Bioconjugate Chem. 2006, 17, 52).
An attractive approach for installing azides into biomolecules is based on metabolic labeling whereby an azide containing biosynthetic precursor is incorporated into biomolecules using the cells' biosynthetic machinery (Prescher and Bertozzi, Nat. Chem. Biol. 2005, 1, 13). This approach has been employed for tagging proteins, gl yeans, and lipids of living systems with a variety of reactive probes. These probes can facilitate the mapping of saccharide-selective glycoproteins and identify glycosylation sites (Hanson et al., J. Am. Chem. Soc. 2007, 129, 7266). Alkyne probes have also been used for cell surface imaging of azide-modified bio-molecules and a particularly attractive approach involves the generation of a fluorescent probe from a non-fluorescent precursor by a [3+2] cycloaddition (Sivakumar et al., Org. Lett. 2004, 6, 4603).
We describe here reagents including an alkyne fragment, a cleavable linker fragment, and biotin. Such compounds are expected to be valuable for biological research. Thus, the alkyne fragment of the reagent can react with various biomolecules containing an azide fragment to give stable triazole adducts. The biotin fragment gives an opportunity to retrieve the tagged compounds by affinity chromatography using immobilized avidin. The cleavable linker allows the release of tagged and captured biomolecules for analysis. For example, released proteins or glycoproteins can be characterized by standard proteomics or glycomics analysis (Too, Expert Rev. Proteomics 2007, 4, 603; Bantscheff et al., Anal. Bioanal. Chem. 2007, 389, 1017; Lau et al, Proteomics 2007, 7, 2787). Release of the proteins and glycoproteins is much more practical than previously reported analysis of biomolecules attached to immobilized avidin (Hanson et al., J. Am. Chem. Soc. 2007, 129, 7266). Compound 21 is an example of the new class of reagent (Figure 9). It contains a 4-dibenzocyclooctynol fragment for reaction with azides, a disulfide, which can be cleaved with reducing reagents such as dithiothreitol (DTT), and biotin. The quantification of differences between physiological states of a biological system is a technically challenging task in proteomics (Too, Expert Rev. Proteomics 2007, 4, 603; Bantscheff et al., Anal. Bioanal. Chem. 2007, 389, 1017; Lau et al., Proteomics 2007, 7, 2787). In addition, to the classical methods of differential protein gel or blot staining by dyes and fluorophores, mass- spectrometry-based quantification methods is gaining popularity. Most of the latter methods employ differential stable isotope labeling to create a specific mass tag that can be recognized by a mass spectrometer and at the same time provide the basis for quantification. These mass tags can be introduced into proteins or peptides by (i) metabolical labeling, (ii) by chemical means, (iii) enzymatically, or (iv) by spiking with synthetic peptide standards.
Reagents composed of an alkyne, a cleavable linker and biotin can be employed to introduce mass tags into proteins, glycoproteins and other biomolecules containing an azide fragment. Thus, by employing reagents such as 21 and 22, different mass tags can be introduced to quantify proteins, glycoproteins, glycopeptides, peptides and carbohydrates. The chemical synthesis of 21 and 22 is depicted in Schemes 4 and 5, respectively (Figures 10 and 11). Various alkyne moieties, cleavable linkers and biotin derivatives are depicted in Figure 12 and alkyne and reactive diene derivatives are depicted in Figure 13.
Example 3
Fast Click Reactions for Labeling of Living Cells and Nanoparticles
An alterative approach for preparing 4-diben∑ocyclooctynol and using this compound for the preparation of amine containing click reagents. 4-Dibenzocyclooctynol 45 could be prepared by an alternative synthetic route (Scheme 6; Figure 14). Thus, known of dibenzosuberenone (41) was treated trimethylsilyl diazomethane in the presence Of BF3OEt2 in CH2Cl2 (20 ml) at -10 0C to give όT-Z-Dibenzofαejcyclooctatrien-S-one (42) in good yield. The ketone of 42 was reduced with sodium borohydride in a mixture of ethanol and THF to give alcohol 43, which could be converted into 4- dibenzocyclooctynol 45 by bromination of the double bond followed by elimination of the resulting compound 44 by treatment LDA in THF. Compound 45 could be oxidized to the corresponding ketone 46 by employing Dess-Martin reagent. Compounds 45 and 46 were converted into amine containing derivatives
49, 50 and 51. The attraction of these compounds is that the can easily be derivatized with various probes such as fluorescent tags and biotin. Furthermore, the amine gives an easy chemical handle for attachment to polymeric supports. Thus, alcohol 45 was converted into jp-nitrophenyl ester 49 by reaction with 4- nitro-phenyl chloroform ate (0.4 g, 2 mmol) and pyridine. The target compound compound 49 was obtained by reaction of 49 with an excess of tris(ethylene glycol)- 1 ,8-diamine. Compound 50 was obtained by reaction of 46 with bromoacetic acid in the presence of lithium diisopropylamide in tetrahydrofuran followed by condensation of the resulting acid 48 with tris(ethylene glycol)- 1,8- diamine in DMF in the presence of the coupling reagent HATU and the base DIPEA. Finally, derivative 51 was prepared by oxime formation be reaction of ketone 51 with 7V-{2-[2-(2-amino-ethoxy)-ethoxy]-ethyl}-2-aminooxy-acetamide (84 mg, 0.251 mmol) in the presence of acetic acid and in a mixture of methanol and dichloromethane. A feature of 51 is that the oxime linkage can be cleaved by treatment with aqueous acid to detach the captured compound from the click reagent.
Experimental
6H-Dibenzo[a,e]cyclooctatrien-5-one (42). To a stirred solution of dibenzosuberenone 41 (2.888 g, 14.0 mmol) and BF3OEt2 (2.59 ml, 21.0 mmol) in CH2Cl2 (30 ml) was added dropwise a solution of trimethylsilyl diazomethane (10.5 ml, 21.9 mmol) in CH2Cl2 (20 ml) at -10 0C over 1 hour. The mixture was stirred at -10 0C for 2 hours, and then poured into ice water. The aqueous layer was extracted with CH2Cl2 (3x 100 ml) and the organic layers were combined. -The combined organic layers were washed with brine and dried (MgSO4). The solvent was removed under reduced pressure, and the crude product was purified by flash chromatography on silica gel (2: 1 — 1 :2 v/v hexanes/C^Cl?) to give the product as pale solid (2.220 g, 72%). 1H NMR (300 MHz, CDCl3): δ 8.26 (1 H, q, J= 1.4, 6.6 Hz), 7.13-7.43 (7 H, m), 7.05 (2 H, q, J= 3.8, 12.9 Hz), 4.06 (2 H, s). 13C NMR (75 MHz, CDCl3): δ 196.6, 136.9, 136.3, 135.4, 133.8, 133.1, 132.4, 131.4, 130.6, 129.3, 128.8, 128.0, 127.3, 126.9, 48.4. MALDI HRMS: m/z [M+Na+]. Calcd for C6Hi2NaO: 243.0786 (Chaffms, S.; Brettreich, M.; Wudl, F. Synthesis 2002, 1 191-1 194).
5 ,6-Dihydro-dibenzo[a,e] 'cycloocten-5-ol (43). To a stirred solution of 42 (2.203 g, 10 mmol) in 1 : 1 EtOH/THF (120 ml) at room temperature was added slowly sodium borohydride (0.757 g, 20 mmol), and the reaction mixture was stirred at room temperature for 7 hours. TLC indicated that the reaction was complete, and the reaction mixture was quenched by slow addition of acetic acid (1 ml). The solvent was evaporated, and the residue was dissolved in CH2Cl2 (100 ml) and brine (100 ml), extracted with CH2Cl2 (4χ 100 ml). The organic phase was combined, dried (MgSO4) and evaporated to give the product as white solid (2.223 g, 100%), which is directly used in the next step reaction without further purification. 1H NMR (300 MHz, CDCl3): δ 7.50 (1 H, m), 7.14-7.30 (7 H, m), 6.90 (2 H, q, J= 2.7, 12.0 Hz), 5.31 (1 H, q, J= 6.3, 10.0 Hz), 3.41 (2 H, m). 13C NMR (75 MHz, CDCl3): δ 141.7, 136.7, 136.2, 134.5, 131.7, 131.5, 130.1, 129.9, 129.3, 128.7, 127.4, 127.2, 126.9, 125.9, 74.4, 42.7. MALDI HRMS: m/z [M+Na+]. Calcd for C|6H,4Na0: 245.0942. ll,12-Dibromo-5, 6, 11 , 12-tetrahydro-dibenzo [a,e] cycloocten-5-ol (44). To a stiiτed solution of 43 (2.223 g, 10 mmol) in 1 :1 CHCl3 (50 ml) at room temperature was added dropwise bromine (0.512 ml, 10 mmol), and the reaction mixture was stirred at room temperature for 0.5 hour TLC indicated that the reaction was complete, and the solvent was removed at room temperature under reduced pressure. The residue was purified by flash chromatography on silica gel (2: 1 — 1 :2 v/v hexanes/CH2Cl2) to give the product as yellowish oil (2.220 g, 58%). 1H NMR (300 MHz, CDCl3): δ 7.54-7.47 (2 H, aromatics), 7.31 -6.72 (6 H, aromatics), 5.77 (1 H, d, J= 5.4 Hz, CHBr), 5.22 (1 H, dd, J = 3.6, 15.9 Hz, CHOH), 5.19 (1 H, d, J= 5.4 Hz, CHBr), 3.50 (1 Η, dd, J = 3.6, 15.9 Hz, CH2), 2.75(1 Η, dd, J= 3.6, 15.9 Hz5 CH2). 13C NMR (75 MHz, CDCl3): δ 141.3, 140.0, 137.2, 134.0, 133.4, 131.5, 131.3, 130.9, 127.8, 126.2, 123.7, 121.3, 76.5, 70.0, 62.3, 32.2. MALDI HRMS: m/z 402.9313 [M+Na+]. Calcd for C6Hi4Br2NaO: 402.9309.
5, 6-Dihydro-l 1 ,12-didehydro-dibenzo[a,e] cycloocten-5-ol (45). To a stirred solution of 44 (1.528 g, 4 mmol) in tetrahydrofuran (40 ml) was added dropwise lithium diisopropylamide in tetrahydrofuran (2.0 M) (8 ml, 16 mmol) under an atmosphere of Argon at room temperature. The reaction mixture was stirred for 0.5 hour at room temperature, after which it was quenched by addition of dropwise water (0.5 ml). The solvents were evaporated under reduced pressure, and the residue was purified by flash chromatography on silica gel (2:1 — 0: 1 v/v hexanes/GHbCh) to give the product as white solid (0.503 g, 57%). 1H NMR (300 MHz, CDCl3): δ 7.67 (1 H, aromatics), 7.37-7.18 (7 H, aromatics), 4.57 (1 H, dd, J= 2.1, 14.7 Hz, CHOH), 3.04 (1 H, dd, J= 2.1, 14.7 Hz, CH2), 2.86(1 Η, dd, J= 2.1 , 14.7 Hz, CH2). 13C NMR (75 MHz, CDCl3): δ 154.5, 150.6, 128.6, 127.1, 1 127.0, 126.0, 125.8, 125.1, 124.7, 123.0, 122.7, 121.7, 1 1 1.9, 109.6, 74.2, 47.7. 6H- 1 l,12-Didehydro-dibenzo[a,e]cyclooctatrien-5-one (46). To a stirred solution of 45 (0.172 g, 0.781 mmol) in CH2Cl2 (40 ml) was added Dess-Martin reagent (0.397 g, 0.937 mmol). The reaction mixture was stirred for 0.5 hour at room temperature, TLC indicated that the reaction was complete. The reaction mixture was filter through a short pad of silica gel, and washed with CH2Cb. The filtrate was concentrated, and the residue was purified by flash chromatography on silica gel (1 :1 — 0: 1 v/v hexanes/CIHbCb) to give the product as white solid (0.158 g, 93%). 1H NMR (300 MHz, CDCl3): δ 7.29-7.57 (8H, m), 4.17 (1 H, d, J= 10.6 Hz), 3.64 (1 H, J = 10.6 Hz). 13C NMR (75 MHz, CDCl3): δ 200.4, 154.7, 148.2, 131.21 (2 C), 131.18, 129.3, 128.2, 127.8, 126.3, 125.9, 122.2, 1 1 1.1, 109.4, 49.3. MALDI HRMS: m/z [M+Na+]. Calcd for Ci6H10NaO: 241.0629. Carbonic acid 5, 6-dihydro- 11 , 12-didehydro-dibenzofa, e] 'cycloocten-5-yl ester 4 -nitro -phenyl ester (47). To a solution of 45 (0.22 g, 1 mmol) in CH2Cl? (30 mL) was added 4-nitro-phenyl chloroformate (0.4 g, 2 mmol) and pyridine (0.4"ml, 5 mmol). After stirring 4 hours at ambient temperature, the reacting mixture was washed with brine (2x10 mL), and the organic layer was dried (MgSO4). The solvents were evaporated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate, 10/1, v/v) to afford 47 (0.34 g, 89%). 1H NMR (300 MHz, CDCl3): δ 8.23-8.18 (2 H, aromatics), 7.56-7.54 (2 H, aromatics), 7.46-7.18 (8 H, aromatics), 5.52 (1 H, dd, J= 3.9, 15.3 Hz, CHOH), 3.26 (1 H, dd, J = 3.9, 15.3 Hz, CH2), 2.97 (1 Η, dd, J = 3.9, 15.3 Hz, CH2). 13C NMR (75 MHz, CDCl3): δ 154.5, 150.7, 149.1, 148.7, 129.0, 127.4, 127.3, 126.7, 126.5, 125.5, 125.2, 124.3, 124.0, 122.6, 122.4, 120.8, 120.6, 120.2, 112.2, 108.5, 80.6, 44.8. MALDI HRMS: m/z 408.0852 [M+Na+]. Calcd for C23H15NNaO5: 408.0848.
(5, 6-Dihydro-l 1, 12-didehydro-dibenzofa, e] cycloocten-5-yloxy)-acetic acid (48). To a stirred solution of bromoacetic acid (0.280 g, 2 mmol) in tetrahydrofuran (40 ml) at 0 0C was added slowly sodium hydride (60% oil dispersion, 0.120 g, 3.0 mmol). The mixture was stirred at 0 0C for 10 minutes, then 45 (0.220 g, 1.0 mmol) was added. The mixture was stirred at 0 0C for another 10 minutes, then warmed to room temperature and stirred for 1 day. The reaction was quenched by 1 drop of HOAc, filtered through a short pad of silica gel washed by EtOAc, then concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (1 :0-l :1 CH2Cl2ZEtOAc) to give the product as white solid (0.60 g, 22%). 1H NMR (300 MHz, CDCl3): δ 7.69 (1 H, d, J = 7.7 Hz), 7.49 (1 H, d, J = 6.7 Hz), 7.41 (1 H, m), 7.27-7.35 (5 H, m), 4.25 (1 H, m), 4.15 (2 H, d, J = 8.0 Hz), 3.30 (1 H, dd, J = 2.2, 14.8 Hz), 2.72 (1 H, dd, J = 3.6, 14.8 Hz). 13C NMR (75 MHz, CDCl3): δ 154.5, 150.6, 128.6, 127.1, 1127.0, 126.0, 125.8, 125.1, 124.7, 123.0, 122.7, 121.7, 11 1.9, 109.6, 74.2, 47.7.MALDI HRMS: m/z 301.0838 [M+Na+]. Calcd for
Figure imgf000043_0001
{2-[2-(2-Amino-ethoxy)-ethoxy]-ethyl}-carbamic acid 5, 6-dihydro-l 1, 12- didehydro-dibenzo[a,e]cycloocten-5-yl ester (49). To a stirred solution of 47 (0.077 g, 0.2 mmol) and tris(ethylene glycol)- 1 ,8-diamine (0.293 ml, 2 mmol) in CH2Cl2 (20 ml) at room temperature was added Et3N (0.139 ml, 1.0 mmol). The reaction mixture was stirred for 3 hours at room temperature, after which the solvent was removed under reduced pressure. The residue was purified by flash chromatography on Iatrobeads (8-30% v/v MeOHZCH2Cl2) to give the product as yellowish solid (0.063 g, 80%). 1H NMR (300 MHz, CDCl3): δ 7.51 (1 H, d, J = 7.3 Hz), 7.24-7.37 (7 H, m), 5.81 (1 H, s, NH), 5.48, (1 H, br), 3.50-3.68 (8H, m), 3.39 (2H, m), 3.16 (1 H, d, J= 14.8 Hz), 2.91 (2H, br), 2.88 (1 H, d, J = 14.8 Hz), 2.57 (2 H, br, NH2). 13C NMR (75 MHz, CDCl3): δ 155.7, 152.2, 151.1, 130.0, 128.1 , 128.0, 127.2, 127.1 , 126.3, 126.0, 123.9, 123.8, 121.3, 1 13.0, 1 10.0, 76.7, 72.8, 70.3, 70.2, 70.1 , 70.0, 46.2, 41.5, 41.0. MALDI HRMS: m/z 417.1746 [M+Na+]. Calcd for C23H26N2NaO4: 417.1790.
N-{2-[2-(2-Amino-ethoxy)-ethoxy] -ethyl}-2-(5 , 6-dihydro-l 1 , 12- didehydro-dibenzo[a,e]cycloocten-5-yloxy)-acetamide (50). To a stirred solution of 48 (5.6 mg, 0.02 mmol) and tris(ethylene glycol)- 1,8-diamine (0.0292 ml, 0.2 mmol) in DMF (3 ml) at room temperature was added HATU coupling reagent (7.6 mg, 0.02 mmol) and DIPEA (0.0348 ml, 0.2 mmol). The reaction mixture was stirred for 2 hours at room temperature, after which the solvent was removed under reduced pressure. The residue was purified by flash chromatography on Iatrobeads (8-30% v/v MeOH/CH2Cl2) to give the product as colorless oil. MALDI HRMS: m/z 431.1916 [M+Na+]. Calcd for C24H28N2NaO4: 431.1947. N-{2-[2-(2-Amino-ethoxy)-ethoxy]-ethyl}-2-(6H-l 1 , 12-didehydro- dibenzo [a,e] cycloocten-5-ylideneaminooxy)-acetamide (51). A solution of 46 (46 mg, 0.211 mmol), Λ/-{2-[2-(2-amino-ethoxy)-ethoxy]-ethyl}-2-aminooxy- acetamide (84 mg, 0.251 mmol) and acetic acid (0.1 ml) in 1 : 1 v/v MeOHZCH2Cl? (4 ml) was stirred at room temperature for 2 days. The solvent was removed under reduced pressure, and the residue was purified by flash chromatography on Iatrobeads (4-15% v/v MeOHZCH2Cl2) to give the product as yellowish solid.(56 mg, 63%). MALDI HRMS: mZz 444.1835 [M+Na+]. Calcd for C24H27N3NaO4: 444.1899.
Reaction kinetics of cycloaddition of derivatives of 4-diben∑ocyclooctynol. A number of analogs (63-68) of 4-dibenzocyclooctynol (61) were prepared and the influence of these modifications on the reaction rate of the cycloaddition with benzyl azide was determined by integration of the benzylic proton signals in 1H NMR spectrum. Figure 15 shows the second order constant of compounds 61-68. A surprising finding was that compound 62, which does not have a hydroxyl function, reacts approximately 70-times slower that the analogous 4-dibenzocyclooctynol (61). Acylation of the hydroxyl of 61 such as in compounds 63 and 64, led to a slow reduction in reaction rate. Alkylation of 61, as in compound 65, also resulted in a slower rate of reaction. Compound 66, which has a gem-difluoride reacted at a similar rate as compound 61. Interestingly, ketone 67 reacts with a slightly higher reaction rate than 1. Oxime 68 has a similar reaction than 61. These results demonstrate that modification of the hydroxyl of 61 can have a dramatic influence on the rate of cycloaddition.
Experimental
Synthesis of 70. To a stirring solution Of LiAlH4 (0.38 g, 10 mmol) and AlCl3 (1.3 g, 10 mmol) in Et2O (100 mL) was added 69 (1.1 g, 5 mmol). The reaction was kept at O0C for 12 hours, and then it was quenched with water (100 ml). The aqueous layer was extracted with ether (4 X 100 ml), and the combined organic extracts were washed with water (100 ml) and brine (100 ml). The crude product was purified by column chromatography (hexaneZCH2Cl2, 2Zl, vZv) to yield two 70 (0.63 g, 61%; Scheme 7; Figure 16). 1H NMR (CDCl3, 300 MHz) O: 6.90-7.21 (m, 8H, aromatics), 6.67 (s, 2H, CH=CH), 3.11 (s, 4H, -CH2-CH2-). 13C NMR (75 MHz, CDCl3) δ: 140.0, 136.9, 131.6, 130.3, 130.1 , 128.7, 128.5, 127.1, 125.6. HRMS calcd for Ci6Hi4Na (M+Na): 229.0993. Found: 229.1003. Synthesis of 71. A solution of bromine (0.4 g, 2.5 mmol) in CHCl3 (10 mL) was added dropwise to a solution of 70 (0.5 g, 2.5 mmol) in CHCl3 (20 mL) at 00C. The reaction mixture was stirred at room temperature for 12 hours until the reaction was complete (monitored by TLC). The resulting mixture was washed with aqueous saturated sodium thiosulfate solution (15 mL), and dried (MgSO4). The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/CH2Cl2, 10/1, v/v) to afford 71 (0.53 g, 58%; Scheme 7; Figure 16). 1H NMR (CDCl3, 300 MHz) S: 7.06-7.52 (m, 8H, aromatics), 5.79 (s, 2H, Ph-CH-Br), 3.05 (dd, 2H, PhHCH), 2.84 (m, 2Η, PhHCH). 13C NMR (75 MHz, CDCl3) δ: 141.8, 138.7, 131.3, 130.6, 129.3, 126.6, 35.7.
Synthesis of 62. To a solution of 71 (0.36 g, 1 mmol) in tetrahydrofuran (20 mL) was added dropwise /-BuOK in tetrahydrofuran (2.0 M), (2 mL) under an atmosphere of Argon at room temperature. The reaction mixture was stirred for 2 hours at room temperature, after which it was poured into ice water (10 mL) and the resulting mixture was extracted with CH2Cl2 (2 X 50 mL). The combined extracts were washed with water, brine and dried (MgSO4) and the solvents were evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/CH2Cl2, 2/1, v/v) to afford 62 (50 mg, 25%; Scheme 7; Figure 16). 1H NMR (CDCl3, 300 MHz)
Figure imgf000045_0001
1.29-1 ΛA (m, 8H, aromatics), 3.28-3.15 (m, 2H, -HCH-HCH-), 2.40-2.27 (m, 2Η, -HCH-
ΗCH-). 13C NMR (75 MHz, CDCl3) δ: 153.8, 129.6, 127.9, 126.7, 126.3, 124.2, 111.8, 36.7.
Synthesis of 63. To a stirring solution of 72 (38 mg, 0.1 mmol) in CH2Cl2 (15 mL) was added 73 (15 mg, 0.2 mmol) and TEA (10 μL). The reaction mixture was stirred at room temperature for 12 hours. The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (CH2C12/CH3OH, 20/1, v/v) to afford 63 (25 mg, 77%; Scheme 8; Figure 17). 1H NMR (CDCl3, 300 MHz) r5: 6.94-7.43 (m, 8H, aromatics), 5.42 (m, IH, Ph-CH-O), 3.61(m, 2H, CZZ2OH), 3.30 (m, 2H, CZZ2NH), 3.08 (dd, IH, J= 15.0, 1.8 Hz, PhHCZZ), 2.84 (dd, IH, J= 15.0, 3.9 Hz, PhHCZZ), 1.53-1.68 (m, 2H, CZZ2CH2OH). 13C NMR (75 MHz, CDCl3) δ: 150.8, 149.1, 128.9, 128.0, 127.0, 126.1 , 126.0, 125.9, 125.8, 125.3, 125.1, 125.0, 122.8,122.6, 120.3, 1 1 1.9, 108.9, 58.6, 45,2, 36.8, 36.7, 31.6. HRMS calcd for C20Hi9O3Na (M+Na): 344.1263. Found: 344.1896.
Synthesis of 64. To a stirring solution of 61 (22 mg, 0.1 mmol) in pyridine (4 mL) was added Ac2O (1 mL). The reaction mixture was stirred at room temperature for 12 hours. The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/CH2Cl2, 1/1, v/v) to afford 64 (21 mg, 81 %; Scheme 9; Figure 18). 1H NMR (CDCl3, 300 MHz) δ: 1 Al-I M (m, 8H, aromatics), 5.49 (m, IH, Ph-CH- OAc), 3.06 (dd, IH, J= 15.6, 2.4 Hz, PhHCZZ), 2.84 (dd, IH, J= 15.6, 2.4 Hz, PhHCZZ), 2.17(s, 3H, CH3). 13C NMR (75 MHz, CDCl3) δ: 169.9, 151.3, 151.1, 130.1, 128.3, 128.1, 127.4, 127.3, 126.5, 126.2, 124.0, 121.6, 1 13.2, 110.0, 76.6, 46.5, 21.4. HRMS calcd for C18Hi4O2Na (M+Na): 285.0891. Found: 285.1005. Synthesis of 65. To a stirring solution of 61 (22 mg, 0.1 mmol) in DMF (2 mL) was added NaH (8 mg, 0.2 mmol), the mixture was stirred at room temperature for 1 hour and then benzyl bromide (BnBr, 34 mg, 0.2 mmol) was added. The reaction mixture was stirred at room temperature for 12 hours. The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/CH?Cl2, 2/1 , v/v) to afford 65 (18 mg, 59%; Scheme 9; Figure 18). 1H NMR (CDCl3, 300 MHz) δ: 7.70 (m, IH, aromatics), 7.39-7.17 (m, 13H, aromatics), 4.55 (dd, 2H, J = 1 1.6, 3.0 Hz, CH2Ph), 4.26 (m, 1Η, Ph-CH-OBn), 3.23 (dd, 1Η, J= 15.0, 2.4 Hz, PhHCZZ), 2.84 (dd, IH, J= 15.0, 2.4 Hz, PhHCZZ). 13C NMR (75 MHz, CDCl3) δ: 152.6, 151.1, 137.3, 128.4, 127.3, 127.1, 127.0, 126.5, 126.2, 125.8, 125.7, 125.1 , 125.0, 123.6, 123.0, 120.5, 1 1 1.8, 109.5, 81.5, 71.0, 46.1. HRMS calcd for C23H18ONa (M+Na): 333.1255. Found: 333.1905.
Synthesis of 74. To a stirring solution of LDA (20 ml, 40 mmol, 2.0 M solution in THF) in THF (200 mL) at room temperature was added a solution of ketone 69 (4.4 g, 20 mmol) in THF (40 mL) over 1 hour using a syringe pump. After an additional 20 minutes of stirring at room temperature, chlorotriethylsilane (6.7 ml, 40 mmol) was added. The solution was stirred at room temperature for 1 hour. The reaction mixture was concentrated on a rotary evaporator, and the crude product was purified directly by column chromatography (hexane/CH2Cl2, 10/1 , v/v) to yield a clear oil 74 (5.8 g, 85%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) δ: 7.25-6.95 (m, 8H, aromatics), 6.72 (t, 2H, J= 7.0 Hz, CH=CH), 6.1 1 (s, IH, J= 7.0 Hz, CH=C), 0.87-0.82 (m, 9H, CH2-CH3), 0.61-0.48 (m, 6Η, CH2-CH3). 13C NMR (75 MHz, CDCl3) 5: 151.9, 138.2, 137.3, 137.2, 136.7, 136.5, 134.0, 132.6, 130.1, 129.3, 129.0, 128.6, 128.0, 127.1 , 127.0, 126.2, 1 12.7, 6.9, 5.1. HRMS calcd for C22H26OSiNa (M+Na): 357.1651. Found: 357.1993.
Synthesis of 75. To a stirring solution of fluorinating reagent (1- chloromethyl-4-fluoro-l,4-diazoniabicyclo[2.2.2]octane bis-(tetrafluoroborate)) available under the trade designation SELECTFLUOR from Air Products
(Allentown, PA) (6.3. g, 18 mmol) in DMF (20 ml) at 00C was added a solution of silyl enol ether 74 (5.0 g, 15 mmol) DMF (20 ml) via an addition funnel over 10 minutes. The reaction was allowed to slowly warm to room temperature while stirring over 30 minutes, and then it was quenched with water (100 mL). The aqueous layer was extracted with ether (4 X 100 ml), and the combined organic extracts were washed with water (3 X 100 mL) and brine (I X 200 mL). The crude product was purified by column chromatography (hexane/CH?Cl2, 1/1, v/v) to yield 75 (2.4 g, 66%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) δ: 7.80-7.06 (m, 8H, aromatics), 6.85 (s, 2H, CH=CH), 4.41-4.36 (m, 1Η,CHF). 13C NMR (75 MHz, CDCl3) δ: 196.4, 152.8, 144.1, 140.4, 138.5,
135.8, 135.5, 135.4, 131.5, 129.6, 128.3, 127.7, 126.9, 125.3, 124.6, 124.1 , 63.6, 56.9; 19F(CDCl3, 283 MHz) δ: -109.1 (s, IF). HRMS calcd for Ci6HnFONa (M+Na): 261.0692. Found: 261.1237.
Synthesis of 76. To a stirring solution of LDA (10 ml, 20 mmol, 2.0 M solution in THF) in THF (100 mL) at room temperature was added a solution of ketone 75 (2.4 g, 10 mmol) in THF (20 mL) over 1 hour using a syringe pump. After an additional 20 minutes of stirring at room temperature, chlorotriethylsilane (3.3 ml, 20 mmol) was added. The solution was stirred at room temperature for 1 hour. The reaction mixture was concentrated on a rotary evaporator, and the crude product was purified directly by column chromatography (hexane/CH2Cl2, 5/1, v/v) to yield a clear oil 76 (2.8 g, 79%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) δ: 7.41 (m, 2H, aromatics), 7.22(m, 4H, aromatics), 7.08 (m, 2H, aromatics), 6.91 (s, 2H, CH=CH), 0.94 (t, 9H, J= 7.8 Hz, CH2-CH3), 0.63 (m, 6H, CH2-CH3). 13C NMR (75 MHz, CDCl3) δ: 145.8, 142.6, 137.3,137.2, 136.3, 136.1 , 134.2, 133.3,132.6, 132.1 , 130.1, 130.0, 129.6, 129.3, 128.9,128.8, 128.7, 128.5, 128.2, 127.5, 127.4, 6.7, 6.3, 5.2; 19F(CDCl3, 283 MHz) δ: -1 1 1.978 (s, IF). HRMS calcd for C22H23FOSiNa (M+Na): 373.1400. Found: 373.1522.
Synthesis of 77. To a stirring solution of SELECTFLUOR fluorinating reagent (3.5.g, 10 mmol) in DMF (15 ml) at 00C was added a solution of silyl enol ether 76 (2.8 g, 8 mmol) DMF (10 ml) via an addition funnel over 10 minutes. The reaction was allowed to slowly warm to room temperature while stirring over 30 minutes, and then it was quenched with water (100 mL). The aqueous layer was extracted with ether (4 X 100 mL), and the combined organic extracts were washed with water (3 X 100 mL) and brine (1 X 100 mL). The crude product was purified by column chromatography (CH2Cl?) to yield 77 (1.0 g, 51%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) S: 8.05 (m, IH, aromatics), 7.69 (m, IH, aromatics), 7.51 (m, IH, aromatics), 7.39-7.21 (m, 5H, aromatics), 6.92 (d, IH, CH=CH, J= 14.8 Hz ), 6.73 (d, IH, CH=CH, J= 14.8 Hz ). 13C NMR (75 MHz, CDCl3) δ: 188.1 , 134.6, 133.6, 132.8, 132.3, 131.1, 130.8, 130.5, 130.1, 129.8, 129.6, 129.3, 126.8, 15.0, 124.8, 1 15.3; 19F(CDCl3, 283 MHz) δ: -1 10.1 (s, 2F). HRMS calcd for Ci6H10F2ONa (M+Na): 279.0597. Found: 279.1032.
Synthesis of 78. To a stirring solution of 77 (1.0 g, 4 mmol) in EtOH (30 mL) was added NaBH4 (0.3 g, 8 mmol) over 5 minutes. The reaction was kept at room temperature for 2 hours, and then it was quenched with water (100 ml). The aqueous layer was extracted with CH2Cl2 (3 X 100 mL), and the combined organic extracts were washed with water (100 mL) and brine (100 mL). The crude product was purified by column chromatography (hexane/EtOAc, 5/1 , v/v) to yield 78 (0.78 g, 78%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) 6: 7.02-7.74 (m, 8H, aromatics), 6.94 (d, IH, J= 12.3 Hz, CH=CH), 6.85 (d, IH, J= 12.3 Hz, CH=CH), 5.60 (m, 1Η, Ph-CH-OΗ), 2.82 (m, IH5OH). 13C NMR (75 MHz, CDCl3) δ: 136.0, 135.9, 135.0, 133.8, 131.2, 130.6, 130.0,128.1,127.9, 126.8, 124.6, 121.5, 121.4; 19F(CDCl3, 282 MHz) δ: -69.8 (d, IF, J=259.4 Hz), - 1 1 1.7 (dd, IF, J= 259.4, 21.4 Hz). HRMS calcd for C6Hi2F2ONa (M+Na): 281.0754. Found: 281.1 122.
Synthesis of 79. To a stirring solution of bromine (0.16 g, 1.0 mmol) in CHCl3 (10 mL) was added dropwise to a solution of 78 (0.25 g, 1.0 mmol) in CHCl3 (10 mL) at 00C. The reaction mixture was stirred at room temperature for 12 hours until the reaction was complete (monitored by TLC). The resulting mixture was washed with aqueous saturated sodium thiosulfate solution (10 mL), and dried (MgSO4). The solvents were evaporated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/EtOAc, 8/1 , v/v) to afford 79 (0.19 g, 46%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) δ: 6.96-7.82 (m, 8H, aromatics), 5.62 (d, IH, J= 10.8 Hz, Ph-CH-Br), 5.17 (d, 1Η, J= 10.8 Hz, PhHCH). 13C NMR (75 MHz, CDCl3) δ: 132.7,132.4, 131.0,130.8, 129.2, 129.0, 128, 128.4,128.3, 127.8, 125.4, 123.8, 1 11.5, 57.3, 56.5, 50.5; 19F(CDCl3, 282 MHz) δ: -98.8 (d, 1F, J=341.1 Hz), - 1 10.4 (d, 1F, J=341.1 Hz).
Synthesis of 66. To a stirring solution of 79 (40 mg, 0.1 mmol) in tetrahydrofuran (10 mL) was added dropwise f-BuOK in tetrahydrofuran (2.0 M), (0.5 mL) under an atmosphere of Argon at room temperature. The reaction mixture was stirred for 6 hours at room temperature, after which it was poured into ice water (10 mL) and the resulting mixture was extracted with CH2Cl2 (2 X 50 mL). The combined extracts were washed with water, brine and dried (MgSO4) and the solvents were evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/EtOAc, 5/1, v/v) to afford 66 (10 mg, 49%; Scheme 10; Figure 19). 1H NMR (CDCl3, 300 MHz) δ: 7.19-7.92 (m, 8H, aromatics), 5.39 (d, IH3 J= 23.4, 10.8 Hz, -CH-OH). 13C
NMR (75 MHz, CDCl3) δ:131.3, 129.5, 129.4, 128.4, 127.9, 127.5, 126.8, 126.6, 125.9, 125.7, 124.8, 124.5, 120.3, 107.3, 82.8; 19F(CDCl3, 282 MHz) O: -91.5 (d, IF, J=253.8 Hz). -103.4 (d, IF, J=253.8 Hz).
(6H- 11, 12-didehydro-dibenzofa, e]cycloocten-5-ylideneaminooxy)-acetic acid (68). A solution of 6H-1 l,12-didehydro-dibenzo[α,e]cyclooctatrien-5-one 67 (21.8 mg, 0.1 mmol) and carboxymethyl)hydroxylamine hemihydrochloride (21.8 mg, 0.2 mmol) in 1 : 1 :0.02 v/v/v MeOΗ/CΗ2Cl2/ΗOAc (8 ml) was stirred at room temperature for 2 days. The solvent was removed under reduced pressure, and the residue was purified by flash chromatography on silica gel (EtOAc) to give the product as white solid (17.8 mg, 61%). 1H NMR (300 MHz, CDCl3): δ 7.54 (IH, d, J= 7.4 Hz), 7.46 (IH, d, J = 7.4 Hz), 7.18-7.39 (6 H, m), 4.53 (2 H, m), 4.23 (1 H, d, J = 12.8 Hz), 3.16 (1 H, d, J= 12.8 Hz). 13C NMR (75 MHz, CDCl3): δ 175.2 & 173.6, 154.1 153.2, 130.7,129.5, 19.3, 129.2, 129.1, 128.1 , 128.0, 127.1 , 126.9, 125.5, 125.2, 122.7, 1 13.9, 1 1 1.2, 84.7, 68.3 & 67.1, 35.0 & 33.2. MALDI HRMS: m/z 314.0810 [M+Na+]. Calcd for C8H13NNaO3: 314.0793.
Modification of macromolecules and nano-material using cycloadditions with 4- dibenzocyclooctynol
The Cu(I) catalyzed 1 ,3-dipolar cycloaddition of azides with terminal alkynes to give stable triazoles has been employed for tagging a variety of biomolecules including proteins, nucleic acids, lipids, and saccharides. This reaction has also been used to modify polymers and nanoscale materials. Potential difficulties to remove Cu(I), which is highly cytotoxic, complicates the use of the 1,3-dipolar cycloaddition for conjugation of compounds or material for biological or medical application. The use of 4-dibenzocyclooctynol instead of a terminal alkyne for cycloadditions with azides should overcome this problem.
To demonstrate the use of 4-dibenzocyclooctynol in bioconjugation, co- block polymers 83 and 84 were prepared. These materials were employed to form organomicelles in water and it was shown that 4-dibenzocyclooctyne fragment of these materials can be reacted was with azido containing molecules (Figure 2OA, B). It is well known that co-block polymers composed of a polyester and polyethyleneglycol fragment self-assemble in water to form organomicelles These nano-mateπals have attracted attention as drug delivery devises. Deπvatization of organomicelles with, for example, tissue or tumor targeting moieties may lead to smart drug delivery devises. In addition, modification of organomicelles with fluorescent tags or MRI reagents, such as biotin, will be valuable for imaging purposes (Figure 20C)
Copolymeπzation of polyethylene glycol methyl ether (81) or azide (82) (MW -2000 Da) with caprolactone in the presence of a catalytic amount of SnOct gave copolymers 83 and 84, respectively (Scheme 1 1 , Figure 21 ). The azido fragment of 84 was reduced with tπphenylphosphine and the amine of the resulting polymer 85 was reacted with 86 and 87 to give dibenzocyclooctyl derivatives 88 and 89, respectively. A mixture of 83 and 88 or 89 (9/1, w/w) dissolved in a small amount of THF were added to water Cryo-TEM showed that organomicelles that have a diameter of approximately 4OA were formed The resulting micelles were incubated with azido-contaimng saccharide 90 and after a reaction time of 24 hours, unreacted saccharide was lemoved by dialysis. The micelles were analyzed for sugar content by hydrolysis with TFA followed by quantification by high pH anion exchange chromatography. It was established that approximately 45% of the cyclooctynes were modified by saccharides It is to be expected that compound 84 can also be employed for the formation of micelles and the azido moieties of the resulting azides employed in cycloaddition with compounds modified with a dibenzocyclooctyl fragment
Experimental
Synthesis of PEG44-b-PCL2β 83 PEG45-O-PCL23 block copolymers were synthesized as reported. A predetermined volume (12.0 mL) of ε-caprolactone monomer was placed in a flask containing an amount (9.0 g) of PEG 81 under an argon atmosphere Then, a drop of SnOct was added After cooling to liquid- nitrogen temperature, the flask was evacuated for 12 hours, sealed off, and kept at 1300C for 24 hours The synthesized polymers were dissolved in THF, recovered by precipitation by cold hexane, and dried under vacuum at room temperature. The degree of polymerization of the PCL was calculated by 1H NMR relative to the degree of polymerization of the PEG 81.
Synthesis of azιde-PEG44-b-PCL26 84. Azide-PEG-Z?-PCL was synthesized by a one-pot catiomc πng opening polymerization at 1300C under a stream of argon adopting a previously reported method for the preparation of PEG-ό-PCL with some modifications. Briefly, a predetermined volume (3.3 mL) of ε-caprolactone monomer was placed in a flask containing a preweighed amount (2.5 g) of azide-PEG-OH 82 under a nitrogen atmosphere. Then a drop of SnOct was added. After cooling to liquid-nitrogen temperature, the flask was evacuated, sealed off, and kept at 1300C for 24 hours. The synthesized polymers were then dissolved in THF, recovered by precipitation into cold hexane, and dried under vacuum at room temperature. The number average molecular weight (Mn) of azide-PEO44-έ>-PCL26 84 block copolymer was determined by 1H NMR. Synthesis of amιne-PEG44-b-PCL26 85. Pd/C (10 wt. % on activated carbon, 50 mg) was added to a solution of azide-PEG44-ό-PCL2684 (200 mg)in EtOH and HOAc (50 μL), after which H2 was bubbled through the solution for 1 hr followed by stirring under an H? atmosphere for 16 hours. The mixture was filtered, concentrated in vacuum The residues were then dissolved in THF, recovered by precipitation into cold hexane, and dried under vacuum at room temperature to afford amine-PEG44-&-PCL26 85
DIDO-PEO-PCL copolymer (88) To a stirred solution of carbonic acid 5,6-dihydro-l l ,12-didehydro-dibenzo[a,e]cycloocten-5-yl ester 4-nitro-phenyl ester 86 (1 1.6 mg, 0.03 mmol) and copolymer 85 (98 mg, 0.02 mmol) in CH2Cl2 (10 ml) at room temperature was added Et3N (0.014 ml, 0.1 mmol). The reaction mixture was stirred overnight at room temperature, after which the solvent was removed under reduced pressure. The residue was purified by size exclusion chromatography (SEC) on LH-20 column (1 : 1 v/v MeOH/CH2Cl2) to give the product as yellowish solid (101 mg, 97%).
DIDO-PEO-PCL copolymer (89). To a stirred solution of (5,6-Dihydro- 1 l,12-didehydro-dibenzo[α,e]cycloocten-5-yloxy)-acetic acid 88 (8.3 mg, 0.03 mmol) and copolymer 85 (98 mg, 0.02 mmol) in DMF (15 ml) at room temperature was added HATU coupling reagent (1 1.4 mg, 0.03 mmol) and DIPEA (0.0104 ml, 0.06 mmol). The reaction mixture was stirred for 5 hours at room temperature, after which the solvent was removed under reduced pressure. The residue was purified by SEC chromatography on LH-20 column (1 : 1 v/v MeOH/CH2Cl2) to give the product as yellowish solid (100 mg, 96%).
Cy do additions of dibenzocyclooctanol with various 1,3-dipoles.
It has been found that 4-dibenzocyclooctynol can react in the absence of catalyst or promoter at ambient temperature with 1 ,3-dipoles such as nitrones and acyl diazo derivatives, which can provide unique opportunities for bioconjugation reactions.
Nitrones were prepared by a modification of the procedures disclosed in Dicken et al, J. Org. Chem. 1982, 47, 2047-2051 ; and Inouye et al., Bull. Chem. Soc. Jpn. 1983, 56, 3541-3542. N-alkylhydroxylamine hydrochloride (10.0 mmol), glyoxylic acid (0.92 g, 10.0 mmol), and sodium bicarbonate (1.68 g, 20.0 mmol) in toluene (20 ml) were stirred at room temperature overnight. The solid was filtered and the filtrate was concentrated to afford the nitrone. This nitrone was then used directly without any purification.
Thus nitrones 91-95 were mixed with 4-dibenzocyclooctynol and after a reaction time of 3 minutes to 3.5 hours the corresponding 2,3-dihydro-issoxazole cycloaddition products were isolated in almost a quantitative yield. It can be seen in figure 18 that the chemical nature of the nitrone has a dramatic impact of the reaction rate. In particular electron poor nitrones 93 and 94 react at much faster rates than corresponding azides.
Experimental
General method for calculating second order rate constants by NMR.
Substrates were dissolved separately in the appropriate solvent and mixed 1 : 1 at 6 mM concentrations. Percent conversion was monitored both by disappearance of starting material and appearance of the two regioisomeric products as determined by integration at multiple chemical shifts. Second order rate constants for the reaction were determined by plotting the 1 /[substrates] versus time and analysis by linear regression. Second order rate constants correspond to one half of the determined slope. The complete disclosure of all patents, patent applications, and publications, and electronically available material (e.g., GenBank amino acid and nucleotide sequence submissions; and protein data bank (pdb) submissions) cited herein are incorporated by reference. The foregoing detailed description and examples have been given for clarity of understanding only. No unnecessary limitations are to be understood therefrom. The invention is not limited to the exact details shown and described, for variations obvious to one skilled in the art will be included within the invention defined by the claims.

Claims

What is claimed is:
1. An alkyne of the formula:
Figure imgf000055_0001
Formula I,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group;
X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group.
2. The alkyne of claim 1 wherein each R1 represents hydrogen.
3. The alkyne of claim 1 or 2 wherein each R~ represents hydrogen.
4. The alkyne of any one of the preceding claims wherein R3 comprises a covalently bound organic dye.
5. The alkyne of claim 4 wherein the organic dye is a fluorescent dye.
6. The alkyne of any one of the preceding claims wherein X represents C=N-OR3 and R3 represents an organic group having the formula -(CH2^C(O)Y, wherein: a is 1-3; Y represents OH or NHR5; and
R5 represents hydrogen or a biotinylation product of a primary amine- contaming organic group.
7. The alkyne of claim 6 wherein the biotinylation product is the biotinylation product of a pπmary amme-contammg gioup of the formula -(CH2CH2θ)b(CH2)c-Ld-(CH2CH2θ)e(CH2)fNH2 and/or -(CD2CD2O)b(CD2)c-Ld- (CD2CD2COC(CD2)INH2, wherein b = 0 to 100, c = 0 to 100; d = 0 to 100, e = 0 to 100, f = 0 to 100; and L is an optional cleavable linker.
8. The alkyne of claim 7 wherein the cleavable linker, if present, is a -disulfide.
9 The alkyne of any one of claims 1 to 5 wherein X represents CHOR3 and R is selected from the group consisting of an alkyl group, an aryl group, an alkaryl group, and an aralkyl group
10 The alkyne of claim 9 wherein R3 represents -C(O)Z, wherein. Z represents an alkyl group, OR6, or NHR7,
R6 and R7 are each independently selected from the group consisting of an alkyl group, an aiyl group, an alkaryl group, and an aralkyl group.
1 1 The alkyne of claim 10 wherein R7 is a biotinylation product of a primary amme-contammg organic gioup
12. The alkyne of claim 1 1 wherein the biotinylation product is the biotinylation product of a pπmary amme-contammg group of the formula -(CH2CH2O)b(CH2)c-Ld-(CH2CH2O)e(CH2)fNH2 and/or -(CD2CD2O)b(CD2)c-Ld- (CD2CD2O)e(CD2)tNH2, wherein b = 0 to 100, c = 0 to 100; d = 0 to 100, e = 0 to 100, f = 0 to 100, and L is an optional cleavable linker.
13. The alkyne of claim 12 wherein the cleavable linker, if present, is a disulfide.
14. The alkyne of any one of claims 1 to 5 wherein R3 represents a polymeric or a copolymeric group.
15. The alkyne of claim 14 wherein the copolymeric group comprises a hydrophilic segment and a hydrophobic segment.
16. The alkyne of claim 14 or 15 wherein the copolymeric group comprises a fragment of the formula -[CH2CH2O]11-[C(O)(CH2)SO]111-H, wherein n = O to 100 and m = 0 to 100.
17. An alkyne of the formula:
Figure imgf000057_0001
18. An alkyne of the formula:
Figure imgf000057_0002
Formula V.
19. An alkyne of the formula:
Figure imgf000058_0001
Formula VI.
20. An alkyne of the formula:
Figure imgf000058_0002
Formula VII, wherein RJ represents hydrogen or an organic group.
21. A composition comprising a blend of: an alkyne according to any one of claims 14 to 16; and a polymer or a copolymer.
22. The composition of claim 21 wherein the copolymer comprises a hydrophilic segment and a hydrophobic segment.
23. The composition of claim 21 or 22 wherein the copolymer is of the ffoorrmmuullaa RR99OO--[[CCHH22CCHH22OO]]pp--[[CC((OO))((CCHH;2)5O]o-H, wherein R9 represents an alkyl group, p = 0 to 100, and o = 0 to 100.
24. The composition of claim 23 wherein R9 represents methyl.
25. The composition of any one of claims 21 to 24 wherein the composition forms a copolymer micelle.
26. A method for controlling the delivery of drugs, the method comprising: combining at least one 1, 3 -dipole- functional drug with a copolymer micelle according to claim 25 comprising an alkyne; and allowing the at least one 1,3 -dipole- functional drug and the copolymer micelle comprising the alkyne to react under conditions effective to form a heterocyclic compound.
27. A compound of the formula:
Figure imgf000059_0001
Formula II Formula III,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl -ClO organic group; each R" is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group;
X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; each R > 3 a „nd j r R>4 independently represents hydrogen or an organic group; and
R' represents an organic group.
28. A compound of the formula:
Figure imgf000060_0001
Formula VIII
Figure imgf000060_0002
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group;
X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; each R3 and R4 independently represents hydrogen or an organic group; and
R represents an organic group.
29. The compound of claim 27 or 28 wherein R represents a biomolecule.
30. The compound of claim 29 wherein the biomolecule is selected from the group consisting of peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, polysaccharides, and combinations thereof.
31. A compound of the formula:
Figure imgf000061_0001
Formula X Formula XI,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R~ is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-Cl O organic .group;
X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3, R4, and R10 independently represents hydrogen or an organic group, with the proviso that at least one R10 represents an organic group.
32. A compound of the formula:
Figure imgf000061_0002
Formula XII Formula XIII,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R" is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3, R4, and R10 independently represents hydrogen or an organic group, with the proviso that at least one R10 represents an organic group.
33. The compound of claim 31 or 32 wherein the at least one R1 representing an organic group represents a biomolecule.
34. The compound of claim 33 wherein the biomolecule is selected from the group consisting of peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, polysaccharides, and combinations thereof.
35. A method of preparing a heterocyclic compound, the method comprising: combining at least one 1 ,3-dipole-functional compound with at least one alkyne according to any one of claims 1 to 20; and allowing the at least one 1,3-dipole-functional compound and the at least one alkyne to react under conditions effective to form the heterocyclic compound.
36. The method of claim 35 wherein the 1 ,3-dipole-functional compound is selected from the group consisting of an azide-functional compound, a nitrile oxide-functional compound, a nitrone-functional compound, an azoxy-functional compound, an acyl diazo-functional compound, and combinations thereof.
37. The method of claim 35 or 36 wherein conditions effective to form the heterocyclic compound comprise the substantial absence of added catalyst.
38. The method of any one of claims 35 to 37 wherein the reaction takes place within or on the surface of a living cell.
39. The method of any one of claims 35 to 38 wherein the at least one 1 ,3- dipole-functional compound comprises a 1,3-dipole-functionalized biomolecule.
40. The method of any one of claims 35 to 39 wherein the at least one 1 ,3- dipole-functional compound comprises a detectable label.
41. The method of claim 40 further comprising detecting the heterocyclic compound.
42. The method of claim 40 or 41 wherein the detectable label is an affinity label.
43. The method of claim 42 further comprising isolating the heterocyclic compound using affinity binding.
44. An alkyne comprising: a cleavable linker fragment comprising at least two ends; an alkyne fragment attached to a first end of the cleavable linker fragment; and a biotinylated fragment attached to a second end of the cleavable linker fragment.
45. The alkyne of claim 44 wherein the alkyne fragment comprises a strained, cyclic alkyne fragment.
46. The alkyne of claim 44 or 45 further comprising at least one heavy mass isotope.
47. The alkyne of any one of claims 44 to 46 further comprising at least one detectable label.
48. The alkyne of claim 47 wherein the detectable label comprises a fluorescent label.
49. A heterocyclic compound formed by the reaction of at least one alkyne according to one of claims 44 to 48 with at least one 1,3 dipole- functional compound.
50. The heterocyclic compound of claim 49 wherein the 1 ,3-dipole- functional compound is selected from the group consisting of an azide- functional compound, a nitrile oxide-functional compound, a nitrone-functional compound, an azoxy-functional compound, an acyl diazo-functional compound, and combinations thereof.
51. A method of preparing a heterocyclic compound, the method comprising: combining at least one 1 ,3-dipole-functional compound with at least one alkyne according to any one of claims 44 to 48; and allowing the at least one 1 ,3-dipole-functional compound and the at least one alkyne to react under conditions effective to form the heterocyclic compound.
52. The method of claim 51 wherein the 1,3-dipole-functional compound is selected from the group consisting of an azide-functional compound, a nitrile oxide-functional compound, a nitrone-functional compound, an azoxy-functional compound, an acyl diazo-functional compound, and combinations thereof.
53. The method of claim 51 or 52 wherein conditions effective to form the heterocyclic compound comprise the substantial absence of added catalyst.
54. The method of any one of claims 51 to 53 wherein the reaction takes place within or on the surface of a living cell.
55. The method of any one of claims 51 to 54 wherein the at least one 1,3- dipole-functional compound comprises a 1 ,3 -dipole- functionalized biomolecule.
56. The method of any one of claims 51 to 55 further comprising contacting the heterocyclic compound with a compound that binds biotin.
57. The method of claim 56 wherein the compound that binds biotin comprises avidin and/or streptavidin.
58. The method of any of claims 51 to 57 further comprising detecting the heterocyclic compound.
59. A method of preparing an alkyne, the method comprising: brominating an alkene of the formula:
Figure imgf000065_0001
Formula XIV
to provide a dibromide of the formula:
Figure imgf000065_0002
Formula XV;
and dehydrobrominating the dibromide of Formula XV to provide the alkyne of the formula:
Figure imgf000066_0001
Formula I,
wherein: each R1 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group; each R2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, alkoxy, nitrate, nitrite, sulfate, and a Cl-ClO organic group;
X represents C=O, C=N-OR3, C=N-NR3R4, CHOR3, or CHNHR3; and each R3 and R4 independently represents hydrogen or an organic group.
60. A substrate having on the surface thereof an alkyne according to any one of claims 1 to 20.
61. The substrate of claim 60 wherein the substrate is in the form of a resin, a gel, nanoparticles, or combinations thereof.
62. The substrate of claim 60 or 61 wherein the substrate is a three- dimensional matrix.
63. The substrate of any one of claims 60 to 62 wherein the X group of the alkyne represents a point of attachment to the surface of the substrate.
64. A method of immobilizing a biomolecule on a substrate, the method comprising: providing a substrate according to any one of claims 60 to 63; contacting the substrate with a 1, 3 -dipole- functional biomolecule under conditions effective to form a heterocyclic compound.
65. The method of claim 64 wherein the 1,3-dipole-functional biomolecule is selected from the group consisting of an azide-functional biomolecule, a nitrile oxide-functional biomolecule, a nitrone-functional biomolecule, an azoxy- functional biomolecule, an acyl diazo-functional biomolecule, and combinations thereof.
66. The method of claim 64 or 65 wherein the biomolecule is selected from the group consisting of peptides, proteins, glycoproteins, nucleic acids, lipids, saccharides, oligosaccharides, polysaccharides, and combinations thereof.
67. An article comprising an immobilized biomolecule prepared by the method of any one of claims 64 to 66.
68. An article comprising a protein immobilized on a three-dimensional matrix.
69. A method for immobilizing a cell, the method comprising: providing a substrate according to any one of claims 60 to 63; contacting the substrate with a cell comprising a 1,3-dipole-functional biomolecule under conditions effective to form a heterocyclic compound.
PCT/US2008/084345 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds WO2009067663A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2010535090A JP5498952B2 (en) 2007-11-21 2008-11-21 Process for reacting alkynes with alkynes and 1,3-dipolar functional compounds
CN200880125596.0A CN101925366B (en) 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US12/743,632 US8133515B2 (en) 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
DK08852196.8T DK2222341T3 (en) 2007-11-21 2008-11-21 AND METHODS alkynes of reacting alkynes of 1,3-dipole FUNCTIONAL COMPOUNDS
EP08852196.8A EP2222341B1 (en) 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US13/418,676 US8940859B2 (en) 2007-11-21 2012-03-13 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US14/591,290 US9227943B2 (en) 2007-11-21 2015-01-07 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US14/967,896 US9725405B2 (en) 2007-11-21 2015-12-14 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US15/657,601 US9932297B2 (en) 2007-11-21 2017-07-24 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US402107P 2007-11-21 2007-11-21
US61/004,021 2007-11-21
US767407P 2007-12-14 2007-12-14
US61/007,674 2007-12-14
US13706108P 2008-07-25 2008-07-25
US61/137,061 2008-07-25

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/743,632 A-371-Of-International US8133515B2 (en) 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US13/418,676 Continuation US8940859B2 (en) 2007-11-21 2012-03-13 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds

Publications (2)

Publication Number Publication Date
WO2009067663A1 true WO2009067663A1 (en) 2009-05-28
WO2009067663A8 WO2009067663A8 (en) 2009-07-23

Family

ID=40510055

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/084345 WO2009067663A1 (en) 2007-11-21 2008-11-21 Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds

Country Status (6)

Country Link
US (5) US8133515B2 (en)
EP (2) EP2222341B1 (en)
JP (4) JP5498952B2 (en)
CN (2) CN104529711B (en)
DK (2) DK2222341T3 (en)
WO (1) WO2009067663A1 (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102040530A (en) * 2009-10-15 2011-05-04 原子能与替代能源委员会 Process for the functionalization of biological molecules
WO2011118394A1 (en) * 2010-03-26 2011-09-29 Jnc株式会社 Cyclic compound, method for producing cyclic compound, and method for modifying biological molecule
WO2011136645A1 (en) 2010-04-27 2011-11-03 Stichting Katholieke Universiteit, More Particularly Radboud University Nijmegen Fused cyclooctyne compounds and their use in metal-free click reactions
WO2012016048A1 (en) 2010-07-28 2012-02-02 Life Technologies Corporation Anti- viral azide-containing compounds
WO2012016044A1 (en) 2010-07-28 2012-02-02 Life Technologies Corporation Anti-viral azide containing compounds
WO2012047663A3 (en) * 2010-09-27 2012-05-31 University Of Georgia Reaserch Foundation, Inc. Methods including latent 1,3-dipole-functional compounds and materials prepared thereby
US20120208722A1 (en) * 2010-10-19 2012-08-16 Richard Dluhy Surface enhanced raman spectroscopy platforms and methods
US8258347B2 (en) 2009-02-19 2012-09-04 University Of Georgia Research Foundation, Inc. Cyclopropenones and the photochemical generation of cyclic alkynes therefrom
WO2012121973A1 (en) 2011-03-04 2012-09-13 Life Technologies Corporation Compounds and methods for conjugation of biomolecules
WO2012134925A1 (en) 2011-03-25 2012-10-04 Life Technologies Corporation Heterobifunctional esters for use in labeling target molecules
WO2012142003A2 (en) 2011-04-15 2012-10-18 Life Technologies Corporation Chemical ligation
EP2532639A1 (en) * 2011-06-09 2012-12-12 ModiQuest B.V. Method for preparing a reactive coating
US8426649B2 (en) 2009-02-19 2013-04-23 University Of Georgia Research Foundation, Inc. Cyclopropenones and the photochemical generation of cyclic alkynes therefrom
US20130202652A1 (en) * 2010-07-30 2013-08-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2014009426A3 (en) * 2012-07-13 2014-04-03 Innate Pharma Screening of conjugated antibodies
WO2014066733A2 (en) 2012-10-25 2014-05-01 Life Technologies Corporation Methods and compositions for enzyme-mediated site-specific radiolabeling of glycoproteins
WO2014111344A1 (en) * 2013-01-15 2014-07-24 Novartis Ag Cycloalkyne derivatized saccharides
WO2014189370A1 (en) 2013-05-24 2014-11-27 Stichting Katholieke Universiteit Substituted azadibenzocyclooctyne compounds and their use in metal-free click reactions
US8912322B2 (en) 2010-07-29 2014-12-16 University Of Georgia Research Foundation, Inc. Aza-dibenzocyclooctynes and methods of making and using same
US8940859B2 (en) 2007-11-21 2015-01-27 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US8962580B2 (en) 2008-09-23 2015-02-24 Alnylam Pharmaceuticals, Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
WO2015057066A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Glycoengineered antibody, antibody-conjugate and methods for their preparation
WO2015057064A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Modified glycoprotein, protein-conjugate and process for the preparation thereof
WO2015057065A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Glycoengineered antibody, antibody-conjugate and methods for their preparation
US9427478B2 (en) 2013-06-21 2016-08-30 Innate Pharma Enzymatic conjugation of polypeptides
US9717803B2 (en) 2011-12-23 2017-08-01 Innate Pharma Enzymatic conjugation of polypeptides
WO2017137457A1 (en) 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
WO2017137458A1 (en) 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
US9803201B2 (en) 2015-03-17 2017-10-31 Arrowhead Pharmaceuticals, Inc. Disulfide-containing alkyne linking agents
EP2911699B1 (en) 2012-10-23 2017-11-15 SynAffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof
WO2017218891A1 (en) 2016-06-17 2017-12-21 Life Technologies Corporation Site-specific crosslinking of antibodies
US10036010B2 (en) 2012-11-09 2018-07-31 Innate Pharma Recognition tags for TGase-mediated conjugation
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
WO2019169307A1 (en) 2018-03-02 2019-09-06 Life Technologies Corporation Novel quencher and reporter dye combinations
WO2019240219A1 (en) 2018-06-14 2019-12-19 持田製薬株式会社 Novel crosslinked alginic acid
WO2019243672A1 (en) 2018-06-19 2019-12-26 Glykos Biomedical Oy Conjugate
WO2020002765A1 (en) 2018-06-29 2020-01-02 Glykos Biomedical Oy Conjugates
US10550386B2 (en) * 2010-01-28 2020-02-04 Alnylam Pharmaceuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
US10611824B2 (en) 2013-03-15 2020-04-07 Innate Pharma Solid phase TGase-mediated conjugation of antibodies
US10821196B2 (en) 2008-04-30 2020-11-03 Siemens Medical Solutions Usa, Inc. Substrate based PET imaging agents
WO2020262642A1 (en) 2019-06-28 2020-12-30 持田製薬株式会社 Transplantation device using chemically crosslinked alginic acid
US10905678B2 (en) 2014-04-08 2021-02-02 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glycoconjugates and methods
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
WO2021116037A1 (en) 2019-12-09 2021-06-17 F. Hoffmann-La Roche Ag Dicationic fluorescent dyes
WO2021123506A1 (en) 2019-12-18 2021-06-24 Glykos Biomedical Oy Stabile conjugate
US11168085B2 (en) 2014-01-24 2021-11-09 Synaffix B.V. Process for the cycloaddition of a hetero(aryl) 1,3-dipole compound with a (hetero)cycloalkyne
US11312942B2 (en) 2016-06-08 2022-04-26 Sony Corporation Material-fixing substrate and method for producing same, and material-fixing agent used for material-fixing substrate
US11591408B2 (en) 2017-11-09 2023-02-28 National Research Council Of Canada Antibody glycoconjugates and methods of production and use
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012054784A1 (en) 2010-10-20 2012-04-26 Li-Cor, Inc. Fluorescent imaging with substituted cyanine dyes
WO2014004278A1 (en) * 2012-06-26 2014-01-03 The Curators Of The University Of Missouri Photocleavable drug conjugates
WO2014022535A1 (en) * 2012-07-31 2014-02-06 The University Of Akron Polymeric structures containing strained cycloalkyne functionality for post-fabrication azidealkyne cycloaddition functionalization
CA2883168A1 (en) * 2012-08-21 2014-02-27 Academia Sinica Benzocyclooctyne compounds and uses thereof
EP4036579A1 (en) * 2013-03-15 2022-08-03 Arizona Board of Regents on behalf of Arizona State University Biosensor microarray compositions and methods
JP6245707B2 (en) * 2013-03-28 2017-12-13 静岡県公立大学法人 Method for producing PEGylated bioactive substance labeled with [18F] F or fluorescent dye, and pharmacokinetic analysis thereof
EP2818867A1 (en) 2013-06-27 2014-12-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies conjugated to at least one nucleic acid molecule and their use in multiplex immuno-detection assays
JP6327547B2 (en) * 2013-08-02 2018-05-23 国立研究開発法人理化学研究所 New compounds and their use
JP2017500315A (en) 2013-12-12 2017-01-05 ザ ユニヴァーシティ オブ ジョージア リサーチファウンデーション, インク. Prodrugs for release of cisplatin and cyclooxygenase inhibitors
CN103983764B (en) * 2014-04-17 2016-06-29 深圳先进技术研究院 Cell is carried out the methods and applications of labeled in situ
EP3411475A4 (en) 2016-02-06 2019-09-11 President and Fellows of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
WO2017191817A1 (en) 2016-05-02 2017-11-09 味の素株式会社 Azide group-containing fc protein
CN115305229A (en) 2016-07-13 2022-11-08 哈佛学院院长等 Antigen presenting cell mimetic scaffolds and methods of making and using same
US11951165B2 (en) 2016-12-30 2024-04-09 Vaxcyte, Inc. Conjugated vaccine carrier proteins
WO2019092148A1 (en) 2017-11-10 2019-05-16 Innate Pharma Antibodies with functionalized glutamine residues
WO2019151128A1 (en) * 2018-02-05 2019-08-08 国立大学法人 宮崎大学 Cell labeling agent and cell labeling kit
CA3132959A1 (en) 2019-03-08 2020-09-17 AbTis Co., Ltd. Site-specific antibody conjugation and antibody-drug conjugate as specific embodiment thereof
GB201913598D0 (en) * 2019-09-20 2019-11-06 Univ Birmingham Labelling of biomolecules
WO2021155297A1 (en) * 2020-01-29 2021-08-05 President And Fellows Of Harvard College Methods for labeling and targeting cells
JP2023512036A (en) 2020-01-31 2023-03-23 イナート・ファルマ・ソシエテ・アノニム cancer treatment
CN111620848A (en) * 2020-04-29 2020-09-04 华东师范大学 Tricyclic condensed aromatic system compound containing pyran and medium ring skeleton and synthesis and application thereof
CA3196198A1 (en) 2020-11-25 2022-06-02 Manel KRAIEM Treatment of cancer
US11377424B1 (en) * 2021-05-27 2022-07-05 Massachusetts Institute Of Technology Cyclooctynes for click chemistry
WO2022250679A1 (en) * 2021-05-27 2022-12-01 Massachusetts Institute Of Technology Cyclooctynes for click chemistry
CN116102403A (en) * 2021-11-11 2023-05-12 中国科学院福建物质结构研究所 Preparation method of dienol compound
WO2023227660A1 (en) 2022-05-25 2023-11-30 Innate Pharma Nectin-4 binding agents
CN114853556B (en) * 2022-06-14 2023-08-29 绍兴文理学院 New synthesis method of 5H-dibenzo [ a, d ] cycloheptene skeleton

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007039858A2 (en) * 2005-10-04 2007-04-12 Koninklijke Philips Electronics N.V. Targeted imaging and/or therapy using the [3+2] azide-alkyne cycloaddition

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3711489A (en) * 1971-03-31 1973-01-16 Pfizer Certain 8,9-dihydro(3,4,7,8)cycloocta(1,2-d)imidazoles
SE403775B (en) * 1973-11-21 1978-09-04 Du Pont NEW ETHENOANTHRACENE DERIVATIVES (BENZENOBENZ (E) ISOINDLE DERIVATIVES) INTENDED FOR USE AS INHIBITORS IN FREE RADIUM POLYMERIZATION OF VINYL COMPOUNDS
US5723289A (en) 1990-06-11 1998-03-03 Nexstar Pharmaceuticals, Inc. Parallel selex
DE69108356T2 (en) 1990-11-17 1995-07-20 Nihon Nohyaku Co Ltd Hydrazone derivatives, process for their preparation and their use.
US5767259A (en) 1994-12-27 1998-06-16 Naxcor Oligonucleotides containing base-free linking groups with photoactivatable side chains
SE9500342D0 (en) 1995-01-31 1995-01-31 Marek Kwiatkowski Novel chain terminators, the use thereof for nucleic acid sequencing and synthesis and a method of their preparation
US5843650A (en) 1995-05-01 1998-12-01 Segev; David Nucleic acid detection and amplification by chemical linkage of oligonucleotides
US5874532A (en) 1997-01-08 1999-02-23 Nexstar Pharmaceuticals, Inc. Method for solution phase synthesis of oligonucleotides and peptides
EP0968223B1 (en) 1997-01-08 2016-12-21 Sigma-Aldrich Co. LLC Bioconjugation of macromolecules
US7427678B2 (en) 1998-01-08 2008-09-23 Sigma-Aldrich Co. Method for immobilizing oligonucleotides employing the cycloaddition bioconjugation method
ATE356222T1 (en) 2000-10-06 2007-03-15 Univ Columbia MASSIVE PARALLEL METHOD FOR DECODING DNA AND RNA
US7375234B2 (en) 2002-05-30 2008-05-20 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
US7597876B2 (en) * 2007-01-11 2009-10-06 Immunomedics, Inc. Methods and compositions for improved F-18 labeling of proteins, peptides and other molecules
EP3002289B1 (en) 2002-08-23 2018-02-28 Illumina Cambridge Limited Modified nucleotides for polynucleotide sequencing
WO2004055160A2 (en) 2002-12-13 2004-07-01 The Trustees Of Columbia University In The City Of New York Biomolecular coupling methods using 1,3-dipolar cycloaddition chemistry
US20080075661A1 (en) 2004-10-07 2008-03-27 Koninklijke Philips Electronics, N.V. Compounds, Kits and Methods for Use in Medical Imaging
US20060147963A1 (en) 2004-12-30 2006-07-06 Affymetrix, Inc. Detection of polynucleotides on nucleic acid arrays using azido-modified triphosphate nucleotide analogs
EP2266996A3 (en) 2005-05-02 2011-06-15 baseclick GmbH New labelling strategies for the sensitive detection of analytes
US8114636B2 (en) 2006-02-10 2012-02-14 Life Technologies Corporation Labeling and detection of nucleic acids
EP2222341B1 (en) * 2007-11-21 2015-02-25 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
JP2010122071A (en) * 2008-11-19 2010-06-03 Okayama Univ Material-immobilized carrier having material immobilized thereon, and method for preparing material-immobilized carrier

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007039858A2 (en) * 2005-10-04 2007-04-12 Koninklijke Philips Electronics N.V. Targeted imaging and/or therapy using the [3+2] azide-alkyne cycloaddition

Non-Patent Citations (65)

* Cited by examiner, † Cited by third party
Title
AGARD ET AL., ACS CHEM. BIOL., vol. 1, 2006, pages 644
AGARD ET AL., ACS CHEM. BIOL., vol. 1, 2006, pages 644 - 648
AGARD ET AL., J. AM. CHEM. SOC., vol. 126, 2004, pages 15046 - 15047
AGARD N J ET AL: "A strain-promoted [3 + 2] azide-alkyne cycloaddition for covalent modification of biomolecules in living systems", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, DC., US, vol. 126, no. 46, 24 November 2004 (2004-11-24), pages 15046 - 15047, XP002362785, ISSN: 0002-7863 *
BANTSCHEFF ET AL., ANAL. BIOANAL. CHEM., vol. 389, 2007, pages 1017
BASKIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 16793 - 16797
BASKIN ET AL., QSAR COMB. SCI., vol. 26, 2007, pages 1211 - 1219
BINDER ET AL., MACROMOL. RAPID COMMUN., vol. 2G, 2008, pages 952 - 981
CHIN ET AL., SCIENCE, vol. 301, 2003, pages 964
CHIN ET AL., SCIENCE, vol. 301, 2003, pages 964 - 967
CHOI ET AL., J. DISPERSION SCI. TECH., vol. 24, 2003, pages 475 - 487
CODELLI ET AL., J AM. CHEM. SOC., vol. 130, 2008, pages 11486 - 11493
DEDOLA ET AL., ORG. BIOMOL. CHEM., vol. 5, 2007, pages 1006 - 1017
DEDOLA ET AL., OROG. BIOMOL. CHEM., vol. 5, 2007, pages 1006
DICKEN ET AL., J. ORG. CHEM., vol. 47, 1982, pages 2047 - 2051
GAUCHER ET AL., J. CONTROL. RELEASE, vol. 109, 2005, pages 169 - 188
GIERLICH ET AL., ORG. LETT., vol. 8, 2006, pages 3639
GIERLICH ET AL., ORG. LETT., vol. 8, 2006, pages 3639 - 3642
HANSON ET AL., J AM, CHEM SOC., vol. 129, 2007, pages 7266
HANSON ET AL., J. AM. CHEM. SOC., vol. 129, 2007, pages 7266
HANSON ET AL., J. AM. CHEM. SOC., vol. 129, 2007, pages 7266 - 7267
INOUYE ET AL., BULL. CHEM. SOC. JPN., vol. 56, 1983, pages 3541 - 3542
JOHNSON ET AL., CHEM. COMMUN., 2008, pages 3064 - 3066
JUNG ET AL., J. ORG. CHEM., vol. 43, 1978, pages 3698 - 3701
JUNG; MILLER, J. AM. CHEM. SOC., vol. 103, 1981, pages 1984 - 1992
KHO ET AL., PROC. NATL. ACAD SCI. USA, vol. 101, 2004, pages 12479 - 12484
KHO ET AL., PROC. NATL. ACAD. SCI., vol. 101, 2004, pages 12479
KOLB; SHARPLESS, DRUG DIS. TODAY, vol. 8, 2003, pages 1128
KOLB; SHARPLESS, DRUG DISCOVERY TODAY, vol. 8, 2003, pages 1128 - 1137
LAU ET AL., PROTEOMICS, vol. 7, 2007, pages 2787
LAUGHLIN ET AL., SCIENCE, vol. 320, 2008, pages 664 - 667
LINK A JAMES ET AL: "Cell surface labeling of Escherichia coli via copper(I)-catalyzed (3+2) cycloaddition", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, DC., US, vol. 125, no. 37, 17 September 2003 (2003-09-17), pages 11164 - 11165, XP002329889, ISSN: 0002-7863 *
LINK ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 10180 - 10185
LINK ET AL., PROC. NATL. ACAD. SCI., vol. 103, 2006, pages 10180
LINK; TIRREL, J. AM. CHEM. SOC., vol. 125, 2003, pages 11164 - 11165
LUCHANSKY; BERTOZZI, CHEM-BIOCHEM, vol. 5, 2004, pages 1706 - 1709
MOSES; MOORHOUSE, CHEM. SOC. REV., vol. 36, 2007, pages 1249
MOSES; MOORHOUSE, CHEM. SOC. REV., vol. 36, 2007, pages 1249 - 1262
N J AGARD ET AL.: "A Comparative Study of Bioorthogonal Reactions with Azides", ACS CHEMICAL BIOLOGY, vol. 1, no. 10, 10 November 2006 (2006-11-10), pages 644 - 648, XP002522847 *
NANDIVADA ET AL., ADV. MATER., vol. 19, 2007, pages 2197
NANDIVADA ET AL., ADV. MATER., vol. 19, 2007, pages 2197 - 2208
NISHIYAMA ET AL., ADV. POLYM. SCI., vol. 193, 2006, pages 67 - 101
PRESCHER JENNIFER A ET AL: "Chemistry in living systems", NATURE CHEMICAL BIOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 1, no. 1, 1 June 2005 (2005-06-01), pages 13 - 21, XP002410570, ISSN: 1552-4450 *
PRESCHER; BERTOZZI, NAT. CHEM. BIOL., vol. 1, 2005, pages 13
PRESCHER; BERTOZZI, NAT. CHEM. BIOL., vol. 1, 2005, pages 13 - 21
ROSTOVTSEV ET AL., ANGEW. CHEM. LNT. ED., vol. 41, 2002, pages 2596 - 2599
ROSTOVTSEV ET AL., ANGEW. CHEM., vol. 114, 2002, pages 2708 - 2711
SEITZ ET AL., ANGEW. CHEM. INT. ED ENGL., vol. 8, 1969, pages 447 - 448
SEITZ ET AL., ANGEW. CHEM., vol. 81, 1969, pages 427 - 428
SIVAKUMAR ET AL., ORG. LETT., vol. 6, 2004, pages 4603
SIVAKUMAR ET AL., ORG. LETT., vol. 6, 2004, pages 4603 - 4606
SLETTEN ET AL., ORGANIC LETTERS, vol. 10, 2008, pages 3097 - 3099
SPEERS ET AL., J. AM. CHEM. SOC., vol. 125, 2003, pages 4686
SPEERS ET AL., J. AM. CHEM. SOC., vol. 125, 2003, pages 4686 - 4687
SUN ET AL., BIOCONJUGATE CHEM., vol. 17, 2006, pages 52
SUN ET AL., BIOCONJUGATE CHEM., vol. 17, 2006, pages 52 - 57
TOO, EXPERT REV. PROTEOMICS, vol. 4, 2007, pages 603
TORNOE ET AL., J. ORG. CHEM., vol. 67, 2002, pages 3057 - 3064
TURNER ET AL., J. AM. CHEM. SOC., vol. 95, 1973, pages 790 - 792
VANBERKEL ET AL., CHEM-BIOCHEM, vol. 8, 2007, pages 1504 - 1508
WANG ET AL., J. AM. CHEM. SOC., vol. 125, 2003, pages 3192
WANG ET AL., J. AM. CHEM. SOC., vol. 125, 2003, pages 3192 - 3193
WU; FOKIN, ALDRICHIMICA ACTA, vol. 40, 2007, pages 7
WU; FOKIN, ALDRICHIMICA ACTA, vol. 40, 2007, pages 7 - 17
X NING ET AL.: "Visualizing Metabolically Labeled Glycoconjugates of Living Cells by Copper-Free and Fast Huisgen Cycloaddition", ANGEW CHEM INT ED, vol. 47, 14 February 2008 (2008-02-14), pages 2253 - 2255, XP002522848 *

Cited By (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8940859B2 (en) 2007-11-21 2015-01-27 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US9227943B2 (en) 2007-11-21 2016-01-05 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US9725405B2 (en) 2007-11-21 2017-08-08 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US9932297B2 (en) 2007-11-21 2018-04-03 University Of Georgia Research Foundation, Inc. Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US10821196B2 (en) 2008-04-30 2020-11-03 Siemens Medical Solutions Usa, Inc. Substrate based PET imaging agents
US8962580B2 (en) 2008-09-23 2015-02-24 Alnylam Pharmaceuticals, Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
US8541625B2 (en) 2009-02-19 2013-09-24 University Of Georgia Research Foundation, Inc. Cyclopropenones and the photochemical generation of cyclic alkynes therefrom
US8426649B2 (en) 2009-02-19 2013-04-23 University Of Georgia Research Foundation, Inc. Cyclopropenones and the photochemical generation of cyclic alkynes therefrom
US8258347B2 (en) 2009-02-19 2012-09-04 University Of Georgia Research Foundation, Inc. Cyclopropenones and the photochemical generation of cyclic alkynes therefrom
CN102040530A (en) * 2009-10-15 2011-05-04 原子能与替代能源委员会 Process for the functionalization of biological molecules
US10550386B2 (en) * 2010-01-28 2020-02-04 Alnylam Pharmaceuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
US9422294B2 (en) 2010-03-26 2016-08-23 Jnc Corporation Cyclic compound, method for producing cyclic compound, and method for modifying biological molecule
US8901312B2 (en) 2010-03-26 2014-12-02 Jnc Corporation Cyclic compound, method for producing cyclic compound, and method for modifying biological molecule
WO2011118394A1 (en) * 2010-03-26 2011-09-29 Jnc株式会社 Cyclic compound, method for producing cyclic compound, and method for modifying biological molecule
JP5741572B2 (en) * 2010-03-26 2015-07-01 Jnc株式会社 Cyclic compound, method for producing cyclic compound, and method for modifying biomolecule
US11358921B2 (en) 2010-04-27 2022-06-14 Synaffix B.V. Fused cyclooctyne compounds and their use in metal-free click reactions
EP3604264A1 (en) 2010-04-27 2020-02-05 SynAffix B.V. Fused cyclooctyne compounds
US10239807B2 (en) 2010-04-27 2019-03-26 Synaffix B.V. Fused cyclooctyne compounds and their use in metal-free click reactions
WO2011136645A1 (en) 2010-04-27 2011-11-03 Stichting Katholieke Universiteit, More Particularly Radboud University Nijmegen Fused cyclooctyne compounds and their use in metal-free click reactions
US8859629B2 (en) 2010-04-27 2014-10-14 Synaffix B.V. Fused cyclooctyne compounds and their use in metal-free click reactions
US9222940B2 (en) 2010-04-27 2015-12-29 Synaffix B.V. Fused cyclooctyne compounds and their use in metal-free click reactions
EP2894142A1 (en) 2010-04-27 2015-07-15 SynAffix B.V. Fused cyclooctyne compounds and their use in metal-free click reactions
US9855287B2 (en) 2010-07-28 2018-01-02 Life Technologies Corporation Anti-viral azide containing compounds
WO2012016044A1 (en) 2010-07-28 2012-02-02 Life Technologies Corporation Anti-viral azide containing compounds
WO2012016048A1 (en) 2010-07-28 2012-02-02 Life Technologies Corporation Anti- viral azide-containing compounds
US10179143B2 (en) 2010-07-28 2019-01-15 Life Technologies Corporation Anti-viral azide containing compounds
US10632133B2 (en) 2010-07-28 2020-04-28 The Johns Hopkins University Anti-viral azide containing compounds
US9144575B2 (en) 2010-07-28 2015-09-29 Life Technologies Corporation Anti-viral azide containing compounds
US8912322B2 (en) 2010-07-29 2014-12-16 University Of Georgia Research Foundation, Inc. Aza-dibenzocyclooctynes and methods of making and using same
USRE47539E1 (en) 2010-07-29 2019-07-30 University Of Georgia Research Foundation, Inc. Aza-dibenzocyclooctynes and methods of making and using same
US20130202652A1 (en) * 2010-07-30 2013-08-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
US9315468B2 (en) 2010-09-27 2016-04-19 University Of Georgia Research Foundation, Inc. Methods including latent 1,3-dipole-functional compounds and materials prepared thereby
WO2012047663A3 (en) * 2010-09-27 2012-05-31 University Of Georgia Reaserch Foundation, Inc. Methods including latent 1,3-dipole-functional compounds and materials prepared thereby
CN103347862B (en) * 2010-09-27 2018-07-20 乔治亚大学研究基金公司 Including the method for potential 1,3- dipoles-functional compound and the material thus prepared
CN103347862A (en) * 2010-09-27 2013-10-09 乔治亚大学研究基金公司 Method including latent 1,3-dipole-functional compound and material prepared thereby
US20120208722A1 (en) * 2010-10-19 2012-08-16 Richard Dluhy Surface enhanced raman spectroscopy platforms and methods
EP2966061A1 (en) 2011-03-04 2016-01-13 Life Technologies Corporation Compounds and methods for conjugation of biomolecules
WO2012121973A1 (en) 2011-03-04 2012-09-13 Life Technologies Corporation Compounds and methods for conjugation of biomolecules
WO2012134925A1 (en) 2011-03-25 2012-10-04 Life Technologies Corporation Heterobifunctional esters for use in labeling target molecules
US9440925B2 (en) 2011-03-25 2016-09-13 Life Technologies Corporation SDP-containing heterobifunctional agents
US9145361B2 (en) 2011-03-25 2015-09-29 Life Technologies Corporation SDP-containing heterobifunctional agents
WO2012142003A2 (en) 2011-04-15 2012-10-18 Life Technologies Corporation Chemical ligation
EP2532639A1 (en) * 2011-06-09 2012-12-12 ModiQuest B.V. Method for preparing a reactive coating
US10675359B2 (en) 2011-12-23 2020-06-09 Innate Pharma Enzymatic conjugation of antibodies
US9764038B2 (en) 2011-12-23 2017-09-19 Innate Pharma Enzymatic conjugation of antibodies
US9717803B2 (en) 2011-12-23 2017-08-01 Innate Pharma Enzymatic conjugation of polypeptides
US10132799B2 (en) 2012-07-13 2018-11-20 Innate Pharma Screening of conjugated antibodies
WO2014009426A3 (en) * 2012-07-13 2014-04-03 Innate Pharma Screening of conjugated antibodies
EP2911699B1 (en) 2012-10-23 2017-11-15 SynAffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof
EP3912642A1 (en) 2012-10-23 2021-11-24 Synaffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof
US10745488B2 (en) 2012-10-23 2020-08-18 Synaffix B.V. Modified antibody, antibody-conjugate and process for the preparation thereof
WO2014066733A2 (en) 2012-10-25 2014-05-01 Life Technologies Corporation Methods and compositions for enzyme-mediated site-specific radiolabeling of glycoproteins
US10036010B2 (en) 2012-11-09 2018-07-31 Innate Pharma Recognition tags for TGase-mediated conjugation
WO2014111344A1 (en) * 2013-01-15 2014-07-24 Novartis Ag Cycloalkyne derivatized saccharides
US11135300B2 (en) 2013-01-15 2021-10-05 Glaxosmithkline Biologicals Sa Cycloalkyne derivatized saccharides
US10611824B2 (en) 2013-03-15 2020-04-07 Innate Pharma Solid phase TGase-mediated conjugation of antibodies
US10973922B2 (en) 2013-05-02 2021-04-13 Glykos Finland Oy Glycoprotein-toxic payload conjugates
WO2014189370A1 (en) 2013-05-24 2014-11-27 Stichting Katholieke Universiteit Substituted azadibenzocyclooctyne compounds and their use in metal-free click reactions
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
US9427478B2 (en) 2013-06-21 2016-08-30 Innate Pharma Enzymatic conjugation of polypeptides
US10434180B2 (en) 2013-06-21 2019-10-08 Innate Pharma Enzymatic conjugation of polypeptides
US9987373B2 (en) 2013-10-14 2018-06-05 Synaffix B.V. Modified glycoprotein, protein-conjugate and process for the preparation thereof
WO2015057066A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Glycoengineered antibody, antibody-conjugate and methods for their preparation
WO2015057064A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Modified glycoprotein, protein-conjugate and process for the preparation thereof
EP3929301A1 (en) 2013-10-14 2021-12-29 SynAffix B.V. Glycoengineered antibody, antibody-conjugate and methods for their preparation
WO2015057065A1 (en) 2013-10-14 2015-04-23 Synaffix B.V. Glycoengineered antibody, antibody-conjugate and methods for their preparation
US11168085B2 (en) 2014-01-24 2021-11-09 Synaffix B.V. Process for the cycloaddition of a hetero(aryl) 1,3-dipole compound with a (hetero)cycloalkyne
US11872215B2 (en) 2014-04-08 2024-01-16 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glyconjugates and methods
US10905678B2 (en) 2014-04-08 2021-02-02 University Of Georgia Research Foundation, Inc. Site-specific antibody-drug glycoconjugates and methods
US10047361B2 (en) 2015-03-17 2018-08-14 Arrowhead Pharmaceuticals, Inc. Disulfide-containing alkyne linking agents
US9803201B2 (en) 2015-03-17 2017-10-31 Arrowhead Pharmaceuticals, Inc. Disulfide-containing alkyne linking agents
WO2017137458A1 (en) 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
WO2017137457A1 (en) 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
US11312942B2 (en) 2016-06-08 2022-04-26 Sony Corporation Material-fixing substrate and method for producing same, and material-fixing agent used for material-fixing substrate
WO2017218891A1 (en) 2016-06-17 2017-12-21 Life Technologies Corporation Site-specific crosslinking of antibodies
US11591408B2 (en) 2017-11-09 2023-02-28 National Research Council Of Canada Antibody glycoconjugates and methods of production and use
WO2019169307A1 (en) 2018-03-02 2019-09-06 Life Technologies Corporation Novel quencher and reporter dye combinations
WO2019240219A1 (en) 2018-06-14 2019-12-19 持田製薬株式会社 Novel crosslinked alginic acid
WO2019243672A1 (en) 2018-06-19 2019-12-26 Glykos Biomedical Oy Conjugate
WO2020002765A1 (en) 2018-06-29 2020-01-02 Glykos Biomedical Oy Conjugates
WO2020262642A1 (en) 2019-06-28 2020-12-30 持田製薬株式会社 Transplantation device using chemically crosslinked alginic acid
WO2021116037A1 (en) 2019-12-09 2021-06-17 F. Hoffmann-La Roche Ag Dicationic fluorescent dyes
WO2021123506A1 (en) 2019-12-18 2021-06-24 Glykos Biomedical Oy Stabile conjugate
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle

Also Published As

Publication number Publication date
CN104529711A (en) 2015-04-22
EP2907525A3 (en) 2015-12-02
JP5956487B2 (en) 2016-07-27
EP2222341B1 (en) 2015-02-25
DK2907525T3 (en) 2018-08-06
CN101925366A (en) 2010-12-22
US20100297250A1 (en) 2010-11-25
DK2222341T3 (en) 2015-03-09
CN104529711B (en) 2020-02-07
US8940859B2 (en) 2015-01-27
JP5498952B2 (en) 2014-05-21
US20120172575A1 (en) 2012-07-05
EP2907525B1 (en) 2018-05-16
CN101925366B (en) 2015-02-04
WO2009067663A8 (en) 2009-07-23
US20120322974A9 (en) 2012-12-20
US20160159732A1 (en) 2016-06-09
JP2011504507A (en) 2011-02-10
US8133515B2 (en) 2012-03-13
US9725405B2 (en) 2017-08-08
US9932297B2 (en) 2018-04-03
US20170320815A1 (en) 2017-11-09
US20150126706A1 (en) 2015-05-07
JP2018039841A (en) 2018-03-15
US20120040011A9 (en) 2012-02-16
US9227943B2 (en) 2016-01-05
JP2014177458A (en) 2014-09-25
JP6659397B2 (en) 2020-03-04
JP2016145221A (en) 2016-08-12
EP2222341A1 (en) 2010-09-01
EP2907525A2 (en) 2015-08-19

Similar Documents

Publication Publication Date Title
US9932297B2 (en) Alkynes and methods of reacting alkynes with 1,3-dipole-functional compounds
US8497299B2 (en) Compositions including quinonoid derivatives of cannabinoids for therapeutic use
Genady et al. New functionalized mercaptoundecahydrododecaborate derivatives for potential application in boron neutron capture therapy: Synthesis, characterization and dynamic visualization in cells
US8541604B2 (en) Process for the functionalization of biological molecules
Zhong et al. Amphiphilic drug–drug assembly via dual-responsive linkages for small-molecule anticancer drug delivery
CN108822157A (en) The inclusion compound and preparation method thereof of platinum medicine and open loop Cucurbituril
US20220322945A1 (en) Use of fluorophore compounds of the aza- bodipy type as contrast agents in the short wave infrared region
Wu et al. Strained alkyne substituted near infrared BF 2 azadipyrromethene fluorochrome
WO2022099762A1 (en) Antibody conjugate intermediate and preparation method therefor
CN110396122B (en) Nuclear magnetic resonance contrast agent, preparation method and application thereof in tumor diagnosis
CN101861328A (en) Aminooxime derivatives of 2- and/or 4-substituted androstanes and androstenes as medicaments for cardiovascular disorders
CN111777643A (en) Conjugated oligomer-ruthenium complex, synthesis method thereof and application thereof in preparation of antitumor drugs
CN117924312A (en) Tumor targeting self-assembled molecule based on methylene blue, preparation method and pharmaceutical application thereof
CN117777751A (en) Cyanine dye and application thereof
US8471043B2 (en) Platensimycin derivatives, their intermediates, and process for preparing the same, and new process for preparing platensimycin
Simone I. FUNCTIONALIZATION OF CRYTPOHANE CAGES FOR XENON MRI II. VANADIUM CATALYZED OXIDATIVE COUPLING OF SP 3 C–H BONDS TO HETEROARENES
JPH10330346A (en) Linear chain nitron derivative, and medicine and reagent containing the same
BE878187A (en) DERIVATIVES OF ANTI-TUMOR STEROID HORMONES AND THEIR PREPARATION METHOD

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880125596.0

Country of ref document: CN

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

Ref document number: 08852196

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010535090

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 4153/DELNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008852196

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12743632

Country of ref document: US