US20130184229A1 - Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors - Google Patents

Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors Download PDF

Info

Publication number
US20130184229A1
US20130184229A1 US13/785,439 US201313785439A US2013184229A1 US 20130184229 A1 US20130184229 A1 US 20130184229A1 US 201313785439 A US201313785439 A US 201313785439A US 2013184229 A1 US2013184229 A1 US 2013184229A1
Authority
US
United States
Prior art keywords
alkanyl
compound
alkynyl
alkenyl
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/785,439
Inventor
John L. Magnani
Arun K. Sarkar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Glycomimetics Inc
Original Assignee
Glycomimetics 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 Glycomimetics Inc filed Critical Glycomimetics Inc
Priority to US13/785,439 priority Critical patent/US20130184229A1/en
Publication of US20130184229A1 publication Critical patent/US20130184229A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/26Acyclic or carbocyclic radicals, substituted by hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/207Cyclohexane rings not substituted by nitrogen atoms, e.g. kasugamycins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/22Cyclohexane rings, substituted by nitrogen atoms

Definitions

  • the present invention relates generally to compounds, compositions and methods for treating cancer and inflammatory diseases, and for enhancing retention of cells after releasing into circulating blood. More specifically, the present invention relates to heterobifunctional compounds that inhibit E-selectins and CXCR4 chemokine receptors, and uses thereof.
  • a number of cancers are highly treatable when treated before the cancer has moved beyond the primary site. However, often once the cancer has spread beyond the primary site, the treatment options are limited and the survival statistics decline dramatically. Bones are a common location for cancer to infiltrate once leaving the primary tumor location. Breast and prostate cancer are examples of cancers that migrate to bones. Even leukemic cells that arise in the bloodstream may home to the bone marrow. Once cancer resides in bone, it is frequently a cause of pain to the individual. Further, once in the bone marrow, the cancer cells may also become resistant to chemotherapy. In addition, if the particular bone affected is a source for production of blood cells in the bone marrow, the individual may develop a variety of blood cell related disorders. Thus, it is desirable to prevent cancer cells from leaving the primary site, or to prevent extravasation of cancer cells from the bloodstream and infiltration into other tissues. Retention of cancer cells in the bloodstream makes the cells more susceptible to treatment, such as chemotherapy.
  • Some cancers originate all or in part in bone. For such cancers, it is desirable to mobilize cancer cells from bone to the bloodstream and to prevent those cells (as well as any cancer cells already in the bloodstream) from homing to bone or otherwise leaving the bloodstream. Retention of cancer cells in the bloodstream (or mobilization of cancer cells into the bloodstream and then retention therein) makes the cells more susceptible to treatment, such as chemotherapy.
  • HSCs Hematopoietic stem cells
  • HSCs also reside in the bone marrow and are a source of material for cellular therapy. HSCs adhere to the stroma within the bone marrow and in order to be harvested must break these adhesions and mobilize out of the bone marrow. It is desirable to have improved agents to increase the HSCs available for harvesting. Such HSCs are useful for engraftment.
  • the compounds are heterobifunctional compounds wherein an E-selectin inhibitor is linked to a CXCR4 chemokine receptor inhibitor.
  • Such compounds may be combined with a pharmaceutically acceptable carrier or diluent to form a pharmaceutical composition.
  • the compounds may be used to treat cancer in which the cancer cells may leave the primary site, or to treat an inflammatory disease in which the adhesion or migration of cells occurs in the disease, or to release cells such as stem cells (e.g., bone marrow progenitor cells) into circulating blood and enhance retention of the cells in the blood (e.g., to mobilize cells out of bone marrow and maintain the cells in the peripheral bloodstream).
  • stem cells e.g., bone marrow progenitor cells
  • the present invention provides a heterobifunctional compound for inhibition of E-selectin and the CXCR4 chemokine receptor, comprising E-selectin inhibitor-Linker-CXCR4 chemokine receptor inhibitor, or a physiologically acceptable salt thereof.
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the CXCR4 chemokine receptor inhibitor consists of:
  • the compound has the formula:
  • the compound has the formula:
  • the compound has the formula:
  • the compound has the formula:
  • the compound has the formula:
  • the Linker of the compound is —C( ⁇ O)—NH—(CH 2 ) 2 —NH—.
  • the Linker of the compound is —CH 2 —NH—CH 2 —.
  • the Linker of the compound is —C( ⁇ O)—NH—CH 2 —.
  • linkers as well as the others disclosed herein and those otherwise known in the art, are for use in a compound of the present invention such as the four embodiments depicted above containing a Linker.
  • the present invention provides a method for the treatment of a cancer in which the cancer cells may leave the primary site in an individual who is in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • the present invention provides a method for the treatment of a cancer in which it is desired to mobilize cancer cells from a site into the bloodstream and retain the cancer cells in the bloodstream in an individual who is in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • the present invention provides a method for releasing cells into circulating blood and enhancing retention of the cells in the blood of an individual who is need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • the method further includes the step of collecting the cells released.
  • the step of collecting utilizes apheresis.
  • the cells are stem cells (e.g., bone marrow progenitor cells).
  • G-CSF is administered to the individual.
  • the present invention provides a method for the treatment of an inflammatory disease in which the adhesion or migration of cells occurs in the disease in an individual in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically acceptable carrier or diluent.
  • the above compounds thereof may be used in the manufacture of a medicament, and for any of the uses recited herein.
  • FIG. 1 is a diagram illustrating the synthesis of heterobifunctional Compound #1 (compound 27).
  • FIG. 2 ( FIG. 2A , FIG. 2B and FIG. 2C ) is a diagram illustrating the synthesis of heterobifunctional Compound #2 (compound 28).
  • FIG. 3 depicts the inhibition of binding of anti-CXCR4-PE to SupT 1 cells in a dose-dependent manner by heterobifunctional Compound #1.
  • FIG. 4 depicts the results of an E-selectin assay in which heterobifunctional Compound #1 is used as the inhibitor.
  • FIG. 5 depicts a comparison of IC 50 values of compounds A, B and #1 for E-selectin.
  • Compound A which is a known E-selectin inhibitor, is compound 15 of Thoma et al. ( J. Med. Chem. 42:4909-4913, 1999) and is used as a reference compound.
  • Compound #1 is heterobifunctional Compound #1.
  • Compound B is the glycomimetic portion of Compound #1 (i.e., compound 18 of FIG. 1 except modified to replace COOMe, which is used in the linking process, with H).
  • the present invention provides compounds, compositions and methods for treating diseases in which an E-selectin and a CXCR4 chemokine receptor play a role, and for enhancing retention of cells after releasing into circulating blood.
  • the compounds have a variety of uses in vitro and in vivo.
  • E-selectin inhibitor refers to an inhibitor of E-selectin only, as well as to an inhibitor of E-selectin and either P-selectin or L-selectin, or E-selectin and both P-selectin and L-selectin.
  • E-selectin inhibition regardless of whether there is also inhibition of either P-selectin or L-selectin or both P-selectin and L-selectin.
  • All compounds of the present invention or useful thereto include physiologically acceptable salts thereof.
  • physiologically acceptable salts thereof are Na, K, Li, Mg, Ca, and Cl.
  • a compound of the present invention is a heterobifunctional compound wherein an E-selectin inhibitor is linked (i.e., covalently bonded) to a CXCR4 chemokine receptor inhibitor.
  • Such a compound comprises, or consists of, the formula:
  • E-selectin inhibitor-Linker-CXCR4 chemokine receptor inhibitor Accordingly, the compound functions to inhibit E-selectin and to inhibit the CXCR4 chemokine receptor.
  • E-selectin inhibitors are well known in the art. Some E-selectin inhibitors are specific for E-selectin only. Other E-selectin inhibitors have the ability to inhibit not only E-selectin but additionally P-selectin or L-selectin or both P-selectin and L-selectin. Examples of E-selectin inhibitors (specific for E-selectin or otherwise) are disclosed in U.S. Pat. No. 7,060,685; U.S. Application Publication No. US-2007-0054870; U.S. Application Publication No. US-2008-0161546; and references cited in any of these patent or published application documents. Those examples are small organic molecules. Other known E-selectin inhibitors are amino acid-based, such as antibodies. For example, the humanized monoclonal antibody CDP850 is an E-selectin inhibitor.
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • the E-selectin inhibitor consists of:
  • CXCR4 chemokine receptor inhibitors are well known in the art. Such inhibitors will typically prevent the binding of stromal derived factor-1 (SDF-1) to a CXCR4 receptor.
  • SDF-1 stromal derived factor-1
  • Examples of CXCR4 chemokine receptor inhibitors are AMD-3100 (Hendrix et al., Antimicrob. Agents Chemother. 44:1667-1673, 2000); ALX40-4C (Doranz et al., AIDS Research and Human Retroviruses 17:475-486, 2001); and T134 (Arakaki et al., J. Virol. 73:1719-1723, 1999). These examples include a small organic molecule and amino acid-based molecules, such as the T22 peptide. AMD-3100 is a bicyclam.
  • each of the two cyclam rings is attached to the same phenyl ring (each cyclam ring is para to the other) via a methylene group.
  • the CXCR4 chemokine receptor inhibitor is a phenyl ring to which is attached only one cyclam ring.
  • an E-selectin inhibitor and a CXCR4 chemokine receptor inhibitor are covalently joined via a linker (i.e., interposed between the two inhibitors is a “Linker”).
  • a linker may be (or may include) a spacer group, such as —(CH 2 ) p — or —O(CH 2 ) p — where p is generally about 1-20 (including any whole integer range therein).
  • spacer groups include a carbonyl or carbonyl containing group such as an amide. An embodiment of such spacer groups is
  • linkers e.g., polyethylene glycols (PEG) or —C( ⁇ O)—NH—(CH 2 ) p —C( ⁇ O)—NH 2 where p is as defined above, will be familiar to those in the art or in possession of the present disclosure.
  • PEG polyethylene glycols
  • p is as defined above
  • the linker is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl linker
  • the linker is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl linker
  • the linker is —C( ⁇ O)—NH—(CH 2 ) 2 —NH—.
  • the linker is —CH 2 —NH—CH 2 —.
  • the linker is —C( ⁇ O)—NH—CH 2 —.
  • the E-selectin inhibitor consists of:
  • Me is methyl.
  • Et is ethyl.
  • Ar is aryl.
  • Bz is benzoyl.
  • Selection of a substituent at R 1 includes H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl, halogenated C 1 -C 8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X ⁇ H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl, halogenated C 1 -C 8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C( ⁇ O)OX, alkanyl substituted with C( ⁇ O)OX, C( ⁇ O)NHX, alkanyl substituted with C( ⁇ O)NHX, where X ⁇ H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 al
  • Selection of a substituent at R 2 includes —OH,
  • Selection of a substituent at R 3 includes H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl, CN, CH 2 CN, C( ⁇ O)X where X is H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl, NHOH, NHOCH 3 , NHCN, or NX 2 , or C( ⁇ O)OY where Y is H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl or C 1 -C 8 alkynyl; and
  • cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl or OY where Y is H, C 1 -C 8 alkanyl, C 1 -C 8 alkenyl, C 1 -C 8 alkynyl or C 1 -C 14 aryl.
  • a “C 1 -C 8 alkanyl” refers to an alkane substituent with one to eight carbon atoms and may be straight chain, branched or cyclic (cycloalkanyl). Examples are methyl, ethyl, propyl, isopropyl, butyl and t-butyl.
  • a “halogenated C 1 -C 8 alkanyl” refers to a “C 1 -C 8 alkanyl” possessing at least one halogen. Where there is more than one halogen present, the halogens present may be the same or different or both (if at least three present).
  • a “C 1 -C 8 alkenyl” refers to an alkene substituent with one to eight carbon atoms, at least one carbon-carbon double bond, and may be straight chain, branched or cyclic (cycloalkenyl). Examples are similar to “C 1 -C 8 alkanyl” examples except possessing at least one carbon-carbon double bond.
  • a “C 1 -C 8 alkynyl” refers to an alkyne substituent with one to eight carbon atoms, at least one carbon-carbon triple bond, and may be straight chain, branched or cyclic (cycloalkynyl). Examples are similar to “C 1 -C 8 alkanyl” examples except possessing at least one carbon-carbon triple bond.
  • alkoxy refers to an oxygen substituent possessing a “C 1 -C 8 alkanyl,” “C 1 -C 8 alkenyl” or “C 1 -C 8 alkynyl.” This is —O-alkyl; for example methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy and the like; and alkenyl or alkynyl variations thereof (except for methoxy). It further refers to the group O-alkyl-W-alkyl where W is O or N; for example —O—(CH 2 ) n —W—(CH 2 ) m where n and m are independently 1-10.
  • aryl refers to an aromatic substituent with one to fourteen carbon atoms as ring atoms in one or multiple rings which may be separated by a bond or fused.
  • aryl includes “heteroaryl.”
  • a “heteroaryl” is similar to an “aryl” except the aromatic substituent possesses at least one heteroatom (such as N, O or S) in place of a ring carbon.
  • heteroaryl is typically six to fourteen ring atoms.
  • aryls include phenyl, naphthyl, pyridinyl, pyrimidinyl, triazolo, furanyl, oxazolyl, thiophenyl, quinolinyl and diphenyl.
  • the CXCR4 chemokine receptor inhibitor consists of:
  • the compound has the formula:
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • the linker is —C( ⁇ O)—NH—(CH 2 ) 2 —NH—
  • the compound has the formula:
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • the linker is —CH 2 —NH—CH 2 —
  • the compound has the formula:
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • the linker is —C( ⁇ O)—NH—CH 2 —
  • the compound has the formula:
  • R 1 , R 2 , R 3 and R 4 are as defined above.
  • the linker is —C( ⁇ O)—NH—(CH 2 ) 2 —NH—
  • the compound has the formula:
  • the linker is —CH 2 —NH—CH 2 —
  • the compound has the formula:
  • the linker is —C( ⁇ O)—NH—CH 2 —
  • the compound has the formula:
  • the linker is —C( ⁇ O)—NH—(CH 2 ) 2 —NH—
  • the compound has the formula:
  • All compounds of the present invention or useful thereto include physiologically acceptable salts thereof.
  • physiologically acceptable salts thereof are Na, K, Li, Mg, Ca and Cl.
  • a pharmaceutical composition comprises one or more compounds in combination with (i.e., not covalently bonded to) one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • Such compositions may comprise buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione, adjuvants (e.g., aluminum hydroxide) or preservatives.
  • buffers e.g., neutral buffered saline or phosphate buffered saline
  • carbohydrates e.g., glucose, mannose, sucrose or dextrans
  • mannitol e.g., proteins, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or
  • compositions of the present invention may be formulated as a lyophilizate.
  • compositions of the present invention may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, intravenous, intracranial, intraperitoneal, subcutaneous, or intramuscular administration.
  • compositions described herein may be administered as part of a sustained release formulation (i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration).
  • a sustained release formulation i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration.
  • Such formulations may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of compound release.
  • the amount of compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • the compounds may be used in a method for the treatment of a cancer in which the cancer cells may leave the primary site.
  • a primary site may be, for example, solid tissue (e.g., breast or prostate) or the bloodstream.
  • An individual who is in need of such treatment is administered at least one (i.e., one or more) of the above-described compounds in an amount effective for the treatment.
  • infiltrating diseases include lung cancer and melanoma, as well as the hematological malignancies (e.g., leukemias and myelomas).
  • the term “treatment” includes for the disease or a complication associated with the disease, and includes prevention.
  • a complication associated with the cancer may not have presented itself in an individual with the disease, and a compound may be administered to prevent presentation of the complication in the individual.
  • Complications associated with a cancer in which the cancer cells may leave the primary site include, for example, metastasis and infiltration of cancer cells to other tissues.
  • AML acute myelogenous leukemia
  • MM multiple myeloma
  • Administration of a compound described herein may prevent adhesion or migration of cancer cells. Such prevention can result in making the cancer cells more susceptible to treatment with chemotherapy.
  • Administration of a compound described herein in the context of prevention may be to an individual who is at risk of occurrence of a cancer for the first time, or for recurrence of a cancer. For example, while a brain cancer such as glioblastoma multiforme is typically treated with another type of therapy (such as radiation or chemotherapy) for the first occurrence, such therapy is usually not effective to prevent recurrence.
  • treatment refers to any of a variety of positive effects from the treatment including, for example, eradicating a complication associated with the disease, relieving to some extent a complication, slowing or stopping progression of the disease, enhancing the effectiveness of one or more therapies for the disease, and prolonging the survival time of the recipient.
  • the treatment may be used in conjunction with one or more other therapies for a cancer or a complication associated therewith.
  • the above-described compounds including equivalents may be used in a method for the treatment of a cancer in which it is desired to mobilize cancer cells from a site into the bloodstream and retain the cancer cells in the bloodstream.
  • An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment.
  • cancers for such treatment include leukemias and myelomas (e.g., AML and MM). Mobilizing cancer cells into the bloodstream from a site and retaining the cells therein can result in making the cancer cells more susceptible to treatment with chemotherapy.
  • An example of a site from which to mobilize cancer cells is bone. Cancer cells may, for example, be in circulation and then home to bone.
  • a compound described herein may be used, for example, to mobilize cancer cells from bone into the bloodstream and prevent cancer cells from homing to bone, thereby retaining the cancer cells in the bloodstream.
  • Administration of a compound described herein in the context of prevention may be to an individual who is at risk of occurrence of a cancer for the first time, or for recurrence of a cancer.
  • a brain cancer such as glioblastoma multiforme is typically treated with another type of therapy (such as radiation or chemotherapy) for the first occurrence, such therapy is usually not effective to prevent recurrence.
  • the above-described compounds including equivalents may be used in a method for releasing cells (such as hematopoietic stem cells) into circulating blood and enhancing retention of the cells in the blood.
  • An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment.
  • One use of the method is, for example, for stem cell harvesting.
  • Stem cells may be needed, for example, after high-dose chemotherapy treatment.
  • Many chemotherapies suppress bone marrow which disrupts the production of certain components of blood in an individual. As a result, the individual may develop a variety of blood cell related disorders and continuation of chemotherapy may be compromised.
  • a compound described herein may be used, for example, to release stem cells into circulating blood and enhance retention of the stem cells in the blood.
  • the method may include a further step of collecting cells that are released.
  • released stem cells may be collected.
  • a variety of techniques are known in the art for collecting cells.
  • apheresis may be utilized.
  • An example of a stem cell is a bone marrow progenitor cell.
  • the release of such cells from bone marrow into circulating blood and retention therein has a variety of uses.
  • the mobilized bone marrow progenitor cells may be collected from the blood.
  • a use of such collected cells is to obtain healthy bone marrow progenitor cells from an individual prior to treatment of the individual in a manner such that bone marrow is suppressed.
  • the individual can receive a bone marrow transplantation utilizing the bone marrow progenitor cells collected prior to treatment. This is useful, for example, where an individual needs to be subjected to a chemotherapy protocol that will suppress bone marrow.
  • G-CSF granulocyte-colony stimulating factor
  • a compound described herein aids in releasing stem cells into circulating blood.
  • Stem cells produced in bone marrow and released into circulating blood, as a result of the combination of the administration (separately or together) of a compound described herein and G-CSF, may be collected as described above. Such collected stem cells may be, for example, administered to the individual after chemotherapy. The stem cells return to the bone marrow and produce blood cells.
  • Application of a compound described herein to mobilization and harvesting of healthy bone marrow progenitor cells from bone marrow treated with G-CSF provides cells useful, for example, for bone marrow transplantation.
  • the above-described compounds including equivalents may be used in a method for the treatment of an inflammatory disease in which the adhesion or migration of cells occurs in the disease.
  • An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment.
  • inflammatory diseases include inflammatory skin disorders such as atopic dermatitis and psoriasis.
  • the treatment may reduce (partially or totally) the disease or a complication associated therewith, such as pain.
  • the treatment may be used in conjunction with one or more other therapies for such an inflammatory disease or a complication associated therewith.
  • the above-described compounds may be administered in a manner appropriate to the disease to be treated.
  • Appropriate dosages and a suitable duration and frequency of administration may be determined by such factors as the condition of the patient, the type and severity of the patient's disease and the method of administration.
  • an appropriate dosage and treatment regimen provides the compound(s) in an amount sufficient to provide therapeutic or prophylactic benefit.
  • a compound may be administered at a dosage ranging from 0.001 to 1000 mg/kg body weight (more typically 0.01 to 1000 mg/kg), on a regimen of single or multiple daily doses.
  • Appropriate dosages may generally be determined using experimental models or clinical trials. In general, the use of the minimum dosage that is sufficient to provide effective therapy is preferred.
  • Patients may generally be monitored for therapeutic effectiveness using assays suitable for the condition being treated, which will be familiar to those of ordinary skill in the art.
  • At least one (i.e., one or more) of the above-described compounds may be administered in combination with at least one (i.e., one or more) agent, e.g., chemotherapeutic agent or anti-inflammatory agent.
  • the administration may be in conjunction with one or more other therapies for reducing toxicities of chemotherapy.
  • at least one (i.e., one or more) agent to counteract (at least in part) a side effect of chemotherapy may be administered.
  • At least one compound described herein may be administered before, after or simultaneous with administration of at least one chemotherapeutic agent or anti-inflammatory agent. Where administration is simultaneous, the combination may be administered from a single container or two (or more) separate containers.
  • reaction mixture is cooled down to RT and transferred into a separatory funnel with EtOAc (100 ml) and washed with H 2 O (100 ml).
  • the aqueous layer is extracted with EtOAc (2 ⁇ 200 ml).
  • the combined organic layers are dried (Na 2 SO 4 ), filtered and concentrated to dryness.
  • the crude product is purified by column chromatography (silica) to afford compound 4 as yellowish oil (1.1 g).
  • a mixture of compound 14 (0.16 g) and compound 15 (0.35 g, synthesized as described by Banteli et al., Helvetica Chimica Acta 83:2893-2907, 2000) is co-evaporated with toluene twice and then dried under vacuum.
  • the mixture is dissolved in dry CH 2 Cl 2 (10 ml) and stirred with flame dried molecular sieve (4 A) and 2,6-di-tert-Bu-pyridine (0.59 ml) for 30 min at RT.
  • the reaction mixture is cooled to 0° C. and MeOTf (0.25 ml) is added with stirring.
  • reaction mixture is stirred for 4 h at RT, filtered through a bed of Celite, washed with CH 2 Cl 2 (2 ⁇ 10 ml) and then transferred to a reparatory funnel.
  • the organic layer is washed with a cold saturated solution of NaHCO 3 (25 ml) and brine (25 ml), dried (Na 2 SO 4 ), filtered, and concentrated to dryness.
  • the residue is purified by column chromatography (silica) to give compound 16 (0.23 g).
  • Methyl shikimate (II, 10 g), 2,2 dimethoxypropane (10 ml) and p-TsOH (0.8 g) are dissolved in acetonitrile (125 ml) and stirred at rt for 1 h. The reaction mixture is then neutralized with triethylamine (2 ml) and evaporated to dryness. The residue is chromatographed on silica gel to yield III (11 g).
  • EDA-XIX (82 mg) is heated at 70° C. with ethylenediamine (EDA) (1 ml) with stirring for 5 h. Solvent is evaporated off and the purified by sephadex G-25 column to give EDA-XIX (82 mg).
  • Example 2 Compound 26 of Example 1 is reacted with EDA-XIX (and the product purified) using the procedures described in Example 1 (for the synthesis of compound 27) to give compound 28 which is heterobifunctional Compound #2 (also referred to herein as “Compound #2”).
  • the assay assesses the ability of glycomimetic compounds to inhibit binding of an anti-CXCR4 antibody conjugated to phycoerythrin (“PE”), to CXCR4 on the surface of SupT1 cells.
  • SupT1 cells are a T lymphoblast derived from a lymphoblastic leukemia and constitutively express CXCR4 on the cell surface.
  • the cells are purchased from ATCC (ATCC number CRL-1942).
  • Anti-human CXCR4-phycoerythrin monoclonal antibody (anti-CXCR4-PE) is purchased from R&D Systems (catalog number FAB 170P, clone 12G5).
  • the cells are grown in RPMI 1640 medium supplemented with 10% FBS.
  • each tube receives 10 ⁇ l of anti-CXCR4-PE, except one tube of cells receives 10 ⁇ l of mouse IgG 2a isotype control antibody.
  • the tubes are incubated at 4° C. for 45 minutes.
  • the cells are washed twice with PBS plus 0.05% BSA and the final cell pellet is resuspended in 100 ⁇ l of PBS/BSA.
  • 100 ⁇ l of 2% formaldehyde Polysciences, Inc. ultrapure EM grade, catalog number 040128 are added to each tube.
  • Flow cytometry is performed using a Cytomation MoFlo instrument.
  • Compound #1 inhibits binding of anti-CXCR4-PE to SupT1 cells in a dose-dependent manner with an IC 50 of 8.25 ⁇ M ( FIG. 3 ).
  • SDF-1 ⁇ efficiently inhibits binding of the antibody to CXCR4.
  • the inhibition assay to screen glycomimetic antagonists of E-selectin is a competitive binding assay, which allows the determination of IC 50 values. Briefly, E-selectin/Ig chimera is immobilized by incubation at 37° C. in 96 well microtiter plates for 2 hours. To reduce nonspecific binding, bovine serum albumin is added to each well and incubated at room temperature for 2 hours. The plate is washed and serial dilutions of the test compounds are added to the wells in the presence of conjugates of biotinylated, sLe a polyacrylamide with streptavidin/horseradishperoxidase and incubated for 2 hours at room temperature.
  • the peroxidase substrate 3,3′,5,5′ tetramethylbenzidin (TMB) is added. After 3 minutes, the enzyme reaction is stopped by the addition of H 3 PO 4 and the absorbance of light at a wavelength of 450 nm is determined. The concentration of test compound required to inhibit binding by 50% is determined and reported as the IC 50 value for each glycomimetic E-selectin antagonist. In addition to reporting the absolute IC 50 value as measured above, relative IC 50 values are determined by a ratio of the IC 50 measured for the test compound to that of an internal control (reference) stated for each assay.
  • Compound #1 is a potent E-selectin antagonist (as well as possessing anti-CXCR4 activity— FIG. 3 ).

Abstract

Compounds, compositions and methods are provided for treating cancer and inflammatory diseases, and for releasing cells such as stem cells (e.g., bone marrow progenitor cells) into circulating blood and enhancing retention of the cells in the blood. More specifically, heterobifunctional compounds that inhibit both E-selectins and CXCR4 chemokine receptors are described.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 12/768,173 filed Apr. 27, 2010, now allowed; which application claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Patent Application No. 61/174,580 filed May 1, 2009; which applications are incorporated herein by reference in their entirety.
  • BACKGROUND
  • 1. Technical Field
  • The present invention relates generally to compounds, compositions and methods for treating cancer and inflammatory diseases, and for enhancing retention of cells after releasing into circulating blood. More specifically, the present invention relates to heterobifunctional compounds that inhibit E-selectins and CXCR4 chemokine receptors, and uses thereof.
  • 2. Description of the Related Art
  • A number of cancers are highly treatable when treated before the cancer has moved beyond the primary site. However, often once the cancer has spread beyond the primary site, the treatment options are limited and the survival statistics decline dramatically. Bones are a common location for cancer to infiltrate once leaving the primary tumor location. Breast and prostate cancer are examples of cancers that migrate to bones. Even leukemic cells that arise in the bloodstream may home to the bone marrow. Once cancer resides in bone, it is frequently a cause of pain to the individual. Further, once in the bone marrow, the cancer cells may also become resistant to chemotherapy. In addition, if the particular bone affected is a source for production of blood cells in the bone marrow, the individual may develop a variety of blood cell related disorders. Thus, it is desirable to prevent cancer cells from leaving the primary site, or to prevent extravasation of cancer cells from the bloodstream and infiltration into other tissues. Retention of cancer cells in the bloodstream makes the cells more susceptible to treatment, such as chemotherapy.
  • Some cancers originate all or in part in bone. For such cancers, it is desirable to mobilize cancer cells from bone to the bloodstream and to prevent those cells (as well as any cancer cells already in the bloodstream) from homing to bone or otherwise leaving the bloodstream. Retention of cancer cells in the bloodstream (or mobilization of cancer cells into the bloodstream and then retention therein) makes the cells more susceptible to treatment, such as chemotherapy.
  • Hematopoietic stem cells (HSCs) also reside in the bone marrow and are a source of material for cellular therapy. HSCs adhere to the stroma within the bone marrow and in order to be harvested must break these adhesions and mobilize out of the bone marrow. It is desirable to have improved agents to increase the HSCs available for harvesting. Such HSCs are useful for engraftment.
  • Accordingly, there is a need in the art for the treatment of cancers that may leave the primary site and cancers that originate all or in part in bone, and for improved methods to aid in the preparation of therapeutic-grade stem cells. The present invention fulfills these needs and further provides other related advantages.
  • BRIEF SUMMARY
  • Briefly stated, compounds, compositions and methods for treating diseases and for improving methods in which an E-selectin and a CXCR4 chemokine receptor play a role, are provided. In the present invention, the compounds are heterobifunctional compounds wherein an E-selectin inhibitor is linked to a CXCR4 chemokine receptor inhibitor. Such compounds may be combined with a pharmaceutically acceptable carrier or diluent to form a pharmaceutical composition. The compounds may be used to treat cancer in which the cancer cells may leave the primary site, or to treat an inflammatory disease in which the adhesion or migration of cells occurs in the disease, or to release cells such as stem cells (e.g., bone marrow progenitor cells) into circulating blood and enhance retention of the cells in the blood (e.g., to mobilize cells out of bone marrow and maintain the cells in the peripheral bloodstream).
  • The present invention provides a heterobifunctional compound for inhibition of E-selectin and the CXCR4 chemokine receptor, comprising E-selectin inhibitor-Linker-CXCR4 chemokine receptor inhibitor, or a physiologically acceptable salt thereof.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00001
  • wherein:
      • L=end of bond to Linker;
      • R1=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)OX, alkanyl substituted with C(═O)OX, C(═O)NHX, alkanyl substituted with C(═O)NHX, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)X, OX, NHX, NHC(═O)X, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH;
      • R2=—OH,
  • Figure US20130184229A1-20130718-C00002
      •  —O—C(═O)—X, —NH2, —NH—C(═O)—NHX, or —NH—C(═O)—X where n=0-2 and X is independently selected from C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl,
  • Figure US20130184229A1-20130718-C00003
      •  where Q is H or a physiologically acceptable salt, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, aryl, (CH2)m-aryl where m is 1-10, and where n=0-10, and any of the above ring compounds may be substituted with one to three independently selected of Cl, F, CF3, C1-C8 alkoxy, NO2, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, C1-C14 aryl, or OY, C(═O)OY, NY2 or C(═O)NHY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, or C1-C14 aryl;
      • R3=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, CN, CH2CN, C(═O)X where X is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, NHOH, NHOCH3, NHCN, or NX2, or C(═O)OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl or C1-C8 alkynyl; and
  • Figure US20130184229A1-20130718-C00004
      •  where the cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or C1-C14 aryl.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00005
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00006
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00007
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00008
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the CXCR4 chemokine receptor inhibitor consists of:
  • Figure US20130184229A1-20130718-C00009
  • wherein L=end of bond to Linker.
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00010
  • wherein:
      • R1=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)OX, alkanyl substituted with C(═O)OX, C(═O)NHX, alkanyl substituted with C(═O)NHX, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)X, OX, NHX, NHC(═O)X, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH;
      • R2=—OH,
  • Figure US20130184229A1-20130718-C00011
      •  —O—C(═O)—X, —NH2, —NH—C(═O)—NHX, or —NH—C(═O)—X where n=0-2 and X is independently selected from C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl,
  • Figure US20130184229A1-20130718-C00012
      •  where Q is H or a physiologically acceptable salt, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, aryl, (CH2)m-aryl where m is 1-10, and where n=0-10, and any of the above ring compounds may be substituted with one to three independently selected of Cl, F, CF3, C1-C8 alkoxy, NO2, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, C1-C14 aryl, or OY, C(═O)OY, NY2 or C(═O)NHY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, or C1-C14 aryl;
      • R3=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, CN, CH2CN, C(═O)X where X is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, NHOH, NHOCH3, NHCN, or NX2, or C(═O)OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl or C1-C8 alkynyl; and
  • Figure US20130184229A1-20130718-C00013
      •  where the cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or C1-C14 aryl.
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00014
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00015
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00016
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00017
  • In one embodiment, the Linker of the compound is —C(═O)—NH—(CH2)2—NH—.
  • In one embodiment, the Linker of the compound is —CH2—NH—CH2—.
  • In one embodiment, the Linker of the compound is —C(═O)—NH—CH2—.
  • These linkers, as well as the others disclosed herein and those otherwise known in the art, are for use in a compound of the present invention such as the four embodiments depicted above containing a Linker.
  • The present invention provides a method for the treatment of a cancer in which the cancer cells may leave the primary site in an individual who is in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • The present invention provides a method for the treatment of a cancer in which it is desired to mobilize cancer cells from a site into the bloodstream and retain the cancer cells in the bloodstream in an individual who is in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • The present invention provides a method for releasing cells into circulating blood and enhancing retention of the cells in the blood of an individual who is need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent. In an embodiment, the method further includes the step of collecting the cells released. In an embodiment, the step of collecting utilizes apheresis. In an embodiment, the cells are stem cells (e.g., bone marrow progenitor cells). In an embodiment, G-CSF is administered to the individual.
  • The present invention provides a method for the treatment of an inflammatory disease in which the adhesion or migration of cells occurs in the disease in an individual in need of such treatment, comprising administering to the individual a compound of the present invention in an amount effective for treatment, wherein the compound is with or without a pharmaceutically acceptable carrier or diluent.
  • The present invention provides a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically acceptable carrier or diluent.
  • In other embodiments, the above compounds thereof may be used in the manufacture of a medicament, and for any of the uses recited herein.
  • These and other aspects of the present invention will become apparent upon reference to the following detailed description and attached drawings. All references disclosed herein are hereby incorporated by reference in their entirety as if each was incorporated individually.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 (FIG. 1A and FIG. 1B) is a diagram illustrating the synthesis of heterobifunctional Compound #1 (compound 27).
  • FIG. 2 (FIG. 2A, FIG. 2B and FIG. 2C) is a diagram illustrating the synthesis of heterobifunctional Compound #2 (compound 28).
  • FIG. 3 depicts the inhibition of binding of anti-CXCR4-PE to SupT 1 cells in a dose-dependent manner by heterobifunctional Compound #1.
  • FIG. 4 depicts the results of an E-selectin assay in which heterobifunctional Compound #1 is used as the inhibitor.
  • FIG. 5 depicts a comparison of IC50 values of compounds A, B and #1 for E-selectin. Compound A, which is a known E-selectin inhibitor, is compound 15 of Thoma et al. (J. Med. Chem. 42:4909-4913, 1999) and is used as a reference compound. Compound #1 is heterobifunctional Compound #1. Compound B is the glycomimetic portion of Compound #1 (i.e., compound 18 of FIG. 1 except modified to replace COOMe, which is used in the linking process, with H).
  • DETAILED DESCRIPTION
  • As noted above, the present invention provides compounds, compositions and methods for treating diseases in which an E-selectin and a CXCR4 chemokine receptor play a role, and for enhancing retention of cells after releasing into circulating blood. The compounds have a variety of uses in vitro and in vivo.
  • As used herein, the term “E-selectin inhibitor” refers to an inhibitor of E-selectin only, as well as to an inhibitor of E-selectin and either P-selectin or L-selectin, or E-selectin and both P-selectin and L-selectin. Thus, there is E-selectin inhibition regardless of whether there is also inhibition of either P-selectin or L-selectin or both P-selectin and L-selectin.
  • All compounds of the present invention or useful thereto (e.g., for pharmaceutical compositions or methods of treating) include physiologically acceptable salts thereof. Examples of such salts are Na, K, Li, Mg, Ca, and Cl.
  • A compound of the present invention is a heterobifunctional compound wherein an E-selectin inhibitor is linked (i.e., covalently bonded) to a CXCR4 chemokine receptor inhibitor. Such a compound comprises, or consists of, the formula:
  • E-selectin inhibitor-Linker-CXCR4 chemokine receptor inhibitor. Accordingly, the compound functions to inhibit E-selectin and to inhibit the CXCR4 chemokine receptor.
  • E-selectin inhibitors are well known in the art. Some E-selectin inhibitors are specific for E-selectin only. Other E-selectin inhibitors have the ability to inhibit not only E-selectin but additionally P-selectin or L-selectin or both P-selectin and L-selectin. Examples of E-selectin inhibitors (specific for E-selectin or otherwise) are disclosed in U.S. Pat. No. 7,060,685; U.S. Application Publication No. US-2007-0054870; U.S. Application Publication No. US-2008-0161546; and references cited in any of these patent or published application documents. Those examples are small organic molecules. Other known E-selectin inhibitors are amino acid-based, such as antibodies. For example, the humanized monoclonal antibody CDP850 is an E-selectin inhibitor.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00018
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00019
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00020
  • wherein L=end of bond to Linker.
  • In one embodiment of the compound, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00021
  • wherein L=end of bond to Linker.
  • CXCR4 chemokine receptor inhibitors are well known in the art. Such inhibitors will typically prevent the binding of stromal derived factor-1 (SDF-1) to a CXCR4 receptor. Examples of CXCR4 chemokine receptor inhibitors are AMD-3100 (Hendrix et al., Antimicrob. Agents Chemother. 44:1667-1673, 2000); ALX40-4C (Doranz et al., AIDS Research and Human Retroviruses 17:475-486, 2001); and T134 (Arakaki et al., J. Virol. 73:1719-1723, 1999). These examples include a small organic molecule and amino acid-based molecules, such as the T22 peptide. AMD-3100 is a bicyclam. Each of the two cyclam rings is attached to the same phenyl ring (each cyclam ring is para to the other) via a methylene group. In one embodiment of a compound of the present invention, the CXCR4 chemokine receptor inhibitor is a phenyl ring to which is attached only one cyclam ring.
  • In a compound of the present invention, an E-selectin inhibitor and a CXCR4 chemokine receptor inhibitor are covalently joined via a linker (i.e., interposed between the two inhibitors is a “Linker”). A linker may be (or may include) a spacer group, such as —(CH2)p— or —O(CH2)p— where p is generally about 1-20 (including any whole integer range therein). Other examples of spacer groups include a carbonyl or carbonyl containing group such as an amide. An embodiment of such spacer groups is
  • Figure US20130184229A1-20130718-C00022
  • Embodiments of linkers include the following:
  • Figure US20130184229A1-20130718-C00023
  • Other linkers, e.g., polyethylene glycols (PEG) or —C(═O)—NH—(CH2)p—C(═O)—NH2 where p is as defined above, will be familiar to those in the art or in possession of the present disclosure.
  • In another embodiment, the linker is
  • Figure US20130184229A1-20130718-C00024
  • In another embodiment, the linker is
  • Figure US20130184229A1-20130718-C00025
  • In another embodiment, the linker is —C(═O)—NH—(CH2)2—NH—.
  • In another embodiment, the linker is —CH2—NH—CH2—.
  • In another embodiment, the linker is —C(═O)—NH—CH2—.
  • In one embodiment of a compound of the present invention, the E-selectin inhibitor consists of:
  • Figure US20130184229A1-20130718-C00026
  • wherein L is the end of the bond to Linker.
  • In the present disclosure, there are several chemical abbreviations. “Me” is methyl. “Et” is ethyl. “Ar” is aryl. “Bz” is benzoyl.
  • Selection of a substituent at R1 includes H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)OX, alkanyl substituted with C(═O)OX, C(═O)NHX, alkanyl substituted with C(═O)NHX, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)X, OX, NHX, NHC(═O)X, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH.
  • Selection of a substituent at R2 includes —OH,
  • Figure US20130184229A1-20130718-C00027
  • —O—C(═O)—X, —NH2, —NH—C(═O)—NHX, or —NH—C(═O)—X where n=0-2 and X is independently selected from C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl,
  • Figure US20130184229A1-20130718-C00028
  • where Q is H or a physiologically acceptable salt, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, aryl, (CH2)m-aryl where m is 1-10, and where n=0-10, and any of the above ring compounds may be substituted with one to three independently selected of Cl, F, CF3, C1-C8 alkoxy, NO2, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, C1-C14 aryl, or OY, C(═O)OY, NY2 or C(═O)NHY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, or C1-C14 aryl.
  • Selection of a substituent at R3 includes H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, CN, CH2CN, C(═O)X where X is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, NHOH, NHOCH3, NHCN, or NX2, or C(═O)OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl or C1-C8 alkynyl; and
  • Selection of a substituent at R4 includes
  • Figure US20130184229A1-20130718-C00029
  • where the cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or C1-C14 aryl.
  • As used herein, a “C1-C8 alkanyl” refers to an alkane substituent with one to eight carbon atoms and may be straight chain, branched or cyclic (cycloalkanyl). Examples are methyl, ethyl, propyl, isopropyl, butyl and t-butyl. A “halogenated C1-C8 alkanyl” refers to a “C1-C8 alkanyl” possessing at least one halogen. Where there is more than one halogen present, the halogens present may be the same or different or both (if at least three present). A “C1-C8 alkenyl” refers to an alkene substituent with one to eight carbon atoms, at least one carbon-carbon double bond, and may be straight chain, branched or cyclic (cycloalkenyl). Examples are similar to “C1-C8 alkanyl” examples except possessing at least one carbon-carbon double bond. A “C1-C8 alkynyl” refers to an alkyne substituent with one to eight carbon atoms, at least one carbon-carbon triple bond, and may be straight chain, branched or cyclic (cycloalkynyl). Examples are similar to “C1-C8 alkanyl” examples except possessing at least one carbon-carbon triple bond. An “alkoxy” refers to an oxygen substituent possessing a “C1-C8 alkanyl,” “C1-C8 alkenyl” or “C1-C8 alkynyl.” This is —O-alkyl; for example methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy and the like; and alkenyl or alkynyl variations thereof (except for methoxy). It further refers to the group O-alkyl-W-alkyl where W is O or N; for example —O—(CH2)n—W—(CH2)m where n and m are independently 1-10. An “aryl” refers to an aromatic substituent with one to fourteen carbon atoms as ring atoms in one or multiple rings which may be separated by a bond or fused. As used herein, “aryl” includes “heteroaryl.” A “heteroaryl” is similar to an “aryl” except the aromatic substituent possesses at least one heteroatom (such as N, O or S) in place of a ring carbon. Where an aromatic substituent is an aryl in which all the ring atoms are carbon (i.e., not a heteroaryl), there are typically six to fourteen ring atoms. Where an aryl is a heteroaryl, there may be less than six carbon ring atoms. Examples of aryls include phenyl, naphthyl, pyridinyl, pyrimidinyl, triazolo, furanyl, oxazolyl, thiophenyl, quinolinyl and diphenyl.
  • In one embodiment of a compound of the present invention, the CXCR4 chemokine receptor inhibitor consists of:
  • Figure US20130184229A1-20130718-C00030
  • wherein L is the end of the bond to Linker.
  • In one embodiment, the compound has the formula:
  • Figure US20130184229A1-20130718-C00031
  • wherein R1, R2, R3 and R4 are as defined above.
  • In one embodiment in which the linker is —C(═O)—NH—(CH2)2—NH—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00032
  • wherein R1, R2, R3 and R4 are as defined above.
  • In one embodiment in which the linker is —CH2—NH—CH2—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00033
  • wherein R1, R2, R3 and R4 are as defined above.
  • In one embodiment in which the linker is —C(═O)—NH—CH2—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00034
  • wherein R1, R2, R3 and R4 are as defined above.
  • In one embodiment in which the linker is —C(═O)—NH—(CH2)2—NH—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00035
  • In one embodiment in which the linker is —CH2—NH—CH2—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00036
  • In one embodiment in which the linker is —C(═O)—NH—CH2—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00037
  • In one embodiment in which the linker is —C(═O)—NH—(CH2)2—NH—, the compound has the formula:
  • Figure US20130184229A1-20130718-C00038
  • All compounds of the present invention or useful thereto (e.g., for pharmaceutical compositions or methods of treating), include physiologically acceptable salts thereof. Examples of such salts are Na, K, Li, Mg, Ca and Cl.
  • Compounds as described herein may be present within a pharmaceutical composition. A pharmaceutical composition comprises one or more compounds in combination with (i.e., not covalently bonded to) one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions may comprise buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione, adjuvants (e.g., aluminum hydroxide) or preservatives. Within yet other embodiments, compositions of the present invention may be formulated as a lyophilizate. Compositions of the present invention may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, intravenous, intracranial, intraperitoneal, subcutaneous, or intramuscular administration.
  • The compositions described herein may be administered as part of a sustained release formulation (i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration). Such formulations may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site. Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of compound release. The amount of compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • The above-described compounds including equivalents thereof are useful in methods of the present invention. In one embodiment, the compounds may be used in a method for the treatment of a cancer in which the cancer cells may leave the primary site. A primary site may be, for example, solid tissue (e.g., breast or prostate) or the bloodstream. An individual who is in need of such treatment is administered at least one (i.e., one or more) of the above-described compounds in an amount effective for the treatment. In addition to breast cancer and prostate cancer, other examples of infiltrating diseases include lung cancer and melanoma, as well as the hematological malignancies (e.g., leukemias and myelomas). As used herein, the term “treatment” (including variations such as “treating”) includes for the disease or a complication associated with the disease, and includes prevention. For example, a complication associated with the cancer may not have presented itself in an individual with the disease, and a compound may be administered to prevent presentation of the complication in the individual. Complications associated with a cancer in which the cancer cells may leave the primary site include, for example, metastasis and infiltration of cancer cells to other tissues. For example, acute myelogenous leukemia (AML) and multiple myeloma (MM) cells migrate to the endosteal region of the bone marrow where the cells become quiescent and are protected from chemotherapy-induced apoptosis. Administration of a compound described herein may prevent adhesion or migration of cancer cells. Such prevention can result in making the cancer cells more susceptible to treatment with chemotherapy. Administration of a compound described herein in the context of prevention may be to an individual who is at risk of occurrence of a cancer for the first time, or for recurrence of a cancer. For example, while a brain cancer such as glioblastoma multiforme is typically treated with another type of therapy (such as radiation or chemotherapy) for the first occurrence, such therapy is usually not effective to prevent recurrence.
  • The term “treatment” as used herein refers to any of a variety of positive effects from the treatment including, for example, eradicating a complication associated with the disease, relieving to some extent a complication, slowing or stopping progression of the disease, enhancing the effectiveness of one or more therapies for the disease, and prolonging the survival time of the recipient. The treatment may be used in conjunction with one or more other therapies for a cancer or a complication associated therewith.
  • In another embodiment, the above-described compounds including equivalents may be used in a method for the treatment of a cancer in which it is desired to mobilize cancer cells from a site into the bloodstream and retain the cancer cells in the bloodstream. An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment. Examples of cancers for such treatment include leukemias and myelomas (e.g., AML and MM). Mobilizing cancer cells into the bloodstream from a site and retaining the cells therein can result in making the cancer cells more susceptible to treatment with chemotherapy. An example of a site from which to mobilize cancer cells is bone. Cancer cells may, for example, be in circulation and then home to bone. Once in bone, the cancer cells are protected from chemotherapy. A compound described herein may be used, for example, to mobilize cancer cells from bone into the bloodstream and prevent cancer cells from homing to bone, thereby retaining the cancer cells in the bloodstream. Administration of a compound described herein in the context of prevention may be to an individual who is at risk of occurrence of a cancer for the first time, or for recurrence of a cancer. For example, while a brain cancer such as glioblastoma multiforme is typically treated with another type of therapy (such as radiation or chemotherapy) for the first occurrence, such therapy is usually not effective to prevent recurrence.
  • In another embodiment, the above-described compounds including equivalents may be used in a method for releasing cells (such as hematopoietic stem cells) into circulating blood and enhancing retention of the cells in the blood. An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment. One use of the method is, for example, for stem cell harvesting. Stem cells may be needed, for example, after high-dose chemotherapy treatment. Many chemotherapies suppress bone marrow which disrupts the production of certain components of blood in an individual. As a result, the individual may develop a variety of blood cell related disorders and continuation of chemotherapy may be compromised. A compound described herein may be used, for example, to release stem cells into circulating blood and enhance retention of the stem cells in the blood. The method may include a further step of collecting cells that are released. For example, released stem cells may be collected. A variety of techniques are known in the art for collecting cells. For example, apheresis may be utilized. An example of a stem cell is a bone marrow progenitor cell. The release of such cells from bone marrow into circulating blood and retention therein has a variety of uses. For example, the mobilized bone marrow progenitor cells may be collected from the blood. A use of such collected cells is to obtain healthy bone marrow progenitor cells from an individual prior to treatment of the individual in a manner such that bone marrow is suppressed. Following treatment, the individual can receive a bone marrow transplantation utilizing the bone marrow progenitor cells collected prior to treatment. This is useful, for example, where an individual needs to be subjected to a chemotherapy protocol that will suppress bone marrow.
  • It can be desirable to additionally treat an individual with at least one (i.e., one or more) colony stimulating factor. Such a factor may be administered, for example, before or simultaneous with administration of at least one of the above-described compounds. Where administration is simultaneous, the combination may be administered from a single container or two (or more) separate containers. An example of a suitable colony stimulating factor is granulocyte-colony stimulating factor (G-CSF). G-CSF induces the bone marrow to grow and produce more stem cells. A compound described herein aids in releasing stem cells into circulating blood. Stem cells produced in bone marrow and released into circulating blood, as a result of the combination of the administration (separately or together) of a compound described herein and G-CSF, may be collected as described above. Such collected stem cells may be, for example, administered to the individual after chemotherapy. The stem cells return to the bone marrow and produce blood cells. Application of a compound described herein to mobilization and harvesting of healthy bone marrow progenitor cells from bone marrow treated with G-CSF provides cells useful, for example, for bone marrow transplantation.
  • In another embodiment, the above-described compounds including equivalents may be used in a method for the treatment of an inflammatory disease in which the adhesion or migration of cells occurs in the disease. An individual who is in need of such treatment is administered at least one (i.e., one or more) of the compounds in an amount effective for the treatment. Examples of inflammatory diseases include inflammatory skin disorders such as atopic dermatitis and psoriasis. The treatment may reduce (partially or totally) the disease or a complication associated therewith, such as pain. The treatment may be used in conjunction with one or more other therapies for such an inflammatory disease or a complication associated therewith.
  • The above-described compounds may be administered in a manner appropriate to the disease to be treated. Appropriate dosages and a suitable duration and frequency of administration may be determined by such factors as the condition of the patient, the type and severity of the patient's disease and the method of administration. In general, an appropriate dosage and treatment regimen provides the compound(s) in an amount sufficient to provide therapeutic or prophylactic benefit. Within particularly preferred embodiments of the invention, a compound may be administered at a dosage ranging from 0.001 to 1000 mg/kg body weight (more typically 0.01 to 1000 mg/kg), on a regimen of single or multiple daily doses. Appropriate dosages may generally be determined using experimental models or clinical trials. In general, the use of the minimum dosage that is sufficient to provide effective therapy is preferred. Patients may generally be monitored for therapeutic effectiveness using assays suitable for the condition being treated, which will be familiar to those of ordinary skill in the art.
  • At least one (i.e., one or more) of the above-described compounds may be administered in combination with at least one (i.e., one or more) agent, e.g., chemotherapeutic agent or anti-inflammatory agent. In addition, the administration may be in conjunction with one or more other therapies for reducing toxicities of chemotherapy. For example, at least one (i.e., one or more) agent to counteract (at least in part) a side effect of chemotherapy may be administered. At least one compound described herein may be administered before, after or simultaneous with administration of at least one chemotherapeutic agent or anti-inflammatory agent. Where administration is simultaneous, the combination may be administered from a single container or two (or more) separate containers.
  • The following Examples are offered by way of illustration and not by way of limitation.
  • Example 1 Synthesis of Heterobifunctional Compound #1 (Compound 27 of FIG. 1)
  • Synthesis of Compound 2:
  • Commercially available (Aldrich Chemical Co., Milwaukee, Wis.) cis-1,2,3,6-tetrahydrophthalic anhydride (compound 1, 50 g) is added to a suspension of amberlyste 15 (50 g, dried under vacuum) in methanol (1 L) with stirring. Triethylorthoformate (100 ml) is added immediately while stirring. The reaction mixture is then vigorously stirred for 5 days at room temperature and additional triethylorthoformate is added. Stirring is continued for an additional 4 days, then the reaction mixture filtered over celite and washed with methanol. The solvent is removed in vacuum and the residue is dissolved in CH2Cl2 (200 ml). The solution is washed with a cold saturated solution of NaHCO3 (200 ml) and cold brine (200 ml). The organic layer is dried (Na2SO4), filtered and concentrated to dryness to afford compound 2 (55 g).
  • Synthesis of Compound 3:
  • To a suspension of compound 2 (10 g) in phosphate buffer (400 ml, pH 7) is added PLE (40 mg, 1080 unit). The pH of the mixture is maintained at 7 by continuous dropwise addition of 1M NaOH solution via syringe pump. The reaction is stirred at 20° C. until 1 equivalent of NaOH (50 ml) is used. The reaction mixture is transferred to a reparatory funnel and EtOAc (400 ml) is added. The layers are separated and the organic layer is extracted with phosphate buffer (2×250 ml, pH 7). The combined aqueous layers are acidified (pH 2) with aqueous HCl (1M) and extracted with EtOAc (3×400 ml). The combined organic layers are dried (Na2SO4), filtered and concentrated to dryness to afford compound 3 (7.8 g).
  • Synthesis of Compound 4:
  • To a solution of compound 3 (2 g) in dry CH2Cl2 (35 ml) is added (COCl)2 (1.4 ml) and DMF (0.025 ml) and stirred for 3 h at RT. The solution is evaporated to dryness (rotavapor is purged with argon). The residue is dissolved in dry THF (40 ml) and is added dropwise over a period of 20 min to a boiling suspension of 2-mercaptopyridine-1-oxide sodium salt (2 g), t-BuSH (6 ml), and 4-DMAP (52 mg) in dry THF (100 ml). The solution is stirred under reflux for 3 h. The reaction mixture is cooled down to RT and transferred into a separatory funnel with EtOAc (100 ml) and washed with H2O (100 ml). The aqueous layer is extracted with EtOAc (2×200 ml). The combined organic layers are dried (Na2SO4), filtered and concentrated to dryness. The crude product is purified by column chromatography (silica) to afford compound 4 as yellowish oil (1.1 g).
  • Synthesis of Compound 5:
  • To a suspension of compound 4 (4 g) in phosphate buffer (400 ml, pH 7) is added PLE (42 mg) with stirring. The pH is kept at 7 by adding NaOH solution (1M) via syringe pump. The reaction mixture is stirred at RT until 1 equivalent of NaOH is used. The reaction mixture is transferred to a separatory funnel and washed with EtOAc (2×250 ml). The layers are separated and the organic layers are extracted with phosphate buffer (2×250 ml, pH 7). The combined aqueous layers are acidified to pH 2 with aqueous HCl solution and extracted with EtOAc (3×300 ml). The combined organic layers are dried (Na2SO4), filtered and evaporated to dryness. The crude product is filtered through a short plug of silica to afford compound 5 (3 g).
  • Synthesis of Compound 6:
  • Compound 5 (4 g) is suspended in water (90 ml) and cooled down to 0° C. NaHCO3 (8 g) is added followed by a solution of KI (32 g) and I2 (8 g) in water (75 ml). The reaction mixture is stirred at RT for 24 h and then extracted with CH2Cl2 (3×30 ml). The combined organic layers are washed with a saturated solution of Na2S2O3 in water (125 ml). The aqueous layer is extracted with CH2Cl2 (2×30 ml). The combined organic layers are protected from light, dried (Na2SO4), filtered, and concentrated to dryness and quickly under high vacuum to afford iodolactone 6 as an off-white solid (7.5 g).
  • Synthesis of Compound 7:
  • Compound 6 (7 g) is dissolved in dry THF (170 ml) and DBU (7 ml) is added. The reaction mixture is refluxed for 20 h and then cooled down to RT. Diethyl ether (100 ml) is added and transferred into a separatory funnel and extracted with an aqueous solution of HCl (200 ml, 0.5M). The aqueous layers are extracted with Et2O (3×100 ml). The combined organic layers are washed with brine (200 ml), dried (Na2SO4), filtered, and concentrated to dryness. The crude product is purified by column chromatography (silica gel) to afford compound 7 (3.7 g).
  • Synthesis of Compound 8:
  • NaHCO3 (2.2 g) is dried under vacuum and then dry MeOH (132 ml) is added with stirring followed by compound 7 (3 g). The reaction mixture is then stirred at RT under argon for 12 h. The solvent is evaporated off and the residue is transferred into a separatory funnel with CH2Cl2 (35 ml), extracted with water (40 ml), and with brine (40 ml). The aqueous layer is extracted with CH2Cl2 (2×35 ml). The combined organic layers are dried (Na2SO4), filtered, and concentrated to dryness to give compound 8 (5 g).
  • Synthesis of Compound 9:
  • To a solution of compound 8 (4 g) in dry CH2Cl2 (80 ml) is added tert-butyldimethylsilyl chloride (7.2 ml) in small portions, followed by DBU (9.5 ml). The reaction mixture is stirred for 12 h and then quenched with MeOH (12 ml). The reaction mixture is transferred into a separatory funnel with CH2Cl2 (60 ml), washed with cold saturated solution of NaHCO3 (50 ml) and cold brine (50 ml). The aqueous layers are extracted with CH2Cl2 (2×50 ml). The combined organic layers are dried (Na2SO4), filtered and concentrated to dryness. The residue is purified by column chromatography (silica) to give compound 9 (6 g).
  • Synthesis of Compound 10:
  • To a cold (10° C.) solution of compound 9 (5 g) in CH2Cl2 (125 ml) is added m-CPBA (8 g) with stirring, and stirring is continued for 15 h at 10° C. The temperature is raised to RT over a period of 2 h and the mixture is diluted with CH2Cl2 (400 ml). The mixture is transferred into a separatory funnel, and washed with cold saturated solution of Na2S2O3 solution in water (2×400 ml). The organic layer is successively washed with cold saturated solution NaHCO3 (400 ml) and cold brine (100 ml). The aqueous layers are extracted with CH2Cl2 (2×400 ml). The combined organic layers are dried (Na2SO4), filtered, and concentrated to dryness. The crude product is purified by column chromatography (silica) to give compound 10 (4 g).
  • Synthesis of Compound 11:
  • CuCN (1.5 g) is dried in high vacuum at 150° C. for 30 min, suspended in dry THF (25 ml) and cooled down to −78° C. MeLi (1.6 M in Et2O, 22.5 ml) is added slowly via syringe and the temperature is raised to −10° C. over a period of 30 min. The mixture is again cooled down to −78° C., followed by the addition of BF3 etherate (1.4 ml) in THF (5 ml). After stirring for 20 min, compound 10 (1 g) in THF (25 ml) is added and stirring is continued for 5 h at −78° C. The excess of MeLi is quenched with a mixture of MeOH (10 ml) and Et3N (10 ml). The mixture is diluted with Et2O (250 ml) and transferred into a reparatory funnel and extracted with aqueous 25% NH3/satd. NH4Cl (1:9) solution. The organic layer is successively washed with brine (150 ml), 5% AcOH (150 ml), saturated solution of NaHCO3 (150 ml), and brine (150 ml). The aqueous layers are extracted with Et2O (2×250 ml). The combined organic layers are dried (Na2SO4), filtered, and concentrated to dryness. The crude product is purified by column chromatography (silica) to give compound 11 (800 mg).
  • Synthesis of Compound 13:
  • A solution of Br2 (0.08 ml) in CH2Cl2 (1 ml) is added dropwise at 0° C. to a solution of commercially available (Carbosynth Ltd., Compton, Berkshire, UK) compound 12 (640 mg) in CH2Cl2 (10 ml) and stirred at 0° C. for 1 h. Cyclohexene (0.02 ml) is added and the reaction mixture is stirred for anther 30 min. The mixture is added dropwise to a solution of 11 (310 mg) and Et4NBr (280 mg, oven dried at 200° C.) in DMF/CH2Cl2 (20 ml, 1:1) containing molecular sieve (1 g, 3 A) with stirring at RT. The stirring is continued for 60 h. The reaction is quenched with pyridine (2 ml), filtered over celite, and washed with CH2Cl2 (20 ml). The solution is washed with brine (50 ml) and the aqueous layer is extracted 3 times with CH2Cl2 (3×50 ml). The combined organic layers are dried (Na2SO4), filtered, and concentrated to dryness. The crude product is purified by column chromatography (silica) to give compound 13 (144 mg).
  • Synthesis of Compound 14:
  • To a solution of compound 13 (140 mg) in THF (5 ml), TBAF (0.39 ml) is added. After 24 h additional TBAF (0.2 ml) is added and the stirring is continued for 50 h. The reaction mixture is concentrated to dryness and the crude product is purified by column chromatography (silica) to afford compound 14 (95 mg).
  • Synthesis of Compound 16:
  • A mixture of compound 14 (0.16 g) and compound 15 (0.35 g, synthesized as described by Banteli et al., Helvetica Chimica Acta 83:2893-2907, 2000) is co-evaporated with toluene twice and then dried under vacuum. The mixture is dissolved in dry CH2Cl2 (10 ml) and stirred with flame dried molecular sieve (4 A) and 2,6-di-tert-Bu-pyridine (0.59 ml) for 30 min at RT. The reaction mixture is cooled to 0° C. and MeOTf (0.25 ml) is added with stirring. The reaction mixture is stirred for 4 h at RT, filtered through a bed of Celite, washed with CH2Cl2 (2×10 ml) and then transferred to a reparatory funnel. The organic layer is washed with a cold saturated solution of NaHCO3 (25 ml) and brine (25 ml), dried (Na2SO4), filtered, and concentrated to dryness. The residue is purified by column chromatography (silica) to give compound 16 (0.23 g).
  • Synthesis of Compound 17:
  • Compound 16 (0.96 mg) is dissolved in dioxane-water (10:2, 12 ml) and AcOH (0.2 ml) is added. 10% Pd/C (0.8 g) is added and stirred vigorously under hydrogen (40 psi) for 16 h at RT. The reaction mixture is filtered through a bed of Celite and washed with MeOH. Solvent is evaporated off to give compound 17 (700 mg).
  • Synthesis of Compound 18:
  • Compound 17 (500 mg) is treated at RT with 0.01N NaOMe in MeOH (20 ml) for 1 h. The reaction is neutralized with AcOH and the solvent is evaporated off to give compound 18 (300 mg).
  • Synthesis of Compound 19:
  • Compound 18 (200 mg) is dissolved in ethylenediamine (3 ml) and the solution is stirred for 3 h at 70° C. Solvent is evaporated off and the residue is purified by Sep-Pak C18 column to give compound 19 (160 mg).
  • Synthesis of Compound 21:
  • Commercially available (Aldrich Chemical Co., Milwaukee, Wis.) compound 20 (1.47 g) is suspended in CH2Cl2 (70 ml). To this suspension is added a solution of (BOC)2O (3.86 g in 70 ml of CH2Cl2) dropwise with stirring at RT. The stirring is continued for 2 h. The reaction mixture is concentrated to dryness and purified by column chromatography (CombiFlash) to give compound 21 (1.8 g).
  • Synthesis of Compound 23:
  • A suspension of compound 21 (1.59 g), commercially available (Aldrich Chemical Co., Milwaukee, Wis.) compound 22 (0.8 g) and K2CO3 (0.48 g) in DMF (15 ml) is stirred at 60° C. overnight. The reaction mixture is concentrated to a thick oil and filtered through a glass syringe filter, dissolved in CH2Cl2 and purified by column chromatography (silica) to give compound 23 (1.96 g).
  • Synthesis of Compound 24:
  • To a cold (0° C.) solution of compound 23 (0.99 g) in THF (30 ml) is added LiAlH4 (2M solution in THF, 3.05 ml) with stirring. Stirring is continued for 2 h at 0° C. The reaction is quenched with EtOAc and diethylether is added. The mixture is transferred to a reparatory funnel and washed with cold saturated solution of NH4Cl. The organic layer is dried (Na2SO4), filtered, and concentrated to dryness. The residue is purified column chromatography (CombiFlash) to give compound 24 (717 mg).
  • Synthesis of Compound 25:
  • A solution of (COCl)2 (0.15 ml) in CH2Cl2 (3 ml) is cooled down to −78° C. To this solution is added DMSO (0.25 ml) dropwise in the cold (−78° C.) with stirring and stirring is continued for 15 min at −78° C. Compound 24 (717 mg) in CH2Cl2 (3 ml) is added dropwise to the above mixture at −78° C. with stirring. The stirring is continued for 15 min at −78° C. and DIPEA (1.17 ml) is added and stirred for 15 min. The reaction mixture is warmed to RT slowly. The reaction mixture is concentrated to dryness and the crude product is purified by column chromatography (silica) to give compound 25 (701 mg).
  • Synthesis of Compound 26:
  • Compound 25 (77 mg) is dissolved in CH2Cl2 (7 ml) and CF3COOH (1.4 ml) is added with stirring. The reaction mixture is stirred at RT for 2 h, CF3COOH (0.7 ml) is added and stirring is continued for another 1 h. The reaction mixture is evaporated to dryness and purified by Sep-Pak C18 Cartridges to give compound 26 (30 mg).
  • Synthesis of Compound 27:
  • To a solution of compound 19 (5 mg) in DMSO (0.2 ml) is added compound 26 (4 mg) and NaBH3CN (0.8 mg, 0.08 ml from a stock solution of 10 mg/ml) and AcOH (0.8 mg, 0.08 ml from a stock solution of 10 mg/ml). The reaction mixture is stirred at 60° C. for 2 h and the solvent is evaporated off. The residue is purified by HPLC (reverse phase C18 column) to give compound 27 (2.5 mg) which is heterobifunctional Compound #1 (also referred to herein as “Compound #1”).
  • Example 2 Synthesis of Heterobifunctional Compound #2 (Compound 28 of FIG. 2)
  • Synthesis of Intermediate II:
  • (−)-Shikimic acid (20 g) in MeOH (200 ml) and sulfuric acid (2 ml, 98%) are stirred at rt for 50 h. The reaction mixture is neutralized with 2N aqueous NaOH in the cold. After evaporation to dryness, the residue is purified by silica gel chromatography to afford II (19.2 g).
  • Synthesis of Intermediate (III):
  • Methyl shikimate (II, 10 g), 2,2 dimethoxypropane (10 ml) and p-TsOH (0.8 g) are dissolved in acetonitrile (125 ml) and stirred at rt for 1 h. The reaction mixture is then neutralized with triethylamine (2 ml) and evaporated to dryness. The residue is chromatographed on silica gel to yield III (11 g).
  • Synthesis of Intermediate IV:
  • The shikimic acid derivative III (10 g) and PtO2/C (10%, 250 mg) in MeOH (40 ml) are hydrogenated at rt under vigorous stirring. After 16 h the reaction mixture is filtered over celite and evaporated to dryness. The residue is chromatographed on silica gel to yield IV.
  • Synthesis of Intermediate V:
  • To a solution of IV (8 g) in DCM (100 ml) at 0° C. are added pyridine (12 ml), acetic anhydride (7 ml) and a DMAP (25 mg). The reaction mixture is stirred at rt for 1 h, and diluted with EtOAc (250 ml). After washing with 0.5 M aqueous HCl (3×50 ml), saturated solution of KHCO3 (3×50 ml) and brine (3×50 ml), the combined organic layers are dried (Na2SO4) and evaporated to dryness. The residue is purified by chromatography on silica gel to yield V (6.8 g).
  • Synthesis of Intermediate VI:
  • A solution of V (6.0 g) in acetic acid (30 ml, 80%) is stirred at 80° C. for 1 h. Solvent is evaporated off and the residue is purified by chromatography on silica gel (DCM/MeOH 14:1) to yield VI (3.6 g).
  • Synthesis of Intermediate (VII):
  • A solution of VI (3 g) and p-TsCl (3.5 g) in pyridine (30 ml) is stirred at rt for 6 h. MeOH (5 ml) is added and the solvent is evaporated at reduced pressure, the residue dissolved in EtOAc (3×150 ml) and the organic layers are washed with 0.5 M aqueous HCl (0° C.), water (cold) and brine (cold). The combined organic layers are dried (Na2SO4), filtered on Celite and evaporated to dryness. The residue is purified by chromatography on silica gel (toluene/EtOAc 4:1) to yield VII (3.7 g).
  • Synthesis of Compound VIII:
  • A solution of VII (3 g) and NaN3 (2.5 g) in DMF (20 ml) is stirred at 80° C. The reaction mixture is cooled to rt and diluted with EtOAc (200 ml) and water (50 ml). The organic layer is additionally washed twice with water (2×50 ml) and once with brine (50 ml). All aqueous layers are extracted twice with EtOAc (2×50 ml). The combined organic layers are dried with Na2SO4, filtered and the solvent is evaporated off. The residue is purified by chromatography on silica gel (petroleum ether/EtOAc 5:2) to give VIII (2.2 g).
  • Synthesis of Compound X:
  • To a solution of ethyl 2,3,4-tri-O-benzyl-α-L-fucothiopyanoside IX (1.5 g) in DCM (3 ml), bromine (150 μl) is added at 0° C. under argon. After 5 min the cooling bath is removed and the reaction mixture is stirred for additional 25 min at rt. Cyclohexene (200 μl) is added and the reaction mixture is added to a solution of VIII (400 mg), (Et)4NBr (750 mg) and powdered 4 Å molecular sieves in DCM (10 ml) and DMF (5 ml). After 16 h, triethylamine (1.5 ml) is added and stirred for an additional for 10 min, diluted with EtOAc (50 ml) and washed with sat. aqueous NaHCO3, water and brine. The aqueous layers are extracted twice with EtOAc (2×50 ml). The combined organic layers are dried (Na2SO4), filtered and evaporated to dryness. The residue is purified by chromatography on silica gel (toluene/EtOAc 9:1) to yield X (700 mg).
  • Synthesis of Compound XI:
  • To a solution of X (1.5 g) in MeOH (20 ml) is added freshly prepared NaOMe (80 mg) and the reaction mixture is stirred in a pressure tube at 80° C. for 20 h. The reaction mixture is cooled to rt and neutralized with acetic acid. Solvent is evaporated to dryness and the residue is dissolved in ether. Freshly prepared diazomethane is added and the excess diazomethane is neutralized with acetic acid. Solvent is evaporated off to give XI (1.25 g).
  • Synthesis of Building Block XV:
  • This synthesis is done exactly in same way as described previously (Helvetica Chemica Acta 83:2893-2907 (2000)).
  • Synthesis of Compound XVI:
  • A mixture of XI (1.6 g), XV (3 g) and activated powdered molecular sieves 4 Å (1 g) in DCM (17 ml) is stirred at rt under argon for 2 h. Then DMTST (2 g) is added in 4 equal portions over a period of 1.5 h. After 24 h the reaction mixture is filtered over Celite and the filtrate is diluted with DCM (100 ml). The organic layer is washed with sat. aqueous NaHCO3 and brine and the aqueous layers are extracted twice with DCM. The combined organic layers are dried (Na2SO4), filtered and evaporated to dryness. The residue is purified by chromatography on silica gel (toluene/EtOAc 8:1) to yield XVI (1.5 g).
  • Synthesis of Compound XVII:
  • To a solution of XVI (500 mg) and orotic acid chloride (500 mg) in dichloromethane (10 ml) is added a solution of triphenylphosphine (500 mg in 5 ml dichloromethane) dropwise during 10 min. The reaction mixture is stirred at rt for 25 h and the solvent is evaporated off. The residue is purified (chromatography on silica gel DCM/MeOH 19:1) to give XVII (250 mg).
  • Synthesis of Compound XVIII:
  • To a solution of XVII (200 mg) in dioxane-water (5:1, 12 ml) is added 10% Pd—C (100 mg) and the reaction mixture is stirred vigorously under hydrogen (55 psi) for 24 h. Catalyst is filtered through a bed of celite and the solvent is evaporated off. Residue is purified by silica gel chromatography to give compound XVIII (150 mg).
  • Synthesis of XIX:
  • To a solution of compound XVIII (145 mg) in MeOH (5 ml) is added a solution of NaOMe in MeOH (25%, 0.025 ml) and the reaction mixture is stirred at rt for 4 h, neutralized with acetic acid and the solvent is evaporated off. Residue is dissolved in water and passed through a bed of Dowex 50wX-8 (Na-form) resin. Water wash is evaporated off to afford compound XIX (100 mg).
  • Synthesis of EDA-XIX:
  • XIX (80 mg) is heated at 70° C. with ethylenediamine (EDA) (1 ml) with stirring for 5 h. Solvent is evaporated off and the purified by sephadex G-25 column to give EDA-XIX (82 mg).
  • Synthesis of Compound 28:
  • Compound 26 of Example 1 is reacted with EDA-XIX (and the product purified) using the procedures described in Example 1 (for the synthesis of compound 27) to give compound 28 which is heterobifunctional Compound #2 (also referred to herein as “Compound #2”).
  • Example 3 Assay to Assess Binding of Compounds to CXCR4 Methods
  • The assay assesses the ability of glycomimetic compounds to inhibit binding of an anti-CXCR4 antibody conjugated to phycoerythrin (“PE”), to CXCR4 on the surface of SupT1 cells. SupT1 cells are a T lymphoblast derived from a lymphoblastic leukemia and constitutively express CXCR4 on the cell surface. The cells are purchased from ATCC (ATCC number CRL-1942). Anti-human CXCR4-phycoerythrin monoclonal antibody (anti-CXCR4-PE) is purchased from R&D Systems (catalog number FAB 170P, clone 12G5). The cells are grown in RPMI 1640 medium supplemented with 10% FBS. Approximately 2×106 cells are washed three times by centrifuging the cells at 400×g for 10 minutes and the cell pellet is resuspended in PBS plus 0.05% BSA. After the third centrifugation, the cell pellet is resuspended in PBS plus BSA to a concentration of 5×105 cells per ml. To block non-specific binding, human Ig is added to the cells to a concentration of 1 μg per 105 cells. Next, 200 μl (1×105 cells) are added to 5 ml polypropylene round-bottom tubes (Falcon 2063 tubes). Compound #1 (Example 1) (lot 31-190) is added to the cells at final concentrations of 0.5, 5, 10, and 50 μM. To achieve a final concentration of 0.5 μM, 2.2 μl of 50 μM Compound #1 plus 19.8 μl of PBS/BSA are added to 200 μl of cells. To achieve a final concentration of 5 μM, 22 μl of 50 μM Compound #1 are added to 200 μl of cells. To achieve a final concentration of 10 μM, 4.4 μl of 500 μM Compound #1 plus 17.6 μl of PBS/BSA are added to 200 μl of cells. To achieve a final concentration of 50 μM, 22 μl of 500 μM Compound #1 are added to 200 μl of cells. Other aliquots of cells are treated with either 1 or 5 μM of the bicyclam CXCR4 antagonist AMD-3100 (Sigma Aldrich, catalog #A5602). To achieve a final concentration of 1 μM AMD-3100, 4.4 μl of 50 μM AMD-3100 plus 17.6 μl of PBS/BSA are added to 200 μl of cells and to achieve a final concentration of 5 μM, 22 μl of 50 μM AMD-3100 are added to 200 μl of cells. In addition, one tube of cells is treated with 1 μg/ml of SDF-1α (R&D Systems catalog #350-NS), the natural ligand of CXCR4. The tubes are placed at 4° C. for 15 minutes. Subsequently, each tube receives 10 μl of anti-CXCR4-PE, except one tube of cells receives 10 μl of mouse IgG2a isotype control antibody. The tubes are incubated at 4° C. for 45 minutes. The cells are washed twice with PBS plus 0.05% BSA and the final cell pellet is resuspended in 100 μl of PBS/BSA. To fix the samples, 100 μl of 2% formaldehyde (Polysciences, Inc. ultrapure EM grade, catalog number 04018) are added to each tube. Flow cytometry is performed using a Cytomation MoFlo instrument.
  • Results
  • As shown in the table below, Compound #1 inhibits binding of anti-CXCR4-PE to SupT1 cells in a dose-dependent manner with an IC50 of 8.25 μM (FIG. 3). SDF-1α efficiently inhibits binding of the antibody to CXCR4.
  • Mean Median
    fluorescence fluorescence
    % positive intensity intensity
    SupT1 cells only 1.13 2.80 2.55
    Isotype control 1.46 4.73 2.46
    No inhibitor 99.28 104.75 86.60
    0.5 μM Compound #1 98.69 68.49 54.25
    5 μM Compound #1 81.48 22.25 13.34
    10 μM Compound #1 42.06 14.08 6.04
    50 μM Compound #1 12.80 11.08 3.92
    1 μM AMD-3100 57.57 15.60 7.77
    5 μM AMD-3100 12.42 9.98 3.92
    1 μg/ml SDF-1α 12.37 10.25 4.07
  • Example 4 E-Selectin Activity Binding Assay Methods
  • The inhibition assay to screen glycomimetic antagonists of E-selectin is a competitive binding assay, which allows the determination of IC50 values. Briefly, E-selectin/Ig chimera is immobilized by incubation at 37° C. in 96 well microtiter plates for 2 hours. To reduce nonspecific binding, bovine serum albumin is added to each well and incubated at room temperature for 2 hours. The plate is washed and serial dilutions of the test compounds are added to the wells in the presence of conjugates of biotinylated, sLea polyacrylamide with streptavidin/horseradishperoxidase and incubated for 2 hours at room temperature. To determine the amount of sLea bound to immobilized E-selectin after washing, the peroxidase substrate, 3,3′,5,5′ tetramethylbenzidin (TMB) is added. After 3 minutes, the enzyme reaction is stopped by the addition of H3PO4 and the absorbance of light at a wavelength of 450 nm is determined. The concentration of test compound required to inhibit binding by 50% is determined and reported as the IC50 value for each glycomimetic E-selectin antagonist. In addition to reporting the absolute IC50 value as measured above, relative IC50 values are determined by a ratio of the IC50 measured for the test compound to that of an internal control (reference) stated for each assay.
  • Results
  • The results for heterobifunctional Compound #1 are shown in FIGS. 4 and 5. Compound #1 is a potent E-selectin antagonist (as well as possessing anti-CXCR4 activity—FIG. 3).
  • All the above U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications, and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference in their entirety.
  • From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.

Claims (23)

We claim the following:
1. A heterobifunctional compound for inhibition of E-selectin and the CXCR4 chemokine receptor, comprising E-selectin inhibitor-Linker-CXCR4 chemokine receptor inhibitor, or a physiologically acceptable salt thereof.
2. The compound of claim 1 wherein the E-selectin inhibitor consists of:
Figure US20130184229A1-20130718-C00039
wherein:
L=end of bond to Linker;
R1=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)OX, alkanyl substituted with C(═O)OX, C(═O)NHX, alkanyl substituted with C(═O)NHX, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)X, OX, NHX, NHC(═O)X, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH;
R2=—OH,
Figure US20130184229A1-20130718-C00040
 —O—C(═O)—X, —NH2, —NH—C(═O)—NHX, or —NH—C(═O)—X where n=0-2 and X is independently selected from C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl,
Figure US20130184229A1-20130718-C00041
 where Q is H or a physiologically acceptable salt, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, aryl, (CH2)m-aryl where m is 1-10, and where n=0-10, and any of the above ring compounds may be substituted with one to three independently selected of Cl, F, CF3, C1-C8 alkoxy, NO2, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, C1-C14 aryl, or OY, C(═O)OY, NY2 or C(═O)NHY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, or C1-C14 aryl;
R3=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, CN, CH2CN, C(═O)X where X is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, NHOH, NHOCH3, NHCN, or NX2, or C(═O)OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl or C1-C8 alkynyl; and
Figure US20130184229A1-20130718-C00042
 where the cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C1-C8 alkanyl, C1-C8 alkenyl, C alkynyl or OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or C1-C14 aryl.
3. The compound of claim 2 wherein the E-selectin inhibitor consists of:
Figure US20130184229A1-20130718-C00043
wherein L=end of bond to Linker.
4. The compound of claim 2 wherein the E-selectin inhibitor consists of:
Figure US20130184229A1-20130718-C00044
wherein L=end of bond to Linker.
5. The compound of claim 2 wherein the E-selectin inhibitor consists of:
Figure US20130184229A1-20130718-C00045
wherein L=end of bond to Linker.
6. The compound of claim 2 wherein the E-selectin inhibitor consists of:
Figure US20130184229A1-20130718-C00046
wherein L=end of bond to Linker.
7. The compound of claim 1 wherein the CXCR4 chemokine receptor inhibitor consists of:
Figure US20130184229A1-20130718-C00047
wherein L=end of bond to Linker.
8. The compound of claim 1 having the formula:
Figure US20130184229A1-20130718-C00048
wherein:
R1=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, OH, or NHX where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)OX, alkanyl substituted with C(═O)OX, C(═O)NHX, alkanyl substituted with C(═O)NHX, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH; C(═O)X, OX, NHX, NHC(═O)X, where X═H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, halogenated C1-C8 alkanyl, aryl which may be substituted with one or more of Me, OMe, halide, or OH;
R2=—OH,
Figure US20130184229A1-20130718-C00049
 —O—C(═O)—X, —NH2, —NH—C(═O)—NHX, or —NH—C(═O)—X where n=0-2 and X is independently selected from C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl,
Figure US20130184229A1-20130718-C00050
 where Q is H or a physiologically acceptable salt, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, aryl, (CH2)m-aryl where m is 1-10, and where n=0-10, and any of the above ring compounds may be substituted with one to three independently selected of Cl, F, CF3, C1-C8 alkoxy, NO2, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, C1-C14 aryl, or OY, C(═O)OY, NY2 or C(═O)NHY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, or C1-C14 aryl;
R3=H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, CN, CH2CN, C(═O)X where X is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl, NHOH, NHOCH3, NHCN, or NX2, or C(═O)OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl or C1-C8 alkynyl; and
Figure US20130184229A1-20130718-C00051
 where the cyclopropane ring may be substituted with one to two, and the cyclohexane ring may be substituted with one to three, independently selected of Cl, F, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or OY where Y is H, C1-C8 alkanyl, C1-C8 alkenyl, C1-C8 alkynyl or C1-C14 aryl.
9. The compound of claim 1 having the formula:
Figure US20130184229A1-20130718-C00052
10. The compound of claim 1 having the formula:
Figure US20130184229A1-20130718-C00053
11. The compound of claim 1 having the formula:
Figure US20130184229A1-20130718-C00054
12. The compound of claim 1 having the formula:
Figure US20130184229A1-20130718-C00055
13. The compound of claim 1 wherein the Linker is —C(═O)—NH—(CH2)2—NH—, —CH2—NH—CH2—, or —C(═O)—NH—CH2—.
14. The compound of claim 2 wherein the Linker is —C(═O)—NH—(CH2)2—NH—, —CH2—NH—CH2—, or —C(═O)—NH—CH2—.
15. The compound of claim 7 wherein the Linker is —C(═O)—NH—(CH2)2—NH—, —CH2—NH—CH2—, or —C(═O)—NH—CH2—.
16. A method for the treatment of a cancer in which cancer cells may leave the primary site in an individual who is in need of such treatment, said method comprising administering to the individual the compound of claim 1 in an amount effective for treatment.
17. A method for the treatment of a cancer in which it is desired to mobilize cancer cells from a site into the bloodstream and retain the cancer cells in the bloodstream in an individual who is in need of such treatment, said method comprising administering to the individual the compound of claim 1 in an amount effective for treatment.
18. A method for releasing cells into circulating blood and enhancing retention of the cells in the blood of an individual who is need of such treatment, said method comprising administering to the individual the compound of claim 1 in an amount effective for treatment.
19. The method of claim 18, further including the step of collecting the cells released.
20. The method of claim 19 wherein the step of collecting utilizes apheresis.
21. The method of claim 18 wherein the cells are bone marrow progenitor cells.
22. A method for the treatment of an inflammatory disease in which the adhesion or migration of cells occurs in the disease in an individual who is in need of such treatment, said method comprising administering to the individual the compound of claim 1 in an amount effective for treatment.
23. A pharmaceutical composition comprising the compound of claim 1 and a pharmaceutically acceptable carrier or diluent.
US13/785,439 2009-05-01 2013-03-05 Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors Abandoned US20130184229A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/785,439 US20130184229A1 (en) 2009-05-01 2013-03-05 Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US17458009P 2009-05-01 2009-05-01
US12/768,173 US8410066B2 (en) 2009-05-01 2010-04-27 Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors
US13/785,439 US20130184229A1 (en) 2009-05-01 2013-03-05 Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/768,173 Continuation US8410066B2 (en) 2009-05-01 2010-04-27 Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors

Publications (1)

Publication Number Publication Date
US20130184229A1 true US20130184229A1 (en) 2013-07-18

Family

ID=42310713

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/768,173 Active 2031-02-22 US8410066B2 (en) 2009-05-01 2010-04-27 Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors
US13/785,439 Abandoned US20130184229A1 (en) 2009-05-01 2013-03-05 Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/768,173 Active 2031-02-22 US8410066B2 (en) 2009-05-01 2010-04-27 Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors

Country Status (7)

Country Link
US (2) US8410066B2 (en)
EP (1) EP2424544A1 (en)
JP (1) JP5726171B2 (en)
CN (1) CN102421441B (en)
AU (1) AU2010241807B2 (en)
CA (1) CA2760292A1 (en)
WO (1) WO2010126888A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9109002B2 (en) 2011-12-22 2015-08-18 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
WO2016089872A1 (en) * 2014-12-03 2016-06-09 Glycomimetics, Inc. Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors
US9867841B2 (en) 2012-12-07 2018-01-16 Glycomimetics, Inc. Compounds, compositions and methods using E-selectin antagonists for mobilization of hematopoietic cells
US11072625B2 (en) 2016-10-07 2021-07-27 Glycomimetics, Inc. Highly potent multimeric e-selectin antagonists
US11197877B2 (en) 2017-03-15 2021-12-14 Glycomimetics. Inc. Galactopyranosyl-cyclohexyl derivauves as E-selectin antagonists
US11291678B2 (en) 2016-03-02 2022-04-05 Glycomimetics, Inc Methods for the treatment and/or prevention of cardiovascular disease by inhibition of E-selectin
US11433086B2 (en) 2016-08-08 2022-09-06 Glycomimetics, Inc. Combination of T-cell checkpoint inhibitors with inhibitors of e-selectin or CXCR4, or with heterobifunctional inhibitors of both E-selectin and CXCR4
US11548908B2 (en) 2017-12-29 2023-01-10 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11707474B2 (en) 2018-03-05 2023-07-25 Glycomimetics, Inc. Methods for treating acute myeloid leukemia and related conditions
US11712446B2 (en) 2017-11-30 2023-08-01 Glycomimetics, Inc. Methods of mobilizing marrow infiltrating lymphocytes and uses thereof
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012037034A1 (en) 2010-09-14 2012-03-22 Glycomimetics, Inc. E-selectin antagonists
WO2012061662A1 (en) * 2010-11-03 2012-05-10 Glycomimetics, Inc. Glycomimetic-peptidomimetic inhibitors of e-selectins and cxcr4 chemokine receptors
CA2888527A1 (en) * 2012-10-31 2014-05-08 Glycomimetics, Inc. E-selectin antagonists compounds and methods of use
WO2016164394A1 (en) * 2015-04-08 2016-10-13 Glycomimetics, Inc. 2-halo-galactose-containing selectin antagonists
EP3370724A2 (en) 2015-11-03 2018-09-12 GlycoMimetics, Inc. Antibodies for targeting cancer stem cells and treating aggressive cancers
EP3893936A2 (en) 2018-12-10 2021-10-20 GlycoMimetics, Inc. Methods of treating hiv and aids and the elimination of latent reservoirs of hiv infection using selectin, galectin, and siglec antagonists
EP3911307A1 (en) 2019-01-14 2021-11-24 GlycoMimetics, Inc. Selectin or galectin antagonists for treating cytokine release syndrome and crs-induced neurotoxicity
CN114341368A (en) 2019-07-12 2022-04-12 糖模拟物有限公司 Methods of using gene expression as an indicator of E-selectin inhibitor efficacy and clinical outcome for multiple tumor types
JP2022542288A (en) 2019-07-31 2022-09-30 グリコミメティクス, インコーポレイテッド Use of E-Selectin Antagonists to Enhance Survival of Reconstituted Myelodepleted Hosts
WO2021195465A1 (en) * 2020-03-27 2021-09-30 Magnani John L Treatment of acute respiratory distress syndrome and related conditions with antagonists of e-selectin
EP4157285A1 (en) 2020-05-31 2023-04-05 GlycoMimetics, Inc. Compounds and methods for reduction of cancer cell burden and protection of normal hematopoiesis
AU2021292458A1 (en) 2020-06-14 2023-01-19 GlycoMimetics Compositions and methods for overcoming microenvironment-mediated resistance via e-selectin targeting
EP4214218A1 (en) * 2020-09-17 2023-07-26 GlycoMimetics, Inc. E-selectin targeting agents
WO2023014690A1 (en) 2021-08-03 2023-02-09 Glycomimetics, Inc. Compositions and methods for overcoming microenvironment-mediated resistance via e-selectin targeting

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8410666B2 (en) * 2010-05-26 2013-04-02 Kabushiki Kaisha Toshiba Ultrasonic probe and method of manufacturing the same

Family Cites Families (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL7409399A (en) 1973-07-19 1975-01-21 Rhone Poulenc Sa PROCESS FOR THE PREPARATION OF THERMOSTABLE RESINS AND ARTICLES WHOLLY OR PARTIALLY CONSTITUTED FROM THESE RESINS.
US4471057A (en) * 1981-06-30 1984-09-11 The Wistar Institute Detection of colorectal carcinoma
DK17885D0 (en) * 1985-01-14 1985-01-14 Karlsson Karl Anders ANTIVIRAL AGENT
US4876199A (en) * 1985-04-04 1989-10-24 Fred Hutchinson Cancer Research Center Hybridomas producing monoclonal antibodies to mono-, di-, and trifucosylated type 2 chain
US4851511A (en) * 1986-01-30 1989-07-25 Fred Hutchinson Cancer Research Center Monoclonal antibody that specifically binds to disialosyl Lea
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
ES2058070T3 (en) * 1986-05-09 1994-11-01 Pulverer Gerhard USE OF SPECIFIC MONOSACCHARIDES FOR THE PREPARATION OF A MEDICINAL PRODUCT TO PREVENT METASTASIS OF MALIGNANT TUMORS.
US5538724A (en) * 1987-08-11 1996-07-23 The Board Of Trustees For The Leland Stanford Junior Univ. Method of control leukocyte extravasation
US5632991A (en) * 1988-11-14 1997-05-27 Brigham & Women's Hospital Antibodies specific for E-selectin and the uses thereof
US5464778A (en) 1989-03-08 1995-11-07 Board Of Regents Of The University Of Oklahoma Glycoprotein ligand for P-selectin and methods of use thereof
US6033665A (en) * 1989-09-27 2000-03-07 Elan Pharmaceuticals, Inc. Compositions and methods for modulating leukocyte adhesion to brain endothelial cells
US6001988A (en) 1990-06-11 1999-12-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US6280932B1 (en) * 1990-06-11 2001-08-28 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US5576305A (en) 1990-06-15 1996-11-19 Cytel Corporation Intercellular adhesion mediators
US5753631A (en) * 1990-06-15 1998-05-19 Cytel Corporation Intercellular adhesion mediators
US6391857B1 (en) 1990-06-18 2002-05-21 Stanford University Methods and compositions for endothelial binding
US6387884B1 (en) 1990-06-18 2002-05-14 Stanford University Leukocyte homing modulation
US5143712A (en) * 1990-07-30 1992-09-01 Glycomed Incorporated Method of determining a site of inflammation utilizing elam-1 ligands
US5648344A (en) 1990-07-30 1997-07-15 Glycomed Incorporated Methods of treating inflammation using selection binding compounds
US5211937A (en) * 1990-07-30 1993-05-18 Glycomed Incorporated Method of determining a site of inflammation utilizing elam-1 ligands
US5789573A (en) 1990-08-14 1998-08-04 Isis Pharmaceuticals, Inc. Antisense inhibition of ICAM-1, E-selectin, and CMV IE1/IE2
CA2097617A1 (en) * 1990-11-23 1992-05-23 Brian Seed Inhibition of cell adhesion protein-carbohydrate interactions
US5151360A (en) * 1990-12-31 1992-09-29 Biomembrane Institute Effect of n,n,n-trimethylsphingosine on protein kinase-c activity, melanoma cell growth in vitro, metastatic potential in vivo and human platelet aggregation
US6309639B1 (en) 1991-02-05 2001-10-30 The Board Of Regents Of The University Of Oklahoma Method for inhibiting an inflammatory response using antibodies to P-selectin glycoprotein ligand
US6124267A (en) * 1991-02-05 2000-09-26 Southpac Trust Internationals, Inc. O-glycan inhibitors of selectin mediated inflammation derived from PSGL-1
US6121233A (en) * 1991-04-19 2000-09-19 John L. Magnani Methods for the inhibition of cancer metastasis mediated by endothelial adhesion molecules
DE69233136T2 (en) * 1991-05-06 2004-05-13 Genentech, Inc., South San Francisco A SELECTIN LIGAND
US5318890A (en) * 1991-05-06 1994-06-07 The Regents Of The University Of California Assays for inhibitors of leukocyte adhesion
US5352670A (en) * 1991-06-10 1994-10-04 Alberta Research Council Methods for the enzymatic synthesis of alpha-sialylated oligosaccharide glycosides
US5646123A (en) * 1991-06-10 1997-07-08 Alberta Research Council Time dependent administration of oligosaccharide glycosides related to blood group determinants having a type I or type II core structure in reducing inflammation in a sensitized mammal arising form exposure to an antigen
US5580858A (en) * 1991-06-10 1996-12-03 Alberta Research Council Immunosuppressive and tolerogenic modified Lewisx compounds
CA2118695A1 (en) 1991-09-10 1993-03-18 George A. Heavner Peptide inhibitors of inflammation mediated by selectins
US5268364A (en) * 1991-12-12 1993-12-07 The Biomembrane Institute Method for inhibiting selectin-dependent adhesion of leukocytes and platelets by O-glycosylation modification
JPH07501828A (en) * 1991-12-18 1995-02-23 セントコー,インコーポレイテッド Peptide inhibitors of selectin-mediated inflammation
US5591835A (en) * 1992-06-29 1997-01-07 Glycomed Incorporated Substituted lactose derivatives
CA2100412A1 (en) * 1992-07-15 1994-01-16 Yutaka Yamada Glycolipid derivatives
JPH08503694A (en) * 1992-09-08 1996-04-23 セントコー・インコーポレーテッド Peptide-based cell adhesion inhibitor
US5519008A (en) * 1992-09-10 1996-05-21 Glycomed Incorporated Derivatives of triterpenoid acids as inhibitors of cell-adhesion molecules ELAM-1 (E-selectin) and LECAM-1 (L-selectin)
WO1994006442A1 (en) * 1992-09-11 1994-03-31 The Regents Of The University Of California Sulfated ligands for l-selectins and use of chlorates and or sulfatases for the treatment of inflammation
US5695752A (en) * 1992-09-11 1997-12-09 The Regents Of The University Of California Treating inflammation via the administration of specific sulfatase enzymes and/or sulfation inhibitor
US5843707A (en) * 1992-10-23 1998-12-01 Genetics Institute, Inc. Nucleic acid encoding a novel P-selectin ligand protein
US6277975B1 (en) 1992-10-23 2001-08-21 Genetics Institute, Inc. Fusions of P-selectin ligand protein and polynucleotides encoding same
EP0601417A3 (en) * 1992-12-11 1998-07-01 Hoechst Aktiengesellschaft Physiologically compatible and degradable polymer-based carbohydrate receptor blockers, a method for their preparation and their use
WO1994014836A1 (en) 1992-12-18 1994-07-07 Centocor, Inc. Peptide inhibitors of selectin binding
DE69322289T2 (en) 1992-12-29 1999-05-20 Genentech Inc TREATMENT OF INFLAMMABLE INTESTINAL DISEASES WITH INTERFERON GAMMA INHIBITORS
US5412123A (en) * 1993-02-08 1995-05-02 Glycomed Incorporated Anthraquinone and anthracene derivatives as inhibitors of the cell-adhesion molecules of the immune system
EP0687684A4 (en) * 1993-03-04 1998-03-04 Kanto Ishi Pharma Co Ltd Lewis-associated compound, process for producing the same, and anti-inflammatory
US5527890A (en) * 1993-04-16 1996-06-18 Glycomed Incorporated Derivatives of triterpenoid acids and uses thereof
US5811404A (en) 1993-05-14 1998-09-22 Cytel Corporation Sialyl Lex analogues as inhibitors of cellular adhesion
PL176272B1 (en) * 1993-05-14 1999-05-31 Cytel Corp Sialylic le x analoques as inhibitors of cellular adhesion
US5854218A (en) * 1993-05-14 1998-12-29 Cytel Corporation Sialyl Lex analogues as inhibitors of cellular adhesion
US5527785A (en) * 1993-05-14 1996-06-18 The Regents Of The University Of California Selectin receptor modulating compositions
ATE215094T1 (en) 1993-05-17 2002-04-15 Avant Immunotherapeutics Inc COMPLEMENT-RELATED PROTEINS AND CARBOHYDRATES CONTAINING COMPOSITIONS AND METHODS FOR PREPARING AND USING THESE COMPOSITIONS
US5976540A (en) * 1993-05-17 1999-11-02 T Cell Sciences, Inc. Compositions comprising complement related proteins and carbohydrates, and methods for producing and using said compositions
US5646248A (en) * 1993-06-08 1997-07-08 La Jolla Cancer Research Foundation E-selection binding soluble lamp-1 polypeptide
US5658880A (en) * 1993-06-16 1997-08-19 Glycomed Incorporated Sialic acid/fucose based medicaments
US5789385A (en) * 1993-06-16 1998-08-04 Glycomed Incorporated Sialyl Lewisx mimetics containing phenyl backbones
US5837689A (en) * 1993-06-16 1998-11-17 Glycomed Incorporated Sialyl lewis-x mimetics containing naphthyl backbones
US5750508A (en) * 1993-06-16 1998-05-12 Glycomed Incorporated Sialic acid/fucose based medicaments
US5679321A (en) * 1993-06-17 1997-10-21 Glycomed Incorporated Sialic acid/fucose based medicaments
US5559103A (en) * 1993-07-21 1996-09-24 Cytel Corporation Bivalent sialyl X saccharides
US5508387A (en) * 1993-08-04 1996-04-16 Glycomed Incorporated Selectin binding glycopeptides
WO1995005830A1 (en) * 1993-08-20 1995-03-02 The Regents Of The University Of California Polyanion anti-inflammatory agents
US5464815A (en) * 1993-09-08 1995-11-07 Genentech, Inc. Inhibition of heparin-binding
CA2173990A1 (en) 1993-10-12 1995-04-20 Narasinga Rao A library of glyco-peptides useful for identification of cell adhesion inhibitors
US5783693A (en) 1993-11-19 1998-07-21 The Regents Of The University Of California Methods for synthesizing sulfated disaccharide inhibitors of selectins
WO1995014787A1 (en) * 1993-11-22 1995-06-01 Centocor, Inc. Peptide inhibitors of selecting binding
US5663151A (en) * 1994-03-04 1997-09-02 Bristol-Myers Squibb Company Sulfated α-glycolipid derivatives as cell adhesion inhibitors
EP0671409A3 (en) * 1994-03-11 1996-06-12 Hoechst Ag Malonic acid derivatives having anti-adhesive properties.
DE4408248A1 (en) * 1994-03-11 1995-09-14 Hoechst Ag Physiologically acceptable and physiologically degradable carbohydrate mimetics, process for their preparation and their use
HUT77345A (en) * 1994-04-29 1998-03-30 Texas Biotechnology Corporation Mannopyranosyloxy biphenyl derivatives capable of inhibiting the binding of e-selectin,p-selectin or l-selectin to sialyl-lewis x or sialyl-lewis a and pharmaceutical compositions containing them
US5444050A (en) * 1994-04-29 1995-08-22 Texas Biotechnology Corporation Binding of E-selectin or P-selectin to sialyl Lewisx or sialyl-Lewisa
US5486536A (en) * 1994-08-15 1996-01-23 The Regents Of The University Of Michigan Sulfatides as anti-inflammatory compounds
JPH0899989A (en) * 1994-09-30 1996-04-16 Akira Hasegawa New glycolipid derivative and intermediate for its production
DE4436164A1 (en) * 1994-10-10 1996-04-11 Hoechst Ag New conjugates of tetra:carbohydrate and amide-linked peptide or dye etc.
MX9703484A (en) * 1994-11-10 1997-08-30 Pfizer Macrocyclic lactone compounds and their production process.
US5686426A (en) * 1994-11-17 1997-11-11 Bristol-Myers Squibb Company Dicarboxymethylated glycolipid derivatives as cell adhesion inhibitors
US6492332B1 (en) 1995-12-12 2002-12-10 Omeros Corporation Irrigation solution and methods for inhibition of tumor cell adhesion, pain and inflammation
US5639734A (en) * 1994-12-20 1997-06-17 Esko; Jeffrey D. Disaccharide inflammation inhibitors and uses thereof
US20020040008A1 (en) 1995-01-24 2002-04-04 Wagner Denisa D. Method for treating and preventing atherosclerosis
US6506770B1 (en) * 1996-06-06 2003-01-14 Anormed, Inc. Antiviral compounds
US5736533A (en) 1995-06-07 1998-04-07 Neose Technologies, Inc. Bacterial inhibition with an oligosaccharide compound
US5876715A (en) * 1995-08-17 1999-03-02 The Biomembrane Institute Antibodies that bind novel carbohydrate ligands (myelorollins) that cause E-selectin dependent cell rolling, and uses thereof
DE19532902A1 (en) * 1995-09-06 1997-03-13 Hoechst Ag Novel glycomimetics as selectin antagonists and anti-inflammatory drugs made from them
DE19537334A1 (en) * 1995-10-09 1997-04-10 Hoechst Ag New piperidine carboxylic acid and pyrrolidine carboxylic acid derivs.
WO1997015585A1 (en) 1995-10-26 1997-05-01 Kanebo, Ltd. Fucose derivatives, drugs containing the same as active ingredient, and intermediates for producing the same
US5747463A (en) * 1995-11-13 1998-05-05 Bristol-Myers Squibb Company Malonate derivatives of glycolipids as cell adhesion inhibitors
DE19602355A1 (en) 1996-01-24 1997-07-31 Hoechst Ag Multiple fucosylated dicarboxylic acids with anti-adhesive properties
AU1444697A (en) * 1996-01-30 1997-08-22 Novartis Ag Sialyl-lewisa and sialyl-lewisx epitope analogues
EP0886640B1 (en) * 1996-01-30 2007-03-21 GlycoMimetics, Inc. SIALYL-LEWISa AND SIALYL-LEWISx EPITOPE ANALOGUES
US5710023A (en) * 1996-03-01 1998-01-20 Genetics Institute, Inc. IL-13 cytokine receptor chain
EP0902681B1 (en) 1996-03-01 2002-05-22 The Regents of the University of California Inhibition of selectin binding
US5654412A (en) * 1996-05-29 1997-08-05 Glycomed Incorporated Processes for the synthesis of sialyl Lewisx compounds
CA2197058A1 (en) 1996-06-05 1997-12-06 Avery B. Nathens Anti-inflammatory agent
US5919768A (en) * 1996-06-26 1999-07-06 Texas Biotechnology Corporation Di- and trivalent small molecule selectin inhibitors
US5830871A (en) * 1996-10-28 1998-11-03 The Scripps Research Institute Inhibitors of E-, P- and L-selectin binding
GB9618520D0 (en) * 1996-09-05 1996-10-16 Chiroscience Ltd Compounds and their therapeutic use
US6110897A (en) * 1996-10-10 2000-08-29 Glycorex Ab Antiinflammatory cell adhesion inhibitors
WO1998037902A1 (en) 1997-02-28 1998-09-03 The Regents Of The University Of California Inhibition of cell-cell binding by lipid assemblies
US6120751A (en) * 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
SE9701127D0 (en) 1997-03-26 1997-03-26 Karolinska Innovations Ab Antigenic fusion protein carrying GALal, 3GAL epitopes
US5916910A (en) 1997-06-04 1999-06-29 Medinox, Inc. Conjugates of dithiocarbamates with pharmacologically active agents and uses therefore
US6193973B1 (en) 1997-08-22 2001-02-27 B. David Tuttle Dietary supplement for boosting energy and increasing muscular strength
US5948628A (en) 1997-09-05 1999-09-07 The Board Of Regents Of The University Of Oklahoma Methods of screening for compounds which mimic galectin-1
WO1999043353A2 (en) 1998-02-26 1999-09-02 Boehringer Ingelheim Pharmaceuticals, Inc. Combination anti-selectin and immunosuppressant therapy
US6365365B1 (en) 1998-03-20 2002-04-02 The Regents Of The University Of California Method of determining whether an agent modulates glycosyl sulfotransferase-3
US6265192B1 (en) 1998-03-20 2001-07-24 The Regents Of The University Of California Glycosly sulfortransferase-3
US6037333A (en) * 1998-05-07 2000-03-14 Trustees Of Tufts College Microbe-inhibiting compositions
US6593459B1 (en) 1998-06-16 2003-07-15 The Board Of Regents Of The University Of Oklahoma Synthetic glycosulfopeptides and methods of synthesis thereof
US6503885B1 (en) 1998-09-21 2003-01-07 Otsuka Pharmaceutical Co., Ltd. Carboxymethylgalactose derivatives
CA2348781A1 (en) 1998-11-12 2000-05-25 Novolytics Inc. Compositions and methods for producing vascular occlusion
AU6173501A (en) 2000-05-19 2001-12-03 Blood Res Center Methods for diagnosing and treating hemostatic disorders by modulating p-selectin activity
JP4262480B2 (en) * 2000-11-29 2009-05-13 ブラッコ インターナショナル ビー.ヴイ. Sialyl Lewis X analogue with binding ability
US20020132220A1 (en) 2000-12-27 2002-09-19 Berens Kurt L. Use of selectin antagonists in organ preservation solutions
US7087212B2 (en) 2001-08-17 2006-08-08 Mallinckrodt, Inc Multicomponent assemblies having enhanced binding properties for diagnosis and therapy
US7060685B2 (en) 2002-05-16 2006-06-13 Glycomimetics, Inc. Compounds and methods for inhibiting selectin-mediated function
EP1539251A4 (en) * 2002-07-03 2009-02-25 Glycomimetics Inc Compositions and methods for diagnosis and therapy of medical conditions involving angiogenesis
JP2007524658A (en) 2003-11-19 2007-08-30 グリコミメティクス, インコーポレイテッド Glucose mimetic antagonists for both E- and P-selectin
AP2007004095A0 (en) * 2005-01-07 2007-08-31 Univ Emory Cxcr4 antagonists for the treatment of hiv infection
CN103626813B (en) 2005-09-02 2017-05-03 糖模拟物有限公司 Heterobifunctional pan-selectin inhibitors
JP4814039B2 (en) * 2005-10-27 2011-11-09 株式会社リコー Image processing apparatus, image processing program, and recording medium
US20080227799A1 (en) * 2006-07-11 2008-09-18 Liotta Dennis C CXCR4 Antagonists Including Heteroatoms for the Treatment of Medical Disorders
JP5298020B2 (en) 2006-10-12 2013-09-25 グリコミメティクス, インコーポレイテッド Glucose mimic substitution of hexose and N-acetylhexosamine
ES2426784T3 (en) * 2008-06-13 2013-10-25 Glycomimetics, Inc. Treatment of blood cancers using selected glyomimetic compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8410666B2 (en) * 2010-05-26 2013-04-02 Kabushiki Kaisha Toshiba Ultrasonic probe and method of manufacturing the same

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9796745B2 (en) 2011-12-22 2017-10-24 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
US10526361B2 (en) 2011-12-22 2020-01-07 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
US10766916B2 (en) 2011-12-22 2020-09-08 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
US9109002B2 (en) 2011-12-22 2015-08-18 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
US11332491B2 (en) 2011-12-22 2022-05-17 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
US9867841B2 (en) 2012-12-07 2018-01-16 Glycomimetics, Inc. Compounds, compositions and methods using E-selectin antagonists for mobilization of hematopoietic cells
WO2016089872A1 (en) * 2014-12-03 2016-06-09 Glycomimetics, Inc. Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors
US20170305951A1 (en) * 2014-12-03 2017-10-26 Glycomimetics, Inc. Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors
US10519181B2 (en) * 2014-12-03 2019-12-31 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors
US11291678B2 (en) 2016-03-02 2022-04-05 Glycomimetics, Inc Methods for the treatment and/or prevention of cardiovascular disease by inhibition of E-selectin
US11433086B2 (en) 2016-08-08 2022-09-06 Glycomimetics, Inc. Combination of T-cell checkpoint inhibitors with inhibitors of e-selectin or CXCR4, or with heterobifunctional inhibitors of both E-selectin and CXCR4
US11072625B2 (en) 2016-10-07 2021-07-27 Glycomimetics, Inc. Highly potent multimeric e-selectin antagonists
US11780873B2 (en) 2016-10-07 2023-10-10 Glycomimetics, Inc. Highly potent multimeric e-selectin antagonists
US11197877B2 (en) 2017-03-15 2021-12-14 Glycomimetics. Inc. Galactopyranosyl-cyclohexyl derivauves as E-selectin antagonists
US11878026B2 (en) 2017-03-15 2024-01-23 Glycomimetics, Inc. Galactopyranosyl-cyclohexyl derivatives as e-selectin antagonists
US11712446B2 (en) 2017-11-30 2023-08-01 Glycomimetics, Inc. Methods of mobilizing marrow infiltrating lymphocytes and uses thereof
US11548908B2 (en) 2017-12-29 2023-01-10 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
US11707474B2 (en) 2018-03-05 2023-07-25 Glycomimetics, Inc. Methods for treating acute myeloid leukemia and related conditions
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3

Also Published As

Publication number Publication date
AU2010241807A1 (en) 2011-11-10
JP5726171B2 (en) 2015-05-27
US8410066B2 (en) 2013-04-02
US20100279965A1 (en) 2010-11-04
CN102421441B (en) 2015-04-01
CA2760292A1 (en) 2010-11-04
WO2010126888A1 (en) 2010-11-04
CN102421441A (en) 2012-04-18
JP2012525393A (en) 2012-10-22
EP2424544A1 (en) 2012-03-07
AU2010241807B2 (en) 2014-08-14

Similar Documents

Publication Publication Date Title
US8410066B2 (en) Heterobifunctional inhibitors of E-selectins and CXCR4 chemokine receptors
US11548908B2 (en) Heterobifunctional inhibitors of E-selectin and galectin-3
US8518896B2 (en) Treatment of cancers of the blood using selected glycomimetic compounds
US9534009B2 (en) Pan-selectin inhibitor with enhanced pharmacokinetic activity
WO2012061662A9 (en) Glycomimetic-peptidomimetic inhibitors of e-selectins and cxcr4 chemokine receptors
US8039442B2 (en) Compounds and methods for treatment of sickle cell disease or complications associated therewith
US20100104660A1 (en) Composition and method for treating tumor
KR20100102092A (en) Azacytidine analogues and uses thereof
US20230079833A1 (en) Galactose-linked multimeric glycomimetic inhibitors of e-selectins, galectin-3, and/or cxcr4 chemokine receptors
CN101921164A (en) Synthesis of (1)-beta-elemene, (-)-beta-elemenal, (-)-beta-elemenol, (-)-beta-elemene fluoride and their analogues, intermediates, and composition and uses thereof
JP4603684B2 (en) 13-deoxyanthracycline derivative and method for producing the same
US11517580B2 (en) Fucosyltransferase specific inhibition using fucose mimetics
CN113801178A (en) Dual targeting compounds for inhibition of E-selectin and CXCR4 chemokine receptors
US20230132763A1 (en) Fucosyltransferase specific inhibition using fucose mimetics
CN117959459A (en) Tumor microenvironment activated drug conjugate and antibody drug conjugate
KR20200039622A (en) Synthesis of single protected bifunctional prodrug and antibody drug conjugate based thereon and method of preparing antibody drug conjugate
MXPA01009025A (en) L-arabino-disaccharides of anthracyclines, processes for their preparation, and pharmaceutical compositions containing them.

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION