US20070060591A1 - Chemokine receptor binding heterocyclic compounds - Google Patents

Chemokine receptor binding heterocyclic compounds Download PDF

Info

Publication number
US20070060591A1
US20070060591A1 US11/599,066 US59906606A US2007060591A1 US 20070060591 A1 US20070060591 A1 US 20070060591A1 US 59906606 A US59906606 A US 59906606A US 2007060591 A1 US2007060591 A1 US 2007060591A1
Authority
US
United States
Prior art keywords
mmol
methylene
nmr
phenylenebis
pyridine
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
US11/599,066
Inventor
Gary Bridger
Eva Boehringer
Dominique Schols
Renato Skerlj
David Bogucki
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.)
Genzyme Corp
Original Assignee
Bridger Gary J
Boehringer Eva M
Dominique Schols
Skerlj Renato T
Bogucki David E
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 Bridger Gary J, Boehringer Eva M, Dominique Schols, Skerlj Renato T, Bogucki David E filed Critical Bridger Gary J
Priority to US11/599,066 priority Critical patent/US20070060591A1/en
Publication of US20070060591A1 publication Critical patent/US20070060591A1/en
Assigned to GENZYME CORPORATION reassignment GENZYME CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANORMED CORPORATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention generally relates to novel compounds, pharmaceutical compositions and their use. This invention more specifically relates to novel heterocyclic compounds that bind to chemokine receptors, including CXCR4 and CCR5, and demonstrate protective effects against infection of target cells by a human immunodeficiency virus (HIV).
  • chemokine receptors including CXCR4 and CCR5
  • chemokines Approximately 40 human chemokines have been described, that function, at least in part, by modulating a complex and overlapping set of biological activities important for the movement of lymphoid cells and extravasation and tissue infiltration of leukocytes in response to inciting agents (See, for example: Ponath, P., Exp. Opin. Invest. Drugs 7:1-18 (1998); Baggiolini, M., Nature 392:565-568 (1998); Locati, et al., Annu. Rev. Med. 50:425-40 (1999)).
  • chemotactic cytokines, or chemokines constitute a family of proteins, approximately 8-10 kDa in size.
  • Chemokines appear to share a common structural motif, that consists of 4 conserved cysteines involved in maintaining tertiary structure. There are two major subfamilies of chemokines: the “CC” or ⁇ -chemokines and the “CXC” or ⁇ -chemokines. The receptors of these chemokines are classified based upon the chemokine that constitutes the receptor's natural ligand. Receptors of the ⁇ -chemokines are designated “CCR” while those of the ⁇ -chemokines are designated “CXCR.”
  • Chemokines are considered to be principal mediators in the initiation and maintenance of inflammation (see Chemokines in Disease published by Humana Press (1999), Edited by C. Herbert; Murdoch, et al., Blood 95:3032-3043 (2000)). More specifically, chemokines have been found to play an important role in the regulation of endothelial cell function, including proliferation, migration and differentiation during angiogenesis and re-endothelialization after injury (Gupta, et al., J. Biol. Chem. 7:4282-4287 (1998); Volin, et al., Biochem. Biophys Res. Commun. 242:46-53 (1998)). Two specific chemokines have been implicated in the etiology of infection by human immunodeficiency virus (HIV).
  • HAV human immunodeficiency virus
  • HIV initially binds via its gp120 envelope protein to the CD4 receptor of the target cell.
  • CCR5 Wangt, et al., Science 280:1884-1888 (1998); Rizzuto, et al., Science 280:1949-1953 (1998); Berger, et al., Annu. Rev. Immunol. 17:657-700 (1999)
  • virus-cell fusion results which is mediated by members of the chemokine receptor family, with different members serving as fusion cofactors for macrophage-tropic (M-tropic) and T cell line-tropic (T-tropic) isolates of HIV-1
  • M-tropic macrophage-tropic
  • T-tropic T cell line-tropic isolates of HIV-1
  • the M-tropic viral phenotype correlates with the virus's ability to enter the cell following binding of the CCR5 receptor, while the T-tropic viral phenotype correlates with viral entry into the cell following binding and membrane fusion with the CXCR4 receptor.
  • Clinical observations suggest that patients who possess genetic mutations in CCR5 appear resistant, or less susceptible to HIV infection (Liu, et al., Cell 86:367-377 (1996); Samson, et al., Nature 382:722-725 (1996); Michael, et al., Nature Med. 3:338-340 (1997); Michael, et al., J. Virol.
  • CCR5 and CXCR4 appear to be the only physiologically relevant coreceptors used by a wide variety of primary clinical HIV-1 strains (Zhang, et al., J. Virol.
  • CXCR4 or SDF-1 knock-out mice exhibit cerebellar, cardiac and gastrointestinal tract abnormalities and die in utero (Zou, et al., Nature 393:591-594 (1998); Tachibana, et al., Nature 393:591-594 (1998); Nagasawa, et al., Nature 382:635-638 (1996)).
  • CXCR4-deficient mice also display hematopoietic defects (Nagasawa, et al., Nature 382:635-638 (1996)); the migration of CXCR4 expressing leukocytes and hematopoietic progenitors to SDF-1 appears to be important for maintaining B-cell lineage and localization of CD34 + progenitor cells in bone marrow (Bleul, et al., J. Exp. Med. 187:753-762 (1998); Viardot, et al., Ann. Hematol. 77:195-197 (1998); Auiti, et al., J. Exp. Med.
  • the signal provided by SDF-1 on binding to CXCR4 may also play an important role in tumor cell proliferation and regulation of angiogenesis associated with tumor growth (See “Chemokines and Cancer” published by Humana Press (1999); Edited by B. J. Rollins; Arenburg, et al., J. Leukocyte Biol. 62:554-562 (1997); Moore, et al., J. Invest. Med. 46:113-120 (1998); Moore, et al., Trends cardiovasc. Med. 8:51-58 (1998); Seghal, et al., J. Surg. Oncol.
  • bicyclam dose-dependently inhibits binding of 125I-labeled SDF-1 to CXCR4 and the signal transduction (indicated by an increase in intracellular calcium) in response to SDF-1.
  • the bicyclam also functioned as an antagonist to the signal transduction resulting from the binding of stromal derived factor or SDF-1 ⁇ , the natural chemokine to CXCR4.
  • Bicyclams also inhibited HIV gp120 (envelope)-induced apoptosis in non-HIV infected cells (Blanco, et al., Antimicrobial Agents and Chemother. 44:51-56 (2000)).
  • This competitive binding protects these target cells, which utilize the CXCR4 receptor for entry from infection by HIV.
  • the compounds antagonize the binding, signaling and chemotactic effects of the natural ligand for CXCR4, i.e., the chemokine stromal cell-derived factor 1 ⁇ (SDF-1), and also have protective effects against HIV infection of target cells by binding in vitro to the CCR5 receptor.
  • SDF-1 chemokine stromal cell-derived factor 1 ⁇
  • the present invention describes novel compounds that exhibit protective effects against HIV infection of target cells by binding to the chemokine receptors CXCR4 or CCR5, in a similar manner to the previously disclosed macrocyclic compounds, and that are additionally useful in other indications addressed by the compounds as described above.
  • the present invention provides novel compounds that bind chemokine receptors and interfere with the binding of the natural ligand thereto, and are useful as agents demonstrating protective effects on target cells from HIV infection.
  • the invention compounds act as antagonists or agonists of chemokine receptors and exhibit biological activities related to the ability of these compounds to inhibit the binding of chemokines to their receptors.
  • the present invention provides a compound of Formula 1 V—CR 2 —Ar 1 —CR 2 NR—(CR 2 ) x —Ar 2 (1)
  • V is a substituted heterocycle of 9-24 members containing 2-4 optionally substituted amine nitrogen atoms spaced from each other by 2 or more optionally substituted carbon atoms, and which heterocycle may optionally comprise a fused aromatic or heteroaromatic ring, and wherein
  • said heterocycle contains at least one O or S, said O or S spaced from any adjacent heteroatom by at least 2 carbon atoms, and wherein said S is optionally oxidized or
  • each R is independently H or a straight chain, branched or cyclic alkyl containing 1-6C;
  • x is 0-4;
  • Ar 1 is an unsubstituted or substituted aromatic or heteroaromatic moiety
  • Ar 2 is an unsubstituted or substituted aromatic or heterocyclic group.
  • aspects of the invention are directed to the pharmaceutical compositions comprising a therapeutically effective amount of the compound of Formula 1 and to methods of treating a condition of the human body or the bodies of other mammals comprising the administration of a pharmaceutical or veterinary composition which contains a therapeutically effective amount of the compound of Formula 1.
  • the invention is directed to a method for blocking or interfering with the binding of a chemokine receptor with its natural ligand, by contacting the chemokine receptor with an effective amount of the compound of Formula 1.
  • the invention includes the use of a compound of Formula 1 in the manufacture of a medicam ent for the treatment of a disease in which blocking or interfering with binding of a chemokine receptor with its natural ligand is advantageous and for protecting target cells possessing chemokine receptors, the binding to which by a pathogenic agent results in disease or pathology.
  • the invention is also directed to methods of treatment as outlined above.
  • FIG. 1 shows the structure of N-[4-(11-Fluoro-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8897).
  • FIG. 2 shows the structure of N-[4-(11,11-difluoro-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8880).
  • FIG. 3 shows the structure of N-[4-(1,4,7-triazacyclotetradecan-2-one)-yl))-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8748).
  • FIG. 4 shows the structure of N-[12-(5-oxa-1,9-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8922).
  • FIG. 5 shows the structure of Preparation of N-[4-(11-oxa-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8779).
  • FIG. 6 shows the structure of N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8834).
  • FIG. 7 shows the structure of N-[4-(11-sulfoxo-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD9424).
  • FIG. 8 shows the structure of N-[4-(11-sulfono-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD9408).
  • FIG. 9 shows the structure of N-[4-(3-carboxo-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD-Exp40).
  • FIG. 10 shows the structure of 1,1′-[1,4-phenylenebis(methylene))]bis-1,4,8,11-tetraazacyclotetradecane (AMD3100).
  • the present invention is directed to compounds of Formula 1 which can act as agents that modulate chemokine receptor activity.
  • chemokine receptors include but are not limited to CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8 and CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5.
  • the compounds of Formula 1 that demonstrate protective effects on target cells from HIV infection so as to bind specifically to the chemokine receptor, affect the binding of a natural ligand or chemokine to a receptor such as CXCR4 and/or CCR5. They are also useful as agents which affect chemokine receptors, such as CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8 and CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5 where such chemokine receptors have been correlated as being important mediators of many human inflammatory as well as immunoregulatory diseases. Thus, the compounds of Formula 1, which modulate the activity of such chemokine receptors are useful for the treatment or prevention of such diseases.
  • modulators encompasses antagonist, agonist, partial antagonist, and or partial agonist, inhibitors, and activators.
  • compounds of Formula 1 demonstrate protective effects against HIV infection by inhibiting binding of HIV to a chemokine receptor, such as CXCR4 and/or CCR5 of a target cell.
  • the compounds of Formula 1 that inhibit chemokine receptors may be used for the treatment of diseases associated with hematopoiesis, including but not limited to, controlling the side-effects of chemotherapy, enhancing the success of bone marrow transplantation, enhancing wound healing and burn treatment, as well as combating bacterial infections and leukemia.
  • These compounds of Formula 1 are thus also useful for the treatment of diseases that are associated with inflammation, including but are not limited to, inflammatory or allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type hypersensitivity, asthma, interstitial lung disease (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or dermatomyositis); systemic anaphylaxis or hypersensitivity responses, drug allergies, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, psoriatic arthritis, systemic lupus erythematosus, myastenia gravis, juvenile onset diabetes; glomerulonephritis, autoimmune throidit
  • the compounds of the invention that activate or promote chemokine receptor function may be used for the treatment of diseases that are associated with immunosuppression such as individuals undergoing chemotherapy, radiation therapy, enhanced wound healing and burn treatment, therapy for autoimmune disease or other drug therapy (e.g., corticosteroid therapy) or combination of conventional drugs used in the treatment of autoimmune diseases and graft/transplantation rejection, which causes immunosuppression; immunosuppression due to congenital deficiency in receptor function or other causes; and infectious diseases, such as parasitic diseases, including but not limited to helminth infections, such as nematodes (round worms); Trichuriasis, Enterobiasis, Ascariasis, Hookworm, Strongyloidiasis, Trichinosis , filariasis; trematodes; visceral worms, visceral larva migtrans (e.g., Toxocara), eosinophilic gastroenteritis (e.g., Anisaki spp., Phoc
  • Compounds of Formula 1 may be used in combination with any other pharmaceutical composition where such combined therapy may be useful to modulate chemokine receptor activity and thereby prevent and treat inflammatory and immunoregulatory diseases, including, for example, in combinations with one or more agents useful in the prevention or treatment of HIV.
  • agents useful in the prevention or treatment of HIV include:
  • nucleotide reverse transcriptase inhibitor such as zidovudine, didanosine, lamivudine, zalcitabine, abacavir, stavudine, adefovir, adefovir dipivoxil, fozivudine todoxil, etc.;
  • non-nucleotide reverse transcriptase inhibitor including an agent having anti-oxidation activity such as immunocal, oltipraz, etc.
  • non-nucleotide reverse transcriptase inhibitor such as nevirapine, delavirdine, efavirenz, loviride, immunocal, oltipraz, etc.
  • protease inhibitors such as saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, palinavir, lasinavir, KaletraTM (lopinavir/ritonavir), etc.
  • Such combinations the compounds of the present invention and other HIV agents may be administered separately or in conjunction; the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • the compounds of Formula 1 may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • parenteral e.g., intramuscular, intraperitoneal, intravenous, intracisternal injection or infusion, subcutaneous injection, or implant
  • inhalation spray nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • the compounds of Formula 1 may be used to treat animals, including mice, rats, horses, cattle, sheep, dogs, cats, and monkey, and are also effective for use in humans.
  • the compounds of the invention may be supplied as “pro-drugs,” or, protected forms of the compounds of Formula 1, which release the compound after administration to a patient.
  • the compound may carry protective groups which are split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing active compound or is oxidized or reduced in body fluids to release the compound.
  • a discussion of pro-drugs may be found in Smith and Williams' Introduction to the Principles of Drug Design , H. J. Smith, Wright, Second Edition, London 1988.
  • Acid addition salts which are pharmaceutically acceptable, such as salt with inorganic base, a salt with organic base, a salt with inorganic acid, a salt with organic acid, a salt with basic or acidic amino acid, etc. and non-toxic metal complexes are also encompassed in the present invention.
  • a salt with an inorganic base include a salt with alkali metal (e.g. sodium, potassium, etc.), alkaline earth metal (e.g. calcium, magnesium, etc.), aluminum, ammonium, etc.
  • Examples of the salt with an organic base include a salt with trimethylamine, triethylamine, pyridine, picoline, ethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, N,N′-dibenzylethylenediamine etc.
  • Examples of the salt with an inorganic acid include a salt with hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, etc.
  • Examples of the salt with an organic acid include a salt with formic acid, oxalic acid, acetic acid, tartaric acid, methanesulfonic acid, benzenesulfonic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, etc.
  • Examples of salts with basic amino acids include a salt with arginine, lysine, ornithine, etc.
  • Examples of salts with the acidic amino acid include a salt with aspartic acid, glutamic acid, etc.
  • Non-toxic in the present tense has to be considered with reference to the prognosis for the infected subject without treatment. Copper and zinc complexes are preferred although other metals such as nickel, cobalt or rhubidium, may be used.
  • the compounds of Formula 1 may form hydrates or solvates. Some compounds of Formula 1 exist as regioisomers, configurational isomers, conformers, diasteroisomeric forms and mixtures of diasteroisomeric forms thereof; it is possible to isolate individual isomers using known separation and purification methods, if desired.
  • the invention includes mixtures of these stereoisomers as well as isolated forms. The mixtures may contain the stereoisomers in any ratio.
  • Compounds of the invention also include racemates, which can be separated into the (S)-compounds and (R)-compounds by optical resolution; individual optical isomers and mixtures thereof are included in the scope of the present invention.
  • the compounds of Formula 1 may be administered alone or as an admixture with a pharmaceutically acceptable carrier (e.g. solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.) may be orally or non-orally administered.
  • a pharmaceutically acceptable carrier e.g. solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.
  • non-oral formulations include injections, drops, suppositories and pessaryies.
  • V contains 2-4 N, preferably 3-4 N if there is no additional heteroatom.
  • Preferable ring sizes for V are 9-18 members, more preferably 12-16 members.
  • V may also include a fused aromatic or heteroaromatic ring, preferably 1, 2 or 1, 3 or 1,4 phenylene or 2, 6 or 2, 5 or 2, 4 or 2,3 pyridinylene.
  • the fused ring may also be, for example, 2, 5 or 2,6 pyrimidinylene or 2, 4 or 2,3 pyrrolylene.
  • the required electron withdrawing substituents present at at least one C in ring V may be halogen, nitro, cyano, carboxylic acid, a carboxylic ester with an alcohol of 1-6C or amide formed from an amine of 0-12C, a sulfonic or sulfinic acid or a sulfonic or sulfinic ester or amide, CF 3 , and the like.
  • a preferred electron withdrawing substituent is ⁇ O, as well as halo.
  • halogen examples include fluorine, chlorine, bromine, iodine, etc., with fluorine and chlorine preferred.
  • Ar 2 is an optionally substituted heterocyclic group or aromatic group.
  • aromatic groups include benzene and naphthalene, or dihydronaphthalene and tetrahydronaphthalene.
  • heterocyclic groups include 5 to 6-membered saturated, partially saturated, or aromatic heterocyclic rings containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur.
  • the heterocycles may be pyridine, quinoline, isoquinoline, imidazole, benzimidazole, azabenzimidazole, benzotriazole, furan, benzofuran, thiazole, benzothiazole, oxazole, benzoxazole, pyrrole, indole, indoline, indazole, pyrrolidine, pyrrolidone, pyrroline, piperidine, piperazine, tetrahydroquinoline, tetrahydroisoquinoline, pyrazole, thiophene, isoxazole, isothiazole, triazole, tetrazole, oxadiazole, thiadiazole, morpholine, thiamorpholine, pyrazolidine, imidazolidine, imidazoline, tetrahydropyran, dihydropyran, benzopyran, dioxane, dithiane, tetrahydro
  • the optional substituents on Ar 2 include alkyl(1-6C), alkenyl(1-6C), alkynyl(1-6C), halo, nitro, cyano, carboxylic acid, carboxylic ester formed from an alcohol with 1-6C or amide formed from an amine of 0-12C, a sulfonic or sulfinic acid or ester or amide, OR, SR, NR 2 , OCR, OOCR, NRCOR, all wherein R is hydrogen or straight or branched chain alkyl(1-6C), an optionally substituted aromatic or heterocyclic group, CF 3 , and the like.
  • Preferred substituents include alkyl, OR, NR 2 , and halo.
  • Preferred embodiments of Ar 2 include phenyl, pyridinyl, pyrimidinyl and imidazolyl.
  • Ar 1 is a 5-6 membered aromatic system which is bivalent benzene, pyridine, thiophene, pyrimidine, and the like.
  • Ar 1 may optionally be substituted by alkyl, alkenyl, halo, nitro, cyano, CF 3 , COOR, CONR 2 , OCR, OOCR, NRCOR, OR, NR 2 , SR, (where R is H or alkyl 1-6C) sulfonic or sulfinic acids, esters or amides and the like.
  • Preferred embodiments of Ar 1 are phenylene, especially 1,3 and 1,4 phenylene and pyridinylene, preferably 2,6 pyridinylene, and 3,5 pyridinylene.
  • Preferable substituents are alkyl, OR, NR 2 and halo.
  • each R group be hydrogen or alkyl of 1-2C, preferably hydrogen.
  • the R group coupled to a nitrogen is hydrogen or alkyl 1-6C, preferably straight chain alkyl 1-3C, more preferably H or methyl.
  • 1, 2, 3, 4, or 5 of the R groups are methyl or ethyl and the remaining R groups are hydrogen.
  • the compound of Formula 1 is of the formula V—CH 2 —Ar 1 —CH 2 NR—CH 2 —Ar 2
  • V is (a) substituted with halo or ⁇ O or (b) contains O or S or (c) both (a) and (b), and wherein Ar 1 is unsubstituted 1, 3 or 1,4-phenylene, R is H, methyl or ethyl and Ar 2 is unsubstituted phenyl or pyridinyl.
  • Preferred embodiments of x are 0-2 and 1-2.
  • an appropriate dosage level will generally be about 0.01 to 500 mg per kg body weight per day which can be administered in singe or multiple doses. Preferably, the dosage level will be about 0.1 to about 250 mg/kg per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound used, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the patient undergoing therapy.
  • the active compounds may be administered in the form of a pharmaceutical composition formulated according to well known principles and incorporating the compound, preferably in unit dose form, in combination with a pharmaceutically acceptable diluent, carrier or excipient.
  • a pharmaceutically acceptable diluent, carrier or excipient may be in the form of solutions or suspensions for injection, or irrigation or be in capsule, tablet, dragee, or other solid composition or as a solution or suspension for oral administration or formulated into pessaries or suppositories or sustained release forms of any of the above for implantation.
  • Suitable diluents, carriers, excipients and other components are well known. It may be desirable also to formulate a composition for topical administration such as an ointment or cream.
  • compositions according to the invention may be formulated in unit dosages determined in accordance with conventional pharmacological methods, suitably to provide active compounds in the dosage range in humans or animals of from 0.01 to 100 mg/kg body weight per day, in a single dose or in a number of smaller doses. Preferred dosage ranges are 0.01 to 30 mg/kg body weight per day intravenous (iv) or intraperitoneal (ip). Other active compounds may be used in the compositions or such active compounds or supplemental therapy may be included in a course of treatment.
  • the pharmaceutical compositions are useful for treatment of a patient comprising an effective therapeutic amount of the novel compound, where said compound effectively binds to a chemokine receptor.
  • the present invention further contemplates the use of these compositions in the manufacture of a medicament for the treatment of HIV- or FIV-infected patients and/or the treatment of a disease by the regulation of endothelial cell function and/or the treatment of a disease relating to vascularization of the gastrointestinal tract; haematopoiesis; or cerebellar development.
  • the pharmaceutical composition is administered to said patient as a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is administered to said patient as a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier.
  • the present invention further contemplates methods of treating a patient with a disease relating to vascularization of the gastrointestinal tract; haematopoiesis; or cerebellar development, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier.
  • the present invention further contemplates a method of treating a patient with a disease relating to basal leukocyte trafficking or the extravasation and tissue infiltration of leukocytes in response to inciting antigens, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier.
  • the present method also contemplates treating a patient, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier, wherein said compound effectively binds to a chemokine receptor.
  • the present invention further contemplates pharmaceutical compositions and methods of use for the treatment of humans or animals for: renal allograft rejection; inflammatory disease; cancer; central nervous system developmental disease; HIV; vasculature development disease; haematopoiesis and other chemokine mediated diseases or disorders.
  • the invention further provides for the treatment of diseases, which include, but are not limited to: arthritis; asthma; multiple sclerosis; dementia from HIV or FIV infection, Parkinson's disease, Alzheimer's disease and inflammatory diseases.
  • the pharmaceutical compositions and methods of use of the present invention further provide for the treatment of cancers, that include, but are not limited to those associated with: solid tumors; lymphoma; metastatic tumors; glioblastoma tumors; leukemia; and other carcinomas tumors.
  • the pharmaceutical compositions of the present invention are useful for the treatment of cancers that include, but are not limited to: non-small cell lung cancer; lung cancer; breast cancer; prostate cancer; and cancer of other organs.
  • diseases or disorders that are contemplated to be treated with the pharmaceutical compositions of the present invention, include, but are not limited to: disorders treated by inhibiting or promoting angiogenesis or by inducing stasis of angiogenesis; developmental disorders mediated by chemokines.
  • the present invention further provides methods for the prevention of a disease or disorder in a patient by administering a therapeutically effective dosage of the pharmaceutical compositions of the present invention to a patient over a period of time sufficient to effectively prevent the disease or disorder.
  • Acetic acid (15 ml), saturated with anhydrous hydrogen bromide (gas) was added to [11-fluoro-1,7-bis(2-nitro-benzenesulfonyl)-1,4,7-triaza-cyclotetradec-4-yl]-phosphonic acid diethyl ester (560 mg, 0.77 mmol) contained in round-bottomed flask closed by glass stopper. The resulting mixture was allowed to stir for 18 hours at room temperature and then diethyl ether (50 ml) was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the diethyl ether solution was decanted off.
  • Acetic acid (10 ml), saturated with anhydrous hydrogen bromide (gas) was added to [11,11-difluoro-1,7-bis(2-nitro-benzenesulfonyl)-1,4,7-triaza-cyclotetradec-4-yl]-phosphonic acid diethyl ester (311 mg, 0.41 mmol) contained in round-bottomed flask closed by glass stopper.
  • the resulting mixture was allowed to stir for 18 hours at room temperature and then diethyl ether (50 ml) was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the diethyl ether solution was decanted off.
  • reaction mixture was diluted with 50 ml of CH 2 Cl 2 , filtered through celite, and concentrated in vacuo. Purification of the residue by flash chromatography on silica gel eluting with 5% MeOH, 95% CH 2 Cl 2 gave 807 mg (89%) of the desired amide as a yellow oil.
  • Dimethyl malonate (10.00 g, 75.7 nmol) was added to a suspension of NaH (60% in oil, 3.33 g, 83.3 mmol) in THF (100 mL) over 20 minutes at room temperature. The mixture was heated at 80° C. for 15 minutes, then a solution of ⁇ -bromotolunitrile (14.84 g, 75.69 mmol) in THF (100 mL) was added. Heating was continued at 80° C. for 1 hour, then at 50° C. for 64 hours. Water (30 mL) was added, and the aqueous phase was extracted with EtOAc (3 ⁇ 20 mL). The combined organic phases were dried (MgSO 4 ) and concentrated.
  • the desired macrocycle was prepared using standard macrocyclization conditions; Bridger et al. J. Med. Chem. 1995, 38, 366-378): To a stirred solution of the intermediate from above (1.8 g, 3.59 mmol) in DMF (180 ml) containing anhydrous Cs 2 CO 3 (3.6 g, 11.05 mmol) heated to 80° C. was added dropwise, a solution of N-(diethoxyphosphoryl)-O,O′-bis(2-methylsulfonyl)diethanolamine (Bridger, et al. J. Med. Chem . (1995) 38, 366-378) (1.9 g, 4.78 mmol) in DMF (20 ml).
  • AMD8779 N-[4-(11-oxa-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt)
  • AMD8834 N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt)
  • the sulfone (73 mg, 0.96 mmol) was dissolved in a minimum amount of acetic acid and treated with a saturated solution of HBr in acetic acid (5 mL). The mixture was stirred under nitrogen for 63 h at room temperature and diethyl ether (100 mL) was added. A precipitate formed and was allowed to settle to the bottom of the flask and the supernatant solution was decanted. The white solid was washed with diethyl ether (5 ⁇ 100 mL) and the remaining traces of solvent were removed by blowing nitrogen though the flask followed by drying under vacuum overnight at 55° C. to give AMD9408 (70 mg, 84%) as a white solid.
  • Loading dye Fluo-3, AM (Molecular Probes F-1241) is dissolved in anhydrous DMSO and stored frozen in aliquots. To increase the solubility of the dye in the loading medium, 10% (w/v) pluronic acid (Molecular Probes F-127) is added to the Fluo-3 stock solution immediately before use.
  • HBSS 10 ⁇ [(w/o phenol red and sodium bicarbonate (Gibco 14 065-049)]; Hepes buffer 1M (Gibco 15 630-056), BSA (Sigma A3675).
  • the flux buffer is vacuum-filtered and stored refrigerated for a maximum of 5 days. Before use in the experiment, the buffer is warmed at 37° C. in a waterbath.
  • test compounds are diluted in flux buffer and added to 4 wells of a black microplate (4 parallel measurements per compound).
  • the following control wells are used: 100% response control (no inhibition), flux buffer is added; 100% inhibition control: chemokine is added at 5-times the concentration required to induce a Ca flux.
  • the chemokines are diluted in flux buffer to concentrations that are 4-fold higher than the desired concentrations required for stimulation of the cells (i.e. 2.5 nM for SDF-1 ⁇ ).
  • the chemokines were added to untreated 96-well Sero well compound plates (International Medical, Sterilin code 611F96).
  • flux buffer is added instead of chemokine.
  • 20 ⁇ M digitonin final concentration is also included.
  • the agonist plate is incubated in the FLIPR (37° C.) for 15-30 min.
  • SUP-T1 cells are centrifuged at room temperature (RT) and re-suspended in loading medium (RPMI-1640 containing 2% FBS and 4 ⁇ M Fluo-3, AM). The cells are incubate at room temperature for 45 min. then washed twice in flux buffer then incubated in flux buffer at room temperature for 10 min. The cells are centrifuged and re-suspended in flux buffer at a density of 3 ⁇ 10 6 cells per mL. A 100 ⁇ L aliquot of the cell suspension (3 ⁇ 10 5 cells) is added to each well of a black microplate (Costar 3603), which already contains 50 ⁇ L of a solution of the test compound (at concentrations that are 3-fold higher than the desired final compound concentrations).
  • loading medium RPMI-1640 containing 2% FBS and 4 ⁇ M Fluo-3, AM
  • microplate is then gently centrifuged at room temperature. Homogeneous spreading of the cells on the bottom of the microplate wells is then confirmed with a microscope and the microplate was incubated in the FLIPR (37° C.) for 10 min. prior to testing.
  • the FLIPR settings (camera exposure time and laser power) are adjusted to obtain initial fluorescence values between 8,000 and 10,000 units.
  • the agonist chemokine
  • 50 ⁇ L is added by automatic pipettor with black pipette tips. Fluorescence is measured simultaneously in all wells of the microplate every 2 seconds (first 2 min) and thereafter every 6 seconds (additional 2 min). The average ⁇ -flux measured in each set of 4 identical wells (one test compound) was calculated by the FLIPR software.
  • the compounds of the current invention were tested for inhibition of SDF-1 ⁇ induced Ca flux in SUP-T1 cells using the method described above.
  • the compounds described in Examples 1-9 inhibited SDF-1 ⁇ induced Ca flux greater than 50% at 25 ⁇ g/mL.
  • HIV-1 NL4.3 (or III B ) replication assays were performed as previously described (Bridger, et al, J. Med. Chem. 42:3971-3981 (1999); De Clercq, et al., Proc. Natl. Acad. Sci. 89:5286-5290 (1992); De Clercq, et al., Antimicrob. Agents Chemother. 38:668-674 (1994); Bridger, et al. J. Med. Chem. 38:366-378 (1995)). Anti-HIV activity and cytotoxicity measurements were carried out in parallel. They were based on the viability of MT-4 cells that had been infected with HIV in the presence of various concentrations of the test compounds.
  • the number of viable cells was quantified by a tetrazolium-based colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) procedure in 96-well microtrays.
  • viral input viral multiplicity of infection, MOI
  • MOI viral multiplicity of infection
  • CCID 50 50% cell culture infective dose
  • the compounds of the current invention were tested as described above.
  • the compounds described in Examples 1-9 inhibited HIV-1 replication with EC 50 's in the range 0.003-33.3 ug/mL.
  • a compound related to those of the invention demonstrated inhibition of collagen-induced arthritis (CIA) in a mutant mouse model.
  • the control group consisted of ten mice that were injected with collagen as discussed below.
  • the treatment group consisted of eight mice which were also injected with collagen and were further treated by administering 1,1′-[1,4-phenylenebis(methylene))]bis-1,4,8,11-tetraazacyclotetradecane (AMD 3100; see FIG. 10 ) intravenously using osmotic pumps (200 ⁇ l, Alza, 0.5 ⁇ l/hr) at a concentration of 5 mg/ml over a 14-day period following collagen injection.
  • osmotic pumps 200 ⁇ l, Alza, 0.5 ⁇ l/hr
  • IFN- ⁇ RKO IFN- ⁇ receptor
  • IFN- ⁇ RKO mice were back-crossed with DBA/1 wild-type mice for 10 generations to obtain the DBA/1 IFN- ⁇ RKO mice used in the present study.
  • IFN- ⁇ RKO and wild-type mice were bred in the Experimental Animal Centre of the University of Leuven. The experiments were performed in 8- to 12-week old mice, but in each experiment, the mutant and wild-type mice were age-matched with a 5 day limit. The male to female ratio was kept between 0.8 and 1.3 in each experimental group.
  • Collagen-induced arthritis was carried out in the following manner (see: Vermeire, et al., Int. J. Immunol. 158:5507-5513, (1997)).
  • Native chicken collagen type II EPC, Owensvillle, Mo.
  • IFA incomplete
  • CFA complete Freund's adjuvant
  • Mice were sensitized with a single 100 ⁇ l intradermal injection of the emulsion at the base of the tail. Mice were examined daily for signs of arthritis.
  • score 0 normal
  • score 1 redness and/or swelling in one joint
  • score 2 redness and/or swelling in more than one joint
  • score 3 redness and/or welling in the entire paw
  • score 4 deformity and/or ankylosis.
  • Spleens and fore and hind limbs were fixed in buffered saline—B5fixative (10% formalin with quicksilver).
  • tissues were fixed in 10% formalin or pure methanol (see: Vermeire, et al., J. Immunol. 158:5507-5513 (1997)).
  • Limbs were subsequently decalcified overnight with formic acid.
  • Four-micron thick paraffin sections were stained with hematoxylin and eosin. Severity of arthritis was evaluated using three parameters: infiltration of mono- and polymorphonuclear cells, hyperplasia of the synovium and parmus formation. Each parameter was scored on a scale from 0 to 3 (absent; weak, moderate and severe).
  • Monoclonal antibodies were produced from hybridomas grown by intraperitoneal inoculation in Pristane-primed athymic nude mice (nu/nu of NMRI background).
  • Neutralizing monoclonal antibody against MuIFN- ⁇ (F3, rate IgG 24 ) was purified by affinity chromatography on a mouse anti-rat K chain monoclonal antibody (Billiau, A., et al., J. Immunol. 140:1506 (1988)).
  • the neutralizing titer end-point dilution corresponding to 50% neutralization of the antiviral effect of 30 units/ml of mouse IFN- ⁇ on mouse 1929 cells challenged with mengovirus was 10 53 U/ml (IgG content, 1.4 mg/ml).
  • a neutralizing rate IgG 24 antibody against murine IL-12 was produced using hybridoma C17.8 (kindly provided by Dr. G. Trinchieri, Wistar Institute, Philadelphia, Pa.). The antibody was purified by affinity chromatography on proteinG (Pharmacia, Uppsala, Sweden). Antibody against murine IL-6 was prepared from ascites fluid from thymus-less nude mice inoculated with the 20F3 (rat x mouse) hybridoma (American Type Culture Collection, Rockville, Md.). This rat IgG antibody was purified by affinity chromatography on an anti-rat K chain monoclonal antibody-Sepharose column.
  • the neutralizing titer (endpoint dilution corresponding to 50% neutralization of the cell growth effect of 10 U of murine IL-6 per ml) was 10 55 (IgG content: 2.9 mg/ml).
  • Irrelevant rat IgG 24 was used as an isotope control and was prepared from ascites fluid of a rat plasmocytoma (obtained through the courtesy of Dr. H. Bazin, University of Louvain, Medical School, Brussels, Belgium). The IgG was purified by anion exchange chromatography on Hiload Q Sepharose and gel filtration on Superdex 200 (Pharmacia).
  • mice in the control group demonstrated arthritis, while only 1 of the 8 animals treated with AMD 3100 demonstrated disease.
  • the single treated animal did not develop arthritic pathology until after 20 days post-treatment. Additionally, the treated animals compared with the control animals did not demonstrate any significant body weight loss. Further, the treated animals maintained body weight consist with healthy animals not injected with collagen.
  • Compounds of the present invention may be used in the treatment of glioblastomas, fibromas, astrocytomas or myelomas affecting the central nervous system.
  • the compounds may be used according to standard clinical practice and procedures, using dosages as provided in the foregoing examples and according to clinical end points, such as imaging, immunological and other methodologies.
  • Compounds of the present invention may be used in the treatment of non-small cell lung cancer.
  • the compounds may be used according to standard clinical practice and procedures, using dosages as provided in the foregoing examples and according to clinical end points, such as imaging, immunological and other methodologies.
  • CXC chemokines have been found to regulate or are associated with the regulation of angiogenesis in non-small cell lung cancer (see: Arenberg, et al., J. of Leukocyte Biol. 62:554-562 (1997); and Moore, et al., TCM, vol. 8(2):51-58 (1998) Elsevier Science, Inc.).
  • the role of CXC chemokines and the binding to their respective receptors appear to play a significant role in the formation and/or proliferation of non-small cell lung cancer promoted by an increase in angiogenic activity.
  • the inhibition of the binding of these CXC chemokines to their natural receptor ligands by compounds of the present invention offer a new drug in the treatment tumors such as non-small cell lung cancer that are mediated or associated with increased levels of chemokines.
  • the compounds of the invention were tested in a screen by the MTT method ( J. Virol. Methods 120:309-321 (1988)).
  • MT-4 cells 2.5 ⁇ 10 4 /well
  • HIV-1 HIV-1
  • LAV-2 ROD HIV-2
  • CCID 50 concentration of 100 CCID 50
  • MTT tetrazolium
  • Antiviral activity and cytotoxicity of the compounds are expressed in Table 1 below as EC 50 ( ⁇ g/ml) and CC 50 ( ⁇ g/ml), respectively.
  • HIV is one of the most challenging viruses to combat, and the results given above provide an indication of activity against other retroviruses and against other viruses in general.
  • 2,6-Bis(2-aminoethyl)pyridine was prepared as described in Bridger et al. U.S. Pat. No. 5,698,546, which is hereby incorporated in its entirety by reference herein.

Abstract

The present invention is drawn to novel antiviral compounds, pharmaceutical compositions and their use. More specifically this invention is drawn to derivatives of monocyclic polyamines which have activity in standard tests against HIV- or FIV-infected cells as well as other biological activity related to binding of ligands to chemokine receptors that mediate a number of mammalian embryonic developmental processes.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a continuation of U.S. Ser. No. 11/281,296 filed 16 Nov. 2005, which is a continuation of U.S. Ser. No. 10/703,781 filed 7 Nov. 2003, which is a continuation of U.S. Ser. No. 09/740,050 filed 15 Dec. 2000 which claims priority under 35 U.S.C. § 119(e) to application Ser. No. 60/172,153 filed 17 Dec. 1999. The contents of these applications are incorporated herein by reference.
  • TECHNICAL FIELD
  • This invention generally relates to novel compounds, pharmaceutical compositions and their use. This invention more specifically relates to novel heterocyclic compounds that bind to chemokine receptors, including CXCR4 and CCR5, and demonstrate protective effects against infection of target cells by a human immunodeficiency virus (HIV).
  • BACKGROUND OF THE INVENTION
  • Approximately 40 human chemokines have been described, that function, at least in part, by modulating a complex and overlapping set of biological activities important for the movement of lymphoid cells and extravasation and tissue infiltration of leukocytes in response to inciting agents (See, for example: Ponath, P., Exp. Opin. Invest. Drugs 7:1-18 (1998); Baggiolini, M., Nature 392:565-568 (1998); Locati, et al., Annu. Rev. Med. 50:425-40 (1999)). These chemotactic cytokines, or chemokines, constitute a family of proteins, approximately 8-10 kDa in size. Chemokines appear to share a common structural motif, that consists of 4 conserved cysteines involved in maintaining tertiary structure. There are two major subfamilies of chemokines: the “CC” or β-chemokines and the “CXC” or α-chemokines. The receptors of these chemokines are classified based upon the chemokine that constitutes the receptor's natural ligand. Receptors of the β-chemokines are designated “CCR” while those of the α-chemokines are designated “CXCR.”
  • Chemokines are considered to be principal mediators in the initiation and maintenance of inflammation (see Chemokines in Disease published by Humana Press (1999), Edited by C. Herbert; Murdoch, et al., Blood 95:3032-3043 (2000)). More specifically, chemokines have been found to play an important role in the regulation of endothelial cell function, including proliferation, migration and differentiation during angiogenesis and re-endothelialization after injury (Gupta, et al., J. Biol. Chem. 7:4282-4287 (1998); Volin, et al., Biochem. Biophys Res. Commun. 242:46-53 (1998)). Two specific chemokines have been implicated in the etiology of infection by human immunodeficiency virus (HIV).
  • In most instances, HIV initially binds via its gp120 envelope protein to the CD4 receptor of the target cell. A conformational change appears to take place in gp120 which results in its subsequent binding to a chemokine receptor, such as CCR5 (Wyatt, et al., Science 280:1884-1888 (1998); Rizzuto, et al., Science 280:1949-1953 (1998); Berger, et al., Annu. Rev. Immunol. 17:657-700 (1999)). HIV-1 isolates arising subsequently in the infection bind to the CXCR4 chemokine receptor.
  • Following the initial binding by HIV to CD4, virus-cell fusion results, which is mediated by members of the chemokine receptor family, with different members serving as fusion cofactors for macrophage-tropic (M-tropic) and T cell line-tropic (T-tropic) isolates of HIV-1 (Carroll, et al., Science 276:273-276 (1997); Feng, et al., Science 272:872-877 (1996); Bleul, et al., Nature 382:829-833 (1996); Oberlin, et al., Nature 382:833-835 (1996); Cocchi, et al., Science 270:1811-1815 (1995); Dragic, et al., Nature 381:667-673 (1996); Deng, et al., Nature 381:661-666 (1996); Alkhatib, et al., Science 272:1955-1958, (1996)). During the course of infection within a patient, it appears that a majority of HIV particles shift from the M-tropic to the more pathogenic T-tropic viral phenotype (Blaak, et al., Proc. Natl. Acad. Sci. 97:1269-1274 (2000); Miedema, et al., Immune. Rev. 140:35 (1994); Simmonds, et al., J. Virol. 70:8355-8360 (1996); Tersmette, et al., J. Virol. 62:2026-2032, (1988); Connor, R. I., Ho, D. D., J. Virol. 68:4400-4408 (1994); Schuitemaker, et al., J. Virol. 66:1354-1360 (1992)). The M-tropic viral phenotype correlates with the virus's ability to enter the cell following binding of the CCR5 receptor, while the T-tropic viral phenotype correlates with viral entry into the cell following binding and membrane fusion with the CXCR4 receptor. Clinical observations suggest that patients who possess genetic mutations in CCR5 appear resistant, or less susceptible to HIV infection (Liu, et al., Cell 86:367-377 (1996); Samson, et al., Nature 382:722-725 (1996); Michael, et al., Nature Med. 3:338-340 (1997); Michael, et al., J. Virol. 72:6040-6047 (1998); Obrien, et al., Lancet 349:1219 (1997); Zhang, et al., AIDS Res. Hum. Retroviruses 13:1357-1366 (1997); Rana, et al., J. Virol. 71:3219-3227 (1997); Theodorou, et al., Lancet 349:1219-1220 (1997). Despite the number of chemokine receptors which have been reported to HIV mediate entry into cells, CCR5 and CXCR4 appear to be the only physiologically relevant coreceptors used by a wide variety of primary clinical HIV-1 strains (Zhang, et al., J. Virol. 72:9307-9312 (1998); Zhang, et al., J. Virol. 73:3443-3448 (1999); Simmonds, et al., J. Virol. 72:8453-8457 (1988)). Fusion and entry of T-tropic viruses that use CXCR4 are inhibited by the natural CXC-chemokine stromal cell-derived factor-1, whereas fusion and entry of M-tropic viruses that use CCR5 are inhibited by the natural CC-chemokines namely, Regulated on Activation Normal T-cell Expressed and Secreted (RANTES) and Macrophage Inflammatory proteins (MIP-1 alpha and beta).
  • In addition to serving as a co-factor for HIV entry, the direct interaction of virus-associated gp120 with CXCR4 has been recently suggested as a possible cause of CD8+ T-cell apoptosis and AIDS-related dementia via induction of neuronal cell apoptosis (Hesselgesser, et al., Curr. Biol. 8:595-598 (1998); Hesselgesser, et al., Curr. Biol. 7:112-121 (1997); Hesselgesser, et al., “Chemokines and Chemokine receptors in the Brain” in Chemokines in Disease published by Humana Press (1999), Edited by C. Herbert; Herbein, et al., Nature 395:189-194 (1998); Buttini, et al., Nature Med. 4:441-446 (1998); Ohagen, et al., J. Virol. 73:897-906 (1999); Biard-Piechaczyk, et al., Virology 268:329-344 (2000); Sanders, et al., J Neuroscience Res. 59:671-679 (2000); Bajetto, et al., J. Neurochem. 73:2348-2357 (1999); Zheng, et al., J. Virol. 73:8256-8267 (1999)).
  • However, the binding of chemokine receptors to their natural ligands appears to serve a more evolutionary and central role than only as mediators of HIV infection. The binding of the natural ligand, pre-B-cell growth-stimulating factor/stromal cell derived factor (PBSF/SDF-1) to the CXCR4 chemokine receptor provides an important signaling mechanism: CXCR4 or SDF-1 knock-out mice exhibit cerebellar, cardiac and gastrointestinal tract abnormalities and die in utero (Zou, et al., Nature 393:591-594 (1998); Tachibana, et al., Nature 393:591-594 (1998); Nagasawa, et al., Nature 382:635-638 (1996)). CXCR4-deficient mice also display hematopoietic defects (Nagasawa, et al., Nature 382:635-638 (1996)); the migration of CXCR4 expressing leukocytes and hematopoietic progenitors to SDF-1 appears to be important for maintaining B-cell lineage and localization of CD34+ progenitor cells in bone marrow (Bleul, et al., J. Exp. Med. 187:753-762 (1998); Viardot, et al., Ann. Hematol. 77:195-197 (1998); Auiti, et al., J. Exp. Med. 185:111-120 (1997); Peled, et al., Science 283:845-848 (1999); Qing, et al., Immunity 10:463-471 (1999); Lataillade, et al., Blood 95:756-768 (1999); Ishii, et al., J. Immunol. 163:3612-3620 (1999); Maekawa, et al., Internal Medicine 39:90-100 (2000); Fedyk, et al., J Leukocyte Biol. 66:667-673 (1999); Peled, et al., Blood 95:3289-3296 (2000)).
  • The signal provided by SDF-1 on binding to CXCR4 may also play an important role in tumor cell proliferation and regulation of angiogenesis associated with tumor growth (See “Chemokines and Cancer” published by Humana Press (1999); Edited by B. J. Rollins; Arenburg, et al., J. Leukocyte Biol. 62:554-562 (1997); Moore, et al., J. Invest. Med. 46:113-120 (1998); Moore, et al., Trends cardiovasc. Med. 8:51-58 (1998); Seghal, et al., J. Surg. Oncol. 69:99-104 (1998)); the known angiogenic growth factors VEG-F and bFGF, up-regulate levels of CXCR4 in endothelial cells, and SDF-1 can induce neovascularization in vivo (Salcedo, et al., Am. J. Pathol. 154:1125-1135 (1999)); Leukemia cells that express CXCR4 migrate and adhere to lymph nodes and bone marrow stromal cells that express SDF-1 (Burger, et al., Blood 94:3658-3667 (1999); Arai, et al., Eur. J. Haematol. 64:323-332 (2000); Bradstock, et al., Leukemia 14:882-888 (2000); Burger et al., Blood 96:265502663 (2000); Morle, R. et al. Brit. J. Haematol. 110:561-582 (2000).
  • The binding of SDF-1 to CXCR4 has also been implicated in the pathogenesis of atherosclerosis (Abi-Younes, et al., Circ. Res. 86:131-138 (2000)), renal allograft rejection (Eitner, et al., Transplantation 66:1551-1557 (1998)), asthma and allergic airway inflammation (Yssel, et al., Clinical and Experimental Allergy 28:104-109 (1998); Gonzalo, et al., J. Immunol. 165:499-508 (2000)), Alzheimer's disease (Xia, et al., J. Neurovirology 5:32-41 (1999)) and Arthritis (Nanki, et al., J. Immunol. 164:5010-5014 (2000)).
  • In attempting to better understand the relationship between chemokines and their receptors, recent experiments to block the fusion, entry and replication of HIV via the CXCR4 chemokine receptor were carried out through the use of monoclonal antibodies or small molecules that appear to suggest a useful therapeutic strategy (Schols, et al., J. Exp. Med. 186:1383-1388 (1997); Schols, et al., Antiviral Research 35:147-156 (1997); Bridger, et al., J. Med. Chem. 42:3971-3981 (1999); Bridger, et al., “Bicyclam Derivatives as HIV Inhibitors” in Advances in Antiviral Drug Design Volume 3, p 161-229; Published by JAI press (1999); Edited by E. De Clercq). Small molecules, such as bicyclams, appear to specifically bind to CXCR4 and not CCR5 (Donzella, et al., Nature Medicine 4:72-77 (1998)). These experiments demonstrated interference with HIV entry and membrane fusion into the target cell in vitro. More recently, bicyclams were also shown to inhibit fusion and replication of Feline Immunodeficiency Virus (FIV) that uses CXCR4 for entry (Egberink, et al., J. Virol. 73:6346-6352 (1999)).
  • Additional experiments have shown that the bicyclam dose-dependently inhibits binding of 125I-labeled SDF-1 to CXCR4 and the signal transduction (indicated by an increase in intracellular calcium) in response to SDF-1. Thus, the bicyclam also functioned as an antagonist to the signal transduction resulting from the binding of stromal derived factor or SDF-1α, the natural chemokine to CXCR4. Bicyclams also inhibited HIV gp120 (envelope)-induced apoptosis in non-HIV infected cells (Blanco, et al., Antimicrobial Agents and Chemother. 44:51-56 (2000)).
  • U.S. Pat. No. 5,583,131, U.S. Pat. No. 5,698,546, U.S. Pat. No. 5,817,807, U.S. Pat. No. 5,021,409 and U.S. Pat. No. 6,001,826, which are incorporated herein by reference, disclose cyclic compounds that are active against HIV-1 and HIV-2 in in vitro tests. It was subsequently disclosed, in published PCT application PCT/CA99/00619 (WO 00/02870), incorporated herein by reference, that these compounds as well as additional compounds with less complex structures exhibit anti-HIV activity by binding to the chemokine receptor CXCR4 and/or CCR5 expressed on the surface of certain cells of the immune system. This competitive binding protects these target cells, which utilize the CXCR4 receptor for entry from infection by HIV. In addition, the compounds antagonize the binding, signaling and chemotactic effects of the natural ligand for CXCR4, i.e., the chemokine stromal cell-derived factor 1α (SDF-1), and also have protective effects against HIV infection of target cells by binding in vitro to the CCR5 receptor.
  • Additionally, published PCT application PCT/CA00/00104, (WO 00/45814), incorporated herein by reference, discloses that the cyclic polyamine antiviral agents described in the above-mentioned documents have the effect of enhancing production of white blood cells as well as exhibiting antiviral properties. Thus, these agents are useful for controlling the side-effects of chemotherapy, enhancing the success of bone marrow transplantation, enhancing wound healing and burn treatment, as well as combating bacterial infections in leukemia.
  • Published PCT application PCT/CA00/00321 (WO 00/56729), incorporated herein by reference, discloses additional heterocyclic compounds that exhibit anti-HIV activity by binding to the chemokine receptors CXCR4 and CCR5 expressed on the surface of certain cells of the immune system. This competitive binding thereby protects these target cells, which utilize the CXCR4 or CCR5 receptors for entry, from infection by HIV. In addition, these compounds antagonize the binding, signaling and chemotactic effects of the natural ligand for CXCR4, i.e., the chemokine stromal cell-derived factor 1α (SDF-1) and/or the natural ligand for CCR5, the chemokine RANTES.
  • Citation of the above documents is not intended as an admission that any of the foregoing is pertinent prior art. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents. Further, all documents referred to throughout this application are hereby incorporated in their entirety by reference herein.
  • The present invention describes novel compounds that exhibit protective effects against HIV infection of target cells by binding to the chemokine receptors CXCR4 or CCR5, in a similar manner to the previously disclosed macrocyclic compounds, and that are additionally useful in other indications addressed by the compounds as described above.
  • DISCLOSURE OF THE INVENTION
  • The present invention provides novel compounds that bind chemokine receptors and interfere with the binding of the natural ligand thereto, and are useful as agents demonstrating protective effects on target cells from HIV infection. The invention compounds act as antagonists or agonists of chemokine receptors and exhibit biological activities related to the ability of these compounds to inhibit the binding of chemokines to their receptors.
  • Accordingly, the present invention provides a compound of Formula 1
    V—CR2—Ar1—CR2NR—(CR2)x—Ar2  (1)
  • including the pharmaceutically acceptable salts and protected forms thereof,
  • wherein V is a substituted heterocycle of 9-24 members containing 2-4 optionally substituted amine nitrogen atoms spaced from each other by 2 or more optionally substituted carbon atoms, and which heterocycle may optionally comprise a fused aromatic or heteroaromatic ring, and wherein
  • (a) said heterocycle contains at least one O or S, said O or S spaced from any adjacent heteroatom by at least 2 carbon atoms, and wherein said S is optionally oxidized or
  • (b) at least one carbon atom in said ring is substituted by an electron-withdrawing substituent, or
  • (c) both (a) and (b);
  • and wherein each R is independently H or a straight chain, branched or cyclic alkyl containing 1-6C;
  • x is 0-4;
  • Ar1 is an unsubstituted or substituted aromatic or heteroaromatic moiety; and
  • Ar2 is an unsubstituted or substituted aromatic or heterocyclic group.
  • Other aspects of the invention are directed to the pharmaceutical compositions comprising a therapeutically effective amount of the compound of Formula 1 and to methods of treating a condition of the human body or the bodies of other mammals comprising the administration of a pharmaceutical or veterinary composition which contains a therapeutically effective amount of the compound of Formula 1. In other aspects, the invention is directed to a method for blocking or interfering with the binding of a chemokine receptor with its natural ligand, by contacting the chemokine receptor with an effective amount of the compound of Formula 1.
  • In still other aspects, the invention includes the use of a compound of Formula 1 in the manufacture of a medicam ent for the treatment of a disease in which blocking or interfering with binding of a chemokine receptor with its natural ligand is advantageous and for protecting target cells possessing chemokine receptors, the binding to which by a pathogenic agent results in disease or pathology. The invention is also directed to methods of treatment as outlined above.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the structure of N-[4-(11-Fluoro-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8897).
  • FIG. 2 shows the structure of N-[4-(11,11-difluoro-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8880).
  • FIG. 3 shows the structure of N-[4-(1,4,7-triazacyclotetradecan-2-one)-yl))-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8748).
  • FIG. 4 shows the structure of N-[12-(5-oxa-1,9-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8922).
  • FIG. 5 shows the structure of Preparation of N-[4-(11-oxa-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8779).
  • FIG. 6 shows the structure of N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD8834).
  • FIG. 7 shows the structure of N-[4-(11-sulfoxo-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD9424).
  • FIG. 8 shows the structure of N-[4-(11-sulfono-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt) (AMD9408).
  • FIG. 9 shows the structure of N-[4-(3-carboxo-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD-Exp40).
  • FIG. 10 shows the structure of 1,1′-[1,4-phenylenebis(methylene))]bis-1,4,8,11-tetraazacyclotetradecane (AMD3100).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to compounds of Formula 1 which can act as agents that modulate chemokine receptor activity. Such chemokine receptors include but are not limited to CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8 and CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5.
  • The compounds of Formula 1 that demonstrate protective effects on target cells from HIV infection so as to bind specifically to the chemokine receptor, affect the binding of a natural ligand or chemokine to a receptor such as CXCR4 and/or CCR5. They are also useful as agents which affect chemokine receptors, such as CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8 and CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5 where such chemokine receptors have been correlated as being important mediators of many human inflammatory as well as immunoregulatory diseases. Thus, the compounds of Formula 1, which modulate the activity of such chemokine receptors are useful for the treatment or prevention of such diseases.
  • The term “modulators” as used herein encompasses antagonist, agonist, partial antagonist, and or partial agonist, inhibitors, and activators. In a preferred embodiment of the present invention, compounds of Formula 1 demonstrate protective effects against HIV infection by inhibiting binding of HIV to a chemokine receptor, such as CXCR4 and/or CCR5 of a target cell.
  • The compounds of Formula 1 that inhibit chemokine receptors may be used for the treatment of diseases associated with hematopoiesis, including but not limited to, controlling the side-effects of chemotherapy, enhancing the success of bone marrow transplantation, enhancing wound healing and burn treatment, as well as combating bacterial infections and leukemia.
  • These compounds of Formula 1 are thus also useful for the treatment of diseases that are associated with inflammation, including but are not limited to, inflammatory or allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type hypersensitivity, asthma, interstitial lung disease (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or dermatomyositis); systemic anaphylaxis or hypersensitivity responses, drug allergies, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, psoriatic arthritis, systemic lupus erythematosus, myastenia gravis, juvenile onset diabetes; glomerulonephritis, autoimmune throiditis, graft rejection, including allograft rejection or graft-versus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); eosinphilic myotis, eosiniphilic fasciitis; and cancers.
  • The compounds of the invention that activate or promote chemokine receptor function may be used for the treatment of diseases that are associated with immunosuppression such as individuals undergoing chemotherapy, radiation therapy, enhanced wound healing and burn treatment, therapy for autoimmune disease or other drug therapy (e.g., corticosteroid therapy) or combination of conventional drugs used in the treatment of autoimmune diseases and graft/transplantation rejection, which causes immunosuppression; immunosuppression due to congenital deficiency in receptor function or other causes; and infectious diseases, such as parasitic diseases, including but not limited to helminth infections, such as nematodes (round worms); Trichuriasis, Enterobiasis, Ascariasis, Hookworm, Strongyloidiasis, Trichinosis, filariasis; trematodes; visceral worms, visceral larva migtrans (e.g., Toxocara), eosinophilic gastroenteritis (e.g., Anisaki spp., Phocanema ssp.), cutaneous larva migrans (Ancylostona braziliense, Ancylostoma caninum); the malaria-causing protozoan Plasmodium vivax, Human cytomegalovirus, Herpesvirus saimiri, and Kaposi's sarcoma herpesvirus, also known as human herpesvirus 8, and poxvirus Moluscum contagiosum.
  • Compounds of Formula 1 may be used in combination with any other pharmaceutical composition where such combined therapy may be useful to modulate chemokine receptor activity and thereby prevent and treat inflammatory and immunoregulatory diseases, including, for example, in combinations with one or more agents useful in the prevention or treatment of HIV. Examples of such agents include:
  • (1) nucleotide reverse transcriptase inhibitor such as zidovudine, didanosine, lamivudine, zalcitabine, abacavir, stavudine, adefovir, adefovir dipivoxil, fozivudine todoxil, etc.;
  • (2) non-nucleotide reverse transcriptase inhibitor (including an agent having anti-oxidation activity such as immunocal, oltipraz, etc.) such as nevirapine, delavirdine, efavirenz, loviride, immunocal, oltipraz, etc.; and
  • (3) protease inhibitors such as saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, palinavir, lasinavir, Kaletra™ (lopinavir/ritonavir), etc.
  • Such combinations the compounds of the present invention and other HIV agents may be administered separately or in conjunction; the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • The compounds of Formula 1 may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • The compounds of Formula 1 may be used to treat animals, including mice, rats, horses, cattle, sheep, dogs, cats, and monkey, and are also effective for use in humans.
  • The compounds of the invention may be supplied as “pro-drugs,” or, protected forms of the compounds of Formula 1, which release the compound after administration to a patient. For example, the compound may carry protective groups which are split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing active compound or is oxidized or reduced in body fluids to release the compound. A discussion of pro-drugs may be found in Smith and Williams' Introduction to the Principles of Drug Design, H. J. Smith, Wright, Second Edition, London 1988.
  • Acid addition salts, which are pharmaceutically acceptable, such as salt with inorganic base, a salt with organic base, a salt with inorganic acid, a salt with organic acid, a salt with basic or acidic amino acid, etc. and non-toxic metal complexes are also encompassed in the present invention. Examples of a salt with an inorganic base include a salt with alkali metal (e.g. sodium, potassium, etc.), alkaline earth metal (e.g. calcium, magnesium, etc.), aluminum, ammonium, etc. Examples of the salt with an organic base include a salt with trimethylamine, triethylamine, pyridine, picoline, ethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, N,N′-dibenzylethylenediamine etc. Examples of the salt with an inorganic acid include a salt with hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, etc. Examples of the salt with an organic acid include a salt with formic acid, oxalic acid, acetic acid, tartaric acid, methanesulfonic acid, benzenesulfonic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, etc. Examples of salts with basic amino acids include a salt with arginine, lysine, ornithine, etc. Examples of salts with the acidic amino acid include a salt with aspartic acid, glutamic acid, etc. Non-toxic in the present tense has to be considered with reference to the prognosis for the infected subject without treatment. Copper and zinc complexes are preferred although other metals such as nickel, cobalt or rhubidium, may be used.
  • The compounds of Formula 1 may form hydrates or solvates. Some compounds of Formula 1 exist as regioisomers, configurational isomers, conformers, diasteroisomeric forms and mixtures of diasteroisomeric forms thereof; it is possible to isolate individual isomers using known separation and purification methods, if desired. The invention includes mixtures of these stereoisomers as well as isolated forms. The mixtures may contain the stereoisomers in any ratio. Compounds of the invention also include racemates, which can be separated into the (S)-compounds and (R)-compounds by optical resolution; individual optical isomers and mixtures thereof are included in the scope of the present invention.
  • The compounds of Formula 1 may be administered alone or as an admixture with a pharmaceutically acceptable carrier (e.g. solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.) may be orally or non-orally administered. Examples of non-oral formulations include injections, drops, suppositories and pessaryies.
  • In the above Formula 1, V contains 2-4 N, preferably 3-4 N if there is no additional heteroatom. Preferable ring sizes for V are 9-18 members, more preferably 12-16 members. V may also include a fused aromatic or heteroaromatic ring, preferably 1, 2 or 1, 3 or 1,4 phenylene or 2, 6 or 2, 5 or 2, 4 or 2,3 pyridinylene. The fused ring may also be, for example, 2, 5 or 2,6 pyrimidinylene or 2, 4 or 2,3 pyrrolylene.
  • In the above Formula 1, the required electron withdrawing substituents present at at least one C in ring V may be halogen, nitro, cyano, carboxylic acid, a carboxylic ester with an alcohol of 1-6C or amide formed from an amine of 0-12C, a sulfonic or sulfinic acid or a sulfonic or sulfinic ester or amide, CF3, and the like. A preferred electron withdrawing substituent is ═O, as well as halo.
  • Examples of halogen include fluorine, chlorine, bromine, iodine, etc., with fluorine and chlorine preferred.
  • In the above Formula 1, Ar2 is an optionally substituted heterocyclic group or aromatic group. Examples of aromatic groups include benzene and naphthalene, or dihydronaphthalene and tetrahydronaphthalene. Examples of heterocyclic groups include 5 to 6-membered saturated, partially saturated, or aromatic heterocyclic rings containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur. The heterocycles may be pyridine, quinoline, isoquinoline, imidazole, benzimidazole, azabenzimidazole, benzotriazole, furan, benzofuran, thiazole, benzothiazole, oxazole, benzoxazole, pyrrole, indole, indoline, indazole, pyrrolidine, pyrrolidone, pyrroline, piperidine, piperazine, tetrahydroquinoline, tetrahydroisoquinoline, pyrazole, thiophene, isoxazole, isothiazole, triazole, tetrazole, oxadiazole, thiadiazole, morpholine, thiamorpholine, pyrazolidine, imidazolidine, imidazoline, tetrahydropyran, dihydropyran, benzopyran, dioxane, dithiane, tetrahydrofuran, tetrahydrothiophene, dihydrofuran, dihydrothiophene, etc. Oxides of the nitrogen and sulfur containing heterocycles are also included in the present invention.
  • The optional substituents on Ar2 include alkyl(1-6C), alkenyl(1-6C), alkynyl(1-6C), halo, nitro, cyano, carboxylic acid, carboxylic ester formed from an alcohol with 1-6C or amide formed from an amine of 0-12C, a sulfonic or sulfinic acid or ester or amide, OR, SR, NR2, OCR, OOCR, NRCOR, all wherein R is hydrogen or straight or branched chain alkyl(1-6C), an optionally substituted aromatic or heterocyclic group, CF3, and the like.
  • Preferred substituents include alkyl, OR, NR2, and halo. Preferred embodiments of Ar2 include phenyl, pyridinyl, pyrimidinyl and imidazolyl.
  • In Formula 1, Ar1 is a 5-6 membered aromatic system which is bivalent benzene, pyridine, thiophene, pyrimidine, and the like. Ar1 may optionally be substituted by alkyl, alkenyl, halo, nitro, cyano, CF3, COOR, CONR2, OCR, OOCR, NRCOR, OR, NR2, SR, (where R is H or alkyl 1-6C) sulfonic or sulfinic acids, esters or amides and the like. Preferred embodiments of Ar1 are phenylene, especially 1,3 and 1,4 phenylene and pyridinylene, preferably 2,6 pyridinylene, and 3,5 pyridinylene. Preferable substituents are alkyl, OR, NR2 and halo.
  • Further, in the compounds of Formula 1, it is preferred that each R group be hydrogen or alkyl of 1-2C, preferably hydrogen. In another preferred embodiment, the R group coupled to a nitrogen is hydrogen or alkyl 1-6C, preferably straight chain alkyl 1-3C, more preferably H or methyl. In other preferred embodiments, 1, 2, 3, 4, or 5 of the R groups are methyl or ethyl and the remaining R groups are hydrogen.
  • Thus, in one preferred embodiment, the compound of Formula 1 is of the formula
    V—CH2—Ar1—CH2NR—CH2—Ar2
  • wherein V is (a) substituted with halo or ═O or (b) contains O or S or (c) both (a) and (b), and wherein Ar1 is unsubstituted 1, 3 or 1,4-phenylene, R is H, methyl or ethyl and Ar2 is unsubstituted phenyl or pyridinyl.
  • Preferred embodiments of x are 0-2 and 1-2.
  • In the treatment or prevention of conditions which require chemokine receptor modulation an appropriate dosage level will generally be about 0.01 to 500 mg per kg body weight per day which can be administered in singe or multiple doses. Preferably, the dosage level will be about 0.1 to about 250 mg/kg per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound used, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the patient undergoing therapy.
  • Thus, the active compounds may be administered in the form of a pharmaceutical composition formulated according to well known principles and incorporating the compound, preferably in unit dose form, in combination with a pharmaceutically acceptable diluent, carrier or excipient. Such compositions may be in the form of solutions or suspensions for injection, or irrigation or be in capsule, tablet, dragee, or other solid composition or as a solution or suspension for oral administration or formulated into pessaries or suppositories or sustained release forms of any of the above for implantation. Suitable diluents, carriers, excipients and other components are well known. It may be desirable also to formulate a composition for topical administration such as an ointment or cream.
  • The pharmaceutical compositions according to the invention may be formulated in unit dosages determined in accordance with conventional pharmacological methods, suitably to provide active compounds in the dosage range in humans or animals of from 0.01 to 100 mg/kg body weight per day, in a single dose or in a number of smaller doses. Preferred dosage ranges are 0.01 to 30 mg/kg body weight per day intravenous (iv) or intraperitoneal (ip). Other active compounds may be used in the compositions or such active compounds or supplemental therapy may be included in a course of treatment. The pharmaceutical compositions are useful for treatment of a patient comprising an effective therapeutic amount of the novel compound, where said compound effectively binds to a chemokine receptor.
  • The present invention further contemplates the use of these compositions in the manufacture of a medicament for the treatment of HIV- or FIV-infected patients and/or the treatment of a disease by the regulation of endothelial cell function and/or the treatment of a disease relating to vascularization of the gastrointestinal tract; haematopoiesis; or cerebellar development.
  • In a method for treating a patient infected with HIV or FIV, the pharmaceutical composition is administered to said patient as a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier. In a method of treating a patient with a disease related to the regulation of endothelial cell function, the pharmaceutical composition is administered to said patient as a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier. The present invention further contemplates methods of treating a patient with a disease relating to vascularization of the gastrointestinal tract; haematopoiesis; or cerebellar development, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier.
  • The present invention further contemplates a method of treating a patient with a disease relating to basal leukocyte trafficking or the extravasation and tissue infiltration of leukocytes in response to inciting antigens, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier. The present method also contemplates treating a patient, by administering to said patient a therapeutically effective amount of a pharmaceutical composition in a pharmaceutically acceptable carrier, wherein said compound effectively binds to a chemokine receptor.
  • The present invention further contemplates pharmaceutical compositions and methods of use for the treatment of humans or animals for: renal allograft rejection; inflammatory disease; cancer; central nervous system developmental disease; HIV; vasculature development disease; haematopoiesis and other chemokine mediated diseases or disorders. The invention further provides for the treatment of diseases, which include, but are not limited to: arthritis; asthma; multiple sclerosis; dementia from HIV or FIV infection, Parkinson's disease, Alzheimer's disease and inflammatory diseases. The pharmaceutical compositions and methods of use of the present invention further provide for the treatment of cancers, that include, but are not limited to those associated with: solid tumors; lymphoma; metastatic tumors; glioblastoma tumors; leukemia; and other carcinomas tumors. The pharmaceutical compositions of the present invention are useful for the treatment of cancers that include, but are not limited to: non-small cell lung cancer; lung cancer; breast cancer; prostate cancer; and cancer of other organs.
  • Other diseases or disorders that are contemplated to be treated with the pharmaceutical compositions of the present invention, include, but are not limited to: disorders treated by inhibiting or promoting angiogenesis or by inducing stasis of angiogenesis; developmental disorders mediated by chemokines.
  • The present invention further provides methods for the prevention of a disease or disorder in a patient by administering a therapeutically effective dosage of the pharmaceutical compositions of the present invention to a patient over a period of time sufficient to effectively prevent the disease or disorder.
  • The following examples are intended to illustrate but not to limit the invention.
  • EXAMPLE 1 Preparation of N-[4-(11-Fluoro-1,4,7-triazacyclotetradecanyl 1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine. (AMD8897, FIG. 1) Acetic acid, 7-acetoxy-4-hydroxy-heptyl ester
  • Figure US20070060591A1-20070315-C00001
  • To a solution of heptane-1,4,7-triol (162 mg, 1.09 mmol) in pyridine (4 ml) was added acetic anhydrous (216 mg, 2.28 mmol) at 0° C. The resulting mixture was allowed to stir for 4 hours at 0° C. and then diluted with ethyl acetate (100 ml). The organic solution was washed with sat. NaHCO3, brine and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel, using 30% ethyl acetate in hexanes, gave the title compound (120 mg, 50%) as a colorless oil. 1H NMR (CDCl3) δ 1.41-1.86 (m, 8H), 2.05 (s, 6H), 3.62-3.70 (m, 1H), 4.11 (t, 4H, J=6.6 Hz); 13C NMR (CDCl3) δ 21.01, 24.92, 33.82, 64.41, 71.06, 171.20.
  • Acetic acid, 7-acetoxy-4-fluoro-heptyl ester
  • Figure US20070060591A1-20070315-C00002
  • To a solution of (diethylamino)-sulfur trifluoride (571 mg, 3.54 mmol) in CH2Cl2 was added the solution of acetic acid, 7-acetoxy-4-hydroxy-heptyl ester (403 mg, 1.74 mmol) in CH2Cl2 (10 ml) at −78° C. The resulting mixture was allowed to stir for 15 minutes at −78° C., 30 minutes at room temperature and then diluted with ethyl acetate (300 ml). The organic solution was washed with sat. NaHCO3, brine and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel, using 15% ethyl acetate in hexanes, gave the title compound (386 mg, 94%) as a colorless oil. 1H NMR (CDCl3) δ 1.55-1.83 (m, 8H), 2.05 (s, 6H), 4.05-4.15 (m, 4H), 4.39-4.63 (m, 1H); 13C NMR (CDCl3) δ 20.97, 24.42, 24.48, 31.51, 31.79, 64.03, 92.22, 94.45, 171.12; 19F NMR (CDCl3) δ 106.44-105.27(m); ES-MS m/z 257.3 (M+Na).
  • 4-Fluoro-heptane-1,7-diol
  • Figure US20070060591A1-20070315-C00003
  • Methanol (15 ml) saturated with anhydrous NH3 gas was added to acetic acid, 7-acetoxy-4-fluoro-heptyl ester (500 mg, 2.13 mg) contained in round-bottomed flask closed by glass stopper. The mixture was allowed to stir for over 28 hours at room temperature and then concentrated. Purification of the residue by column chromatography on silica gel, using 5% methanol in CH2Cl2, gave the title compound (320 mg, 100%) as pure colorless oil. 1H NMR (CDCl3) δ 1.59-1.83 (m, 8H), 3.67 (t, 4H, J=5.7 Hz), 4.47-4.67 (m, 1H); 3C NMR (CDCl3) δ 28.73, 28.75, 31.78, 32.06, 62.88, 93.56, 95.98; ES-MS m/z 173.2 (M+Na).
  • 1,7-Bis(toluene-4-sulfonic acid)-4-fluoro-heptyl-ester
  • Figure US20070060591A1-20070315-C00004
  • To a pre-cooled (ice bath) solution of 4-fluoro-heptane-1,7-diol (320 mg, 2.13 mmol) and triethylamine (1.0 ml, 6.90 mmol) in CH2Cl2 (10 ml) was added a solution of p-toluensulfonyl chloride (812 mg, 4.27 mmol) in CH2Cl2 (2 ml). The resulting mixture was stirred for 18 hours at room temperature and then diluted with ethyl acetate (200 ml). The organic solution was washed with sat. NaHCO3, brine and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel, using 20% ethyl acetate in hexanes, gave the title compound (627 mg, 100%) as a colorless oil. 1H NMR (CDCl3) δ 1.52-1.86 (m, 8H), 2.45 (s, 6H), 4.00-4.08 (m, 4H), 4.28-4.44 (m, 1H), 7.37 (d, 4H, J=8.4 Hz), 7.77 (d, 4H, J=8.4 Hz); 13C NMR (CDCl3) δ 22.06, 25.09, 25.15, 31.32, 31.60, 70.34, 91.97, 94.21, 128.29, 130.30, 133.36, 145.28; 19F NMR (CDCl3) δ −107.54-107.02 (m); ES-MS m/z 481.3 (M+Na).
  • [11-Fluoro-1,7-bis(2-nitro-benzenesulfonyl)-1,4,7-triaza-cyclotetradec-4-yl]-phosphonic acid diethyl ester
  • Figure US20070060591A1-20070315-C00005
  • To a stirred solution of bis-[2-(2-nitro-benzenesulfonylamino)-ethyl]-phosphoramidic acid diethyl ester (1.07 g, 1.75 mmol) and anhydrous Cs2CO3 (1.5 g, 4.60 mmol) in anhydrous DMF (100 ml) at 80° C. under N2 was added a solution of 1,7-bis(toluene-4-sulfonic acid)-4-fluoro-heptyl-ester (687 mg, 1.49 mmol) in DMF (10 ml) over 10 hours. The reaction mixture was allowed to stir at 85° C. for further 30 hours, cooled to room temperature and then concentrated. The residue was diluted with 200-ml ethyl acetate and washed with a sat. NaHCO3 brines and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel using 50% ethyl acetate in CH2Cl2 gave the title compound (560 mg, 55%) as a light yellow solid. 1H NMR (CDCl3) δ 1.31 (ddd, 6H, J=0.6, 7.2, 7.2 Hz), 1.65-1.89 (m, 8H), 3.22-3.46 (m, 12H), 3.93-4.14 (m, 4H), 4.67-4.83 (m, 1H), 7.60-7.65 (m, 2H), 7.68-7.75 (m, 4H), 7.99-8.05 (m, 2H); 13C NMR (CDCl3) δ 16.54, 16.64, 22.65, 22.74, 29.66, 29.95, 47.04, 47.37, 47.43, 50.22, 63.17, 63.25, 91.38, 93.62, 124.59, 131.67, 131.96, 132.06, 134.22, 148.69; 19F NMR (CDCl3) δ −99.84-99.41 (m); ES-MS m/z 724.6 (M+H).
  • 11-Fluoro-1,7-bis-(2-nitro-benzenesulfonyl)-1,4,7-triazacyclotetradecane
  • Figure US20070060591A1-20070315-C00006
  • Acetic acid (15 ml), saturated with anhydrous hydrogen bromide (gas) was added to [11-fluoro-1,7-bis(2-nitro-benzenesulfonyl)-1,4,7-triaza-cyclotetradec-4-yl]-phosphonic acid diethyl ester (560 mg, 0.77 mmol) contained in round-bottomed flask closed by glass stopper. The resulting mixture was allowed to stir for 18 hours at room temperature and then diethyl ether (50 ml) was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the diethyl ether solution was decanted off. This white solid was re-dissolved in methanol, stirred with K2CO3 (solid) for 30 mins then diluted with 100-ml ethyl acetate, and the solids were removed by filtration. Evaporation of the filtrates gave the title compound (454 mg, 100%) as white foam. 1H NMR (CDCl3) δ 1.75-1.99 (m, 8H), 2.90 (t, 4H, J=5.0 Hz), 3.26-3.39 (m, 8H), 4.61-4.77 (m, 1H), 7.60-7.65 (m, 2H), 7.68-7.72 (m, 4H), 7.94-7.97 (m, 2H); ES-MS m/z 588.3 (M+H).
    Figure US20070060591A1-20070315-C00007
  • To a stirred solution of 11-fluoro-1,7-bis-(2-nitrobenzenesulfonyl)-1,4,7-triaza-cyclotetradecane (454 mg, 0.77 mmol) and anhydrous K2CO3 (400 mg, 2.89 mmol) in anhydrous CH3CN (7 ml) under N2 was added N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (Bridger, et al., U.S. Ser. No. 09/111,895) (743 mg, 1.72 mmol). The reaction mixture was allowed to stir at 85° C. for further 18 hours and then concentrated. The residue was diluted with 100-ml ethyl acetate and washed with a sat. NaHCO3, then brines and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel using 50% ethyl acetate in CH2Cl2 gave the title compound (445 mg, 59%) as white form. 1H NMR (CDCl3) δ 1.67-1.83 (m, 8H), 2.69-2.76(m, 4H), 3.19 (td, 4H, J=8.1, 8.1 Hz), 3.34 (t, 4H, J=6.2 Hz), 3.75 (s, 2H), 4.50 (s, 2H), 4.60 (s, 2H), 4.59-4.80 (m, 1H), 7.08-7.20 (m, 7H), 7.51-7.73 (m, 9H), 7.81 (dd, 2H, J=1.8, 7.5 Hz), 8.01 (d, 1H, J=7.8 Hz), 8.38 (d, 1H); 19F NMR (CDCl3) δ −99.61-98.50 (m); ES-MS m/z 983.3 (M+H).
  • N-[4-(11-Fluoro-1,4,7-triazacyclotetradecanyl)-1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8897)
  • Figure US20070060591A1-20070315-C00008
  • To a stirred solution of the intermediate from above (445 mg, 0.45 mmol) and anhydrous K2CO3 (750 mg, 5.43 mmol) in anhydrous DMF (6 ml) under N2 was added dropwise, thiophenol (348 mg, 3.17 mmol). The reaction mixture was allowed to stir at room temperature for further 4 hours and then concentrated. The residue was diluted with 100-ml ethyl acetate and solids were removed by filtration through a short column of celite. Evaporation of the solvent and purification of the residue on a chromatron using silica gel (1 mm plate) and an eluent of 1:1:98 Methanol/NH4OH/CH2Cl2 gave AMD8897 (80 mg, 62%). 1H NMR (CDCl3) δ 1.52-1.85 (m, 8H), 2.53-2.63 (m, 12H), 3.57 (s, 2H), 3.83 (s, 2H), 3.93 (s, 2H), 5.07-5.30 (m, 1H), 7.14-7.33 (m, 6H), 7.64 (ddd, 1H, J=1.8, 7.8, 7.8 Hz), 8.55-8.57 (m, 1H); 13C NMR (CDCl3) δ 24.43, 32.13, 32.41, 47.32, 48.47, 52.44, 53.21, 54.52, 58.92, 121.94, 122.32, 128.26, 128.87, 136.42, 137.80, 139.04, 149.33, 159.75; 19F NMR (CDCl3) δ −102.30-103.00 (m); ES-MS m/z 428.3 (M+H); Anal. Calcd. for (C25H38FN5): C, 70.22; H, 8.96; N, 16.38. Found: C, 69.99; H, 8.96; N, 16.42.
  • EXAMPLE 2 Preparation of N-[4-(11,11-difluoro-1,47-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine. (AMD8880, FIG. 2) 4,4-Difluoro-heptanedioic acid diethyl ester
  • Figure US20070060591A1-20070315-C00009
  • To a neat solution of (diethylamino)-sulfur trifluoride (2.02 g, 12.55 mmol) in a plastic bottle was added 4-oxo-heptanedioic acid diethyl ester (2.56 g, 11.13 mmol) at room temperature. The resulting mixture was allowed to stir for 12 days and then diluted with ethyl acetate (500 ml). The organic solution was washed with sat. NaHCO3, brine and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel, using 20% ethyl acetate in hexanes, gave the title compound (1.2 g, 43%) as pure colorless oil. 1H NMR (CDCl3) δ 1.26 (t, 6H, J=8.0 Hz), 2.11-2.28 (m, 4H), 2.52 (t, 4H, J=7.8 Hz), 4.16 (q, 4H, J=4H); 19F NMR (CDCl3) δ −25.78-25.42(m); 13C NMR (CDCl3) δ 14.16, 27.09, 27.16, 27.22, 31.52, 31.85, 32.19, 60.80, 120.18, 123.38, 126.58, 172.26; ES-MS m/z 275.1 (M+Na).
  • 4,4-Difluoro-heptane-1,7-diol
  • Figure US20070060591A1-20070315-C00010
  • To a solution of 4,4-difluoro-heptanedioic acid diethyl ester (1.3 g, 5.65 mmol) in diethyl ether (35 ml) at 0° C. under N2 was slowly added solid LAH (638 mg, 11.80 mmol). The resulting mixture was allowed to stir for 30 mins at room temperature and then heated to reflux for two hours. Upon cooling, water (0.5 ml) was added, followed by 15% NaOH (0.5 ml) and water (1.5 ml). The resulting mixture was stirred for another two hours at room temperature and then diluted with ethyl acetate (500 ml). The organic solution was dried over Na2SO4 without aqueous work-up. Evaporation of the solvent gave the title compound (646 g, 68%) as pure colorless oil. 1H NMR (CDCl3) δ 1.71-1.80 (m, 4H), 1.87-2.03 (m, 4H), 3.69 (t, 4H, J=6.0 Hz); 19F NMR (CDCl3) 8-22.64-22.28(m); 13C NMR (CDCl3) δ 25.81, 25.87, 25.92, 32.99, 33.33, 33.67, 122.38, 125.56, 128.74.
  • 1,7-bis(Toluene-4-sulfonic acid)-4,4-difluoro-heptyl-ester
  • Figure US20070060591A1-20070315-C00011
  • To a pre-cooled (ice bath) solution of 4,4-difluoro-heptane-1,7-diol (450 mg, 2.67 mmol) and triethylamine (1.2 ml, 8.31 mmol) in CH2Cl2 (5 ml) was added a solution of p-toluensulfonyl chloride (1.13 g, 5.94 mmol) in CH2Cl2 (2 ml). The resulting mixture was allowed to stir for 18 hours at room temperature and then diluted with ethyl acetate (200 ml). The organic solution was washed with sat. NaHCO3, then brine and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel, using 20% ethyl acetate in hexanes, gave the title compound (1.1 g, 83%) as a colorless oil. 1H NMR (CDCl3) δ 1.75-1.90 (m, 8H), 2.44 (s, 6H), 4.01-4.05 (m, 4H), 7.33 (d, 4H, J=8.1 Hz), 7.75 (d, 4H, J=8.1 Hz); 19F NMR (CDCl3) δ −24.27 (t); 13C NMR (CDCl3) δ 22.06, 22.22, 22.28, 22.34, 32.80, 33.14, 33.48, 33.71, 69.96, 70.07, 121.06, 124.26, 127.46, 128.27, 130.36, 133.18, 145.42. ES-MS m/z 477.1 (M+H).
  • [11,11-Difluoro-1, 7-bis(2-nitro-benzenesulfonyl)-1,4,7-triazacyclotetradec-4-yl]-phosphonic acid diethyl ester
  • Figure US20070060591A1-20070315-C00012
  • To a stirred solution of bis-[2-(2-nitrobenzenesulfonylamino)-ethyl]-phosphoramidic acid diethyl ester (1.5 g, 2.46 mmol) and anhydrous Cs2CO3 (2.2 g, 6.74 mmol) in anhydrous DMF (150 ml) at 80° C. under N2 was added a solution of 1,7-bis(toluene-4-sulfonic acid)-4,4-difluoro-heptyl-ester (1.07 mg, 2.25 mmol) in DMF (10 ml) over 10 hours. The reaction mixture was allowed to stir at 85° C. for a further 30 hours, cooled to room temperature and then concentrated. The residue was diluted with 200-ml ethyl acetate and washed with a sat. NaHCO3, then brines and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel using 50% ethyl acetate in CH2Cl2 gave the title compound (311 mg, 19%) asa light yellow solid. 1H NMR (CDCl3) δ 1.32 (t, 6H, J=7.1 Hz), 1.72-1.79 (m, 4H), 1.95-2.05 (m, 4H), 3.30 (s, 4H), 3.32 (s, 4H), 3.40 (t, 4H, J=6.15), 3.96-4.08 (m, 4H), 7.60-7.66 (m, 2H), 7.69-7.76 (m, 4H), 7.99-8.05 (m, 2H); 19F NMR (CDCl3) δ −14.87-14.61 (m); 13C NMR (CDCl3) δ 16.54, 16.63, 22.52, 32.07, 32.42, 32.77, 47.16, 47.89, 47.95, 50.17, 63.19, 63.27, 122.42, 124.65, 124.87, 125.67, 128.87, 131.55, 131.60, 132.16, 134.44, 148.72; ES-MS m/z 742.2 (M+H).
  • 11,11-Difluoro-1,7-bis-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane
  • Figure US20070060591A1-20070315-C00013
  • Acetic acid (10 ml), saturated with anhydrous hydrogen bromide (gas) was added to [11,11-difluoro-1,7-bis(2-nitro-benzenesulfonyl)-1,4,7-triaza-cyclotetradec-4-yl]-phosphonic acid diethyl ester (311 mg, 0.41 mmol) contained in round-bottomed flask closed by glass stopper. The resulting mixture was allowed to stir for 18 hours at room temperature and then diethyl ether (50 ml) was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the diethyl ether solution was decanted off. This white solid was re-dissolved in methanol, stirred with K2CO3 (solid) for 30 mins and the mixture was diluted with 100-ml ethyl acetate. The solids were removed by filtration and the filtrates were evaporated to give the title compound (239 mg, 94%) as white form. 1H NMR (CDCl3) δ 1.82-1.91 (m, 4H), 1.99-2.15 (m, 4H), 2.91 (t, 4H, J=5.1 Hz), 3.30-3.38 (m, 8H), 7.60-7.63 (m, 2H), 7.69-7.75 (m, 4H), 7.90-7.94 (m, 2H); 13C NMR (CDCl3) δ 23.25, 32.22, 32.57, 32.90, 50.22, 50.86, 50.96, 123.50, 124.60, 126.71, 129.89, 130.73, 132.02, 132.22, 134.27, 148.94; 19F NMR (CDCl3) 6-14.12 (t); ES-MS m/z 606.3 (M+H).
    Figure US20070060591A1-20070315-C00014
  • To a stirred solution of 11,11-difluoro-1,7-bis-(2-nitrobenzenesulfonyl)-1,4,7ˆt-traiazacyclotetradecane (239 mg, 0.39 mmol) and anhydrous K2CO3 (320 mg, 2.30 mmol) in anhydrous CH3CN (4 ml) under N2 was added N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (Bridger et al. U.S. Ser. No. 09/111,895) (512 mg, 1.18 mmol). The reaction mixture was allowed to stir at 85° C. for further 18 hours and then concentrated. The residue was diluted with 100-ml ethyl acetate and washed with a sat. NaHCO3, then brines and dried over Na2SO4. Evaporation of the solvent and purification of the residue by column chromatography on silica gel using 50% ethyl acetate in CHCl2 gave the title compound (289 mg, 73%) as white foam. 1H NMR (CDCl3) δ 1.73-1.77 (m, 4H), 1.89-1.99 (m, 4H), 2.75 (t, 4H, J=6.8 Hz), 3.24 (t, 4H, J=7.5 Hz), 3.34 (t, 4H, J=6.3 Hz), 3.58 (s, 2H), 4.56 (s, 2H), 4.59 (s, 2H), 7.08-7.18 (m, 6H), 7.52-7.75 (m, 10H), 7.83 (dd, 2H, J=1.5, 7.8 Hz), 7.99 (d, 1H, J=7.8 Hz), 8.40 (d, 1H); 13C NMR (CDCl3) δ 22.43, 32.32, 46.84, 48.99, 51.71, 52.36, 54.65, 59.30, 122.90, 122.96, 124.57, 124.61, 128.93, 129.76, 130.35, 131.12, 131.49, 132.12, 132.24, 133.83, 134.14, 134.29, 134.45, 134.84, 137.06, 138.30, 148.33, 148.63, 149.68, 155.96; 19F NMR (CDCl3) δ −14.52 (t); ES-MS m/z 1001.3 (M+H).
  • N-[4-(11,11-difluoro-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8880)
  • Figure US20070060591A1-20070315-C00015
  • To a stirred solution of the intermediate from above (289 mg, 0.29 mmol) and anhydrous K2CO3 (478 mg, 3.46 mmol) in anhydrous DMF (4 ml) under N2 was added dropwise, thiophenol (222 mg, 2.02 mmol). The reaction mixture was allowed to stir at room temperature for a further 4 hours and then concentrated. The residue was diluted with 100-ml ethyl acetate and solids were removed by filtration through a short column of celite. Evaporation of the solvent and purification of the residue on a chromatron using silica gel (1 mm plate) eluted with 1:1:98 Methanol/NH4OH/CH2Cl2 gave AMD8880 (80 mg, 62%) as a light yellow oil. 1H NMR (CDCl3) δ 1.63-1.72 (m, 4H), 2.00-2.15 (m, 4H), 2.60-2.65 (m, 12H), 3.57 (s, 2H), 3.83 (s, 2H), 3.93 (s, 2H), 7.14-7.18 (m, 1H), 7.20-7.34 (m, 5H), 7.60-7.67 (m, 1H), 8.55 (d, 1H, J=4.8 Hz); 13C NMR (CDCl3) δ 23.47, 32.64, 32.98, 33.32, 47.69, 48.66, 53.61, 54.21, 54.94, 59.67, 122.32, 122.71, 127.03, 128.59, 129.28, 136.81, 138.49, 139.36, 149.71, 160.16; 19F NMR (CDCl3) δ −15.50 (q, J=12.65 Hz); ES-MS m/z 446.4 (M+H); Anal. Calcd. for (C25H37F2N5): C, 67.39; H, 8.37; N, 15.72. Found: C, 67.52; H, 8.49; N, 15.43.
  • EXAMPLE 3 Preparation of N-[4-(1,4,7-triazacyclotetradecan-2-one)-yl))-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt). (AMD8748, FIG. 3)
  • Figure US20070060591A1-20070315-C00016
  • Nosylaziridine 11.7 g (51.3 mmol) in anhydrous THF (80 ml) was added dropwise to a stirred solution of 1,7-diaminoheptane 33.4 g (256.3 mmol) in anhydrous THF (220 ml). After completion of addition, the mixture was stirred for 20 min at room temperature under nitrogen atmosphere. The solution was concentrated in vacuo to afford crude product as a yellow oil. The oil was purified by flash chromatography on silica gel eluting with 5% MeOH, 5% NH4OH, 90% CH2Cl2 to give the desired amine as a yellow oil (10.9 g, 59%). 1H NMR (CDCl3, 300 MHz) δ 1.27-1.50 (m, 10H), 2.09 (s, 4H), 2.47 (t, J=6.0 Hz, 2H), 2.68 (t, J=7.5 Hz, 2H), 2.75 (t, J=6.0 Hz, 2H), 3.13 (t, J=6.0 Hz, 2H), 7.72-7.75 (m, 2H), 7.75-7.87 (m, 1H), 8.13-8.16 (m, 1H); exact mass calculated for C15H26N4O4S: 358, found: m/z 359 [M+H]+.
    Figure US20070060591A1-20070315-C00017
  • To a stirred solution of the amine from above (7.0 g, 19.5 mmol) and 4.3 ml (29.3 mmol) of Et3N in THF (300 ml) at −78° C. was added dropwise 4.3 g (19.5 mmol) of Boc2O in THF (100 ml). After 1 h the reaction mixture was diluted with EtOAc, and washed with aqueous NaHCO3, dried (MgSO4), and concentrated in vacuo. Purification by flash chromatography on silica gel eluting with 5% MeOH, 95% CH2Cl2 gave 5.6 g (63%) of the desired BOC intermediate as a pale yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.26-1.27 (m, 10H), 1.36-1.44 (m, 9H), 2.47 (t, J=7.5 Hz, 2H), 2.75 (t, J=6.0 Hz, 2H), 3.00 (br s, 2H), 3.06-3.16 (m, 4H), 4.50 (br s, 1H), 7.72-7.76 (m, 2H), 7.86-7.89 (m, 1H), 8.13-8.16 (m, 1H); exact mass calculated for C20H34N4O6S: 458, found: m/z 459 [M+H]+.
    Figure US20070060591A1-20070315-C00018
  • To a stirred solution of the BOC intermediate from above (1.6 g, 3.49 mmol) and 1.5 ml (10.5 mmol) of Et3N in 100 ml of anhydrous CH2Cl2 was added 504 ul (19.5 mmol) of DepCl, and the reaction mixture was stirred under a nitrogen atmosphere for 24 h. The solvent was removed under reduced pressure and the residue was dissolved in EtOAc, washed with aqueous NaHCO3, dried (Na2SO4), and concentrated in vacuo. Purification by flash chromatography on silica gel, eluting with 5% MeOH, 95% CH2Cl2 gave 1.4 g (67%) of the Dep intermediate as a pale yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.23-1.34 (m, 16H), 1.44 (s, 9H), 2.88-2.94 (m, 2H), 3.08-3.11 (m, 2H), 3.22 (d, J=6.0 Hz, 4H), 4.00-4.09 (m, 4H), 4.50 (br s, 1H), δ 11 (br s, 1H), 7.72-7.75 (m, 2H), 7.83-7.84 (m, 1H), 8.10-8.13 (m, 1H); exact mass calculated for C24H43N4O9SP: 594, found: m/z 595 [M+H]+.
    Figure US20070060591A1-20070315-C00019
  • To a stirred solution of the Dep intermediate from above (1.0 g, 1.74 mmol) in 30 ml of anhydrous CH2Cl2 was added 10 ml of TFA, and the reaction mixture was stirred under a nitrogen atmosphere for 40 min at room temperature. TFA and the solvent were removed under reduced pressure. The residue was dissolved with MeOH. K2CO3 was added to the solution, and the mixture was stirred for 10 min at room temperature. The mixture was diluted with 50 ml of CH2Cl2, filtered through celite, and concentrated in vacuo. Purification by flash chromatography on silica gel eluting with 5% MeOH, 5% NH4OH, 90% CH2Cl2 gave 731 mg (85%) of the desired amine as a yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.28-1.33 (m, 12H), 1.42-1.46 (m, 4H), 2.60-2.67 (m, 5H), 2.91 (q, J=8.7 Hz, 2H), 3.20-3.22 (m, 4H), 4.03 (q, J=7.5 Hz, 4H), 7.70-7.73 (m, 2H), 7.81-7.82 (m, 1H), 8.09-8.12 (m, 1H); exact mass calculated for C19H35N4O7SP: 494, found: m/z 495 [M+H]+.
    Figure US20070060591A1-20070315-C00020
  • To a stirred solution of the amine from above (731 mg, 1.48 mmol) in 31 ml of anhydrous THF was added 1.6 g (14.7 mmol) of Na2CO3, and the mixture was cooled to −15° C. Bromoacetyl bromide 155 ul (1.78 mmol) in 1.6 ml of THF was added to the mixture dropwise. After 1 h, a second portion 51 ul (0.59 mmol) of bromoacetyl bromide was added to the solution. The mixture was stirred at −15° C. under a nitrogen atmosphere for a further 30 min. The reaction mixture was diluted with 50 ml of CH2Cl2, filtered through celite, and concentrated in vacuo. Purification of the residue by flash chromatography on silica gel eluting with 5% MeOH, 95% CH2Cl2 gave 807 mg (89%) of the desired amide as a yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.25-1.34 (m, 14H), 1.48-1.53 (m, 2H), 2.94 (q, J=3 Hz, 2H), 3.21-3.31 (m, 6H), 3.88 (s, 2H), 4.01-4.09 (m, 4H), 6.14 (br s, 1H), 6.58 (br s, 1H), 7.72-7.75 (m, 2H), 7.83-7.84 (m, 1H), 8.10-8.13 (m, 1H); exact mass calculated for C21H36N4O8SPBr: 616, found: m/z 617 [M+H]+.
    Figure US20070060591A1-20070315-C00021
  • To a stirred solution of the amide from above (369 mg, 0.60 mmol) in 1000 ml of anhydrous acetonitrile was added 417 mg (3.1 mmol) of K2CO3, and the mixture was stirred at 60° C. under a nitrogen atmosphere for 24 h. The solution was concentrated under reduced pressure. Purification by flash chromatography on silica gel eluting with 5% MeOH, 5% NH4OH, 90% CH2Cl2 gave 290 mg (90%) of the desired macrocycle as a yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.28 (t, J=7.5 Hz, 6H), 1.30-1.37 (m, 10H), 2.94-2.95 (m, 2H), 3.27-3.45 (m, 6H), 3.96-4.04 (m, 4H), 4.05 (s, 2H), 6.58 (t, J=6.0 Hz, 1H), 7.66-7.68 (m, 1H), 7.73-7.76 (m, 2H), 8.16-8.19 (m, 1H); exact mass calculated for C21H35N4O8SP: 534, found: m/z 535 [M+H]+.
    Figure US20070060591A1-20070315-C00022
  • To a stirred solution of the macrocycle from above (290 mg, 0.54 mmol) in 5 ml of anhydrous DMF was added 374 mg (2.71 mmol) of K2CO3, and 167 μl (1.63 mmol) of thiophenol. The mixture was stirred at room temperature under a nitrogen atmosphere for 4 h. The solution was concentrated under reduced pressure. The residue was dissolved in CH2Cl2, filtered through celite, and concentrated in vacuo. Purification by flash chromatography on silica gel eluting with 3% MeOH, 5% NH4OH, 92% CH2Cl2 gave 145 mg (77%) of the macrocyclic amine as a yellow solid. 1H NMR (CDCl3, 300 MHz) δ 1.31 (t, J=7.5 Hz, 6H), 1.33-1.59 (m, 10H), 2.70 (t, J=7.5 Hz, 2H), 2.96-2.99 (m, 2H), 3.20-3.34 (m, 5H), 3.31 (s, 2H), 3.96-4.05 (m, 4H), 7.40 (br s, 1H), exact mass calculated for C15H32N3O4P: 349, found: m/z 350 [M+H]+.
    Figure US20070060591A1-20070315-C00023
  • To a stirred solution of the macrocyclic amine from above (152 mg, 0.44 mmol) in 4 ml of anhydrous acetonitrile was added 180 mg (1.31 mmol) of K2CO3, and 237 mg (0.61 mmol) of N-[1-methylene-4-(chloromethylene)phenylene]-N-(diethoxyphosphoryl)-2-(aminomethyl)pyridine (Bridger et al. U.S. Ser. No. 09/111,895). The mixture was stirred at 83° C. under a nitrogen atmosphere for 18 h. The solution was concentrated under reduced pressure. The residue was dissolved with CH2Cl2, filtered through celite, and concentrated in vacuo to afford crude product as yellow oil. Purification by flash chromatography on silica gel eluting with 3% MeOH, 97% CH2Cl2 gave 257 mg (85%) of the desired intermediate as a yellow foam. 1H NMR (CDCl3) δ 1.25-1.30 (m, 12H), 1.31-1.61 (m, 10H), 2.50-2.55 (m, 2H), 2.80-2.94 (m, 2H), 2.97 (s, 2H), 3.25-3.50 (m, 5H), 3.97 (s, 2H), 4.02-4.12 (m, 8H), 4.21 (dd, J=16.5 Hz, J=10.5 Hz, 4H), 7.04-7.26 (m, 5H), 7.36 (d, J=6.0 Hz, 1H), 7.62 (t, J=3 Hz, 1H), 8.50 (d, J=3.0 Hz, 1H); exact mass calculated for C33H55N5O7P2: 695, found: m/z 696 [M+H]+.
    Figure US20070060591A1-20070315-C00024
  • N-[4-(1,4,7-triazacyclotetradecan-2-one)-yl))-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD8748)
  • The intermediate from above (25 7 mg, 0.37 mmol) was dissolved in 3 ml of HBr/AcOH and the solution was stirred at room temperature for 24 h. Diethyl ether was added to the mixture resulting in the formation of a yellow precipitate. The solid was collected by filtration, washed with diethyl ether and dried in vacuo to give AMD8748 as a white powder (182 mg, 60%). 1H NMR (D2O, 300 MHz) δ 1.38-1.74 (m, 10H), 3.15 (t, J=7.5 Hz, 2H), 3.26-3.28 (m, 2H), 3.45-3.59 (m, 4H), 3.84 (s, 2H), 4.47 (s, 2H), 4.58 (s, 2H), 7.62-7.76 (m, 5H), 7.79 (d, J=8.7 Hz, 1H), 8.23 (t, J=6.0 Hz, 1H), 8.73 (d, J=5.1 Hz, 1H); 13C NMR (D2O, 75.5 MHz) δ 167.59, 148.23, 147.32, 142.85, 132.44, 132.21, 131.34, 126.44, 126.39, 59.72, 54.77, 51.20, 50.19, 49.45, 45.49, 40.38, 39.05, 26.89, 24.85, 23.37, 23.16, 21.66; exact mass calculated for C25H37N5O: 423, found: m/z 424 [M+H]+; anal calculated for (C25H37N5O) 2.5 (H2O) 3.9 (HBr) 0.4 (C4H10O): C, 39.26; H, 6.18, N, 8.61; Br, 38.29, found: C, 39.15; H, 5.99, N, 8.53; Br, 38.45.
  • EXAMPLE 4 Preparation of N-[12-(5-oxa-1,9-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt). (AMD8922 FIG. 4)
  • Figure US20070060591A1-20070315-C00025
  • Preparation of 2-(4-cyano-benzyl)-malonic acid dimethyl ester
  • Figure US20070060591A1-20070315-C00026
  • Dimethyl malonate (10.00 g, 75.7 nmol) was added to a suspension of NaH (60% in oil, 3.33 g, 83.3 mmol) in THF (100 mL) over 20 minutes at room temperature. The mixture was heated at 80° C. for 15 minutes, then a solution of α-bromotolunitrile (14.84 g, 75.69 mmol) in THF (100 mL) was added. Heating was continued at 80° C. for 1 hour, then at 50° C. for 64 hours. Water (30 mL) was added, and the aqueous phase was extracted with EtOAc (3×20 mL). The combined organic phases were dried (MgSO4) and concentrated. Purification of the crude material on silica gel (25% EtOAc/hexanes) gave the title compound as colourless crystals (8.49 g, 45%). 1H NMR (CDCl3) δ 3.27 (d, 2H, J=9.0 Hz), 3.67 (t, 1H, J=9.0 Hz), 3.70 (s, 6H), 7.32 (d, 2H, J=9.0 Hz), 7.58 (d, 2H, J=9.0 Hz).
  • N-[4-(3-Hydroxy-2-hydroxymethyl-propyl)-benzyl]-2-nitro-N-pyridin-2-ylmethyl-benzenesulfonamide
  • Figure US20070060591A1-20070315-C00027
  • To a solution of 2-(4-cyano-benzyl)-malonic acid dimethyl ester (8.47 g, 34.3 mmol) in THF (30 mL) at 0° C. was added LiAlH4 (1.0 M in THF, 206 mL, 206 mmol), and the mixture was heated at reflux for 19.5 hours. The mixture was cooled to 0° C., and H2O (8 mL) was added dropwise followed by 15% NaOH(aq) (8 mL) and H2O (24 mL). The mixture was stirred at room temperature for 45 minutes, then dried (MgSO4) and filtered. The filtrate was concentrated to give a yellow oil (5.16 g).
  • A solution of the crude diol (5.13 g) in CH2Cl2 (105 mL) was stirred at 0° C. while Et3N (4.00 mL, 28.7 mmol) was added followed by 2-nitrobenzenesulfonyl chloride (5.90 g, 26.6 mmol). The solution was stirred at room temperature for 17 hours, then washed with brine (70 mL). The aqueous phase was extracted with CH2Cl2 (3×20 mL). The combined organic phases were dried (MgSO4) and concentrated. Purification of the crude material on silica gel (80% THF/hexanes) gave a yellow solid (3.06 g).
  • The protected diol (2.87 g), 2-picolyl chloride hydrochloride (1.36 g, 8.29 mmol), K2CO3 (3.13 g, 22.6 mmol), and KBr (90 mg, 0.76 mmol) were heated at reflux in acetonitrile (40 mL) for 17.5 hours. Water (30 mL) was added, and the mixture was extracted with EtOAc (100 mL). The organic extract was washed with brine (20 mL), and the combined aqueous phases were extracted with EtOAc (3×20 mL). The combined organic extracts were dried (MgSO4) and concentrated. Purification of the crude material on silica gel (10:10:1 CH2Cl2/EtOAc/MeOH) gave the title compound as a yellow oil (1.83 g, 9% over 3 steps). 1H NMR (CDCl3) δ 2.00 (m, 1H), 2.56 (d, 2H, J=7.5 Hz), 3.64 (m, 2H), 3.78 (m, 2H), 4.57 (s, 2H), 4.60 (s, 2H), 7.02-7.14 (m, 5H), 7.23 (d, 1H, J=7.8 Hz), 7.56 (m, 2H), 7.67 (d, 2H, J=3.9 Hz), 7.97 (d, 1H, J=7.8 Hz), 8.41 (d, 1H, J=4.8 Hz).
  • N-[4-(3-Cyano-2-cyanomethyl-propyl)-benzyl]-2-nitro-N-pyridin-2-ylmethyl-benzenesulfonamide
  • Figure US20070060591A1-20070315-C00028
  • To a solution of N-[4-(3-hydroxy-2-hydroxymethyl-propyl)-benzyl]-2-nitro-N-pyridin-2-ylmethyl-benzenesulfonamide (1.80 g, 3.82 mmol) and Et3N (1.30 mL, 9.33 mmol) in CH2Cl2 (30 mL) at 0° C. was added MsCl (0.65 mL, 8.4 mmol). The solution was stirred at room temperature for 30 minutes, then concentrated. EtOAc (50 mL) was added, and the mixture was washed with H2O (20 mL), saturated NaHCO3(aq) (2×15 mL), and brine (15 mL) then dried (MgSO4) and concentrated to give a yellow oil (2.36 g).
  • A mixture of the crude mesylate (2.36 g), cetyltrimethylammonium bromide (137 mg, 0.376 mmol), and NaCN (1.3 g, 27 mmol) in 2:1 benzene/H2O (30 mL) was heated at reflux for 18 hours. The mixture was diluted with EtOAc (100 mL) and washed with H2O (20 mL), saturated NaHCO3(aq) (2×20 mL), and brine (10 mL) then dried (MgSO4) and concentrated. Purification of the crude material on silica gel (80% EtOAc/hexanes) gave the title compound as a yellow oil (824 mg, 44% over 2 steps). 1H NMR (CDCl3) δ 2.31-2.55 (m, 5H), 2.79 (d, 2H, J=6.0 Hz), 4.59 (s, 2H), 4.60 (s, 2H), 7.04-7.25 (m, 6H), 7.51-7.64 (m, 2H), 7.69 (m, 2H), 8.03 (d, 1H, J=7.8 Hz), 8.41 (d, 1H, J=4.8 Hz).
  • Methanesulfonic acid 5-methanesulfonyloxy-3-(4-{[(2-nitro-benzenesulfonyl)-pyridin-2-ylmethyl-amino]-methyl}-benzyl)-pentyl ester
  • Figure US20070060591A1-20070315-C00029
  • A solution of N-[4-(3-cyano-2-cyanomethyl-propyl)-benzyl]-2-nitro-N-pyridin-2-ylmethyl-benzenesulfonamide (2.36 g, 4.82 mmol) in 4:1 concentrated HCl(aq)/AcOH (25 mL) was heated at reflux for 17 hours. Water (80 mL) was added, and the mixture was extracted with EtOAc (5×25 mL). The organic phase was extracted with saturated NaHCO3(aq) (5×20 mL) and H2O (2×20 mL). The combined aqueous extracts were acidified (pH 1-2) with concentrated HCl(aq) and extracted with EtOAc (5×25 mL). The combined organic extracts were dried (MgSO4) and concentrated to give a brown foam (1.25 g).
  • To a solution of the crude diacid (1.25 g) in THF (20 mL) was added BH3.Me2S (10 M in BH3, 2.9 mL, 29 mmol). The mixture was heated at reflux for 30 minutes. Methanol (30 mL) was added, and the solution was concentrated. The residue was dissolved in MeOH (30 mL) and the solution was concentrated (repeated) to give a yellow foam (1.04 g).
  • To a solution of the crude diol (1.00 g) and Et3N (1.4 mL, 10 mmol) in CH2Cl2 (20 mL) at 0° C. was added MsCl (0.70 mL, 9.0 mmol). The mixture was stirred at room temperature for 20 minutes, then concentrated. The residue was partitioned between EtOAc (30 mL) and saturated NaHCO3(aq) (15 mL). The organic phase was washed with saturated NaHCO3(aq) (15 mL) and brine (5 mL), then dried (MgSO4) and concentrated. Purification of the crude material on silica gel (10:10:1 CH2Cl2/EtOAc/MeOH) gave the title compound as a yellow oil (450 mg, 15% over 3 steps). 1H NMR (CDCl3) δ 1.75 (m, 4H), 2.01 (m, 1H), 2.59 (d, 2H, J=7.2 Hz), 3.00 (s, 6H), 4.23 (m, 4H), 4.58 (s, 2H), 4.59 (s, 2H), 7.02 (d, 2H, J=8.1 Hz), 7.11 (m, 3H), 7.25 (d, 1H, J=8.4 Hz), 7.53-7.61 (m, 2H), 7.69 (m, 2H), 8.00 (m, 1H), 8.42 (m, 1H).
    Figure US20070060591A1-20070315-C00030
  • To a solution of bis(3-aminopropyl)ether (1.40 mL, 10 mmol) in dichloromethane (50 mL) was added triethylamine (4.2 mL, 30 mmol) and 2-nitrobenzenesulfonyl chloride (5.42 g, 24 mmol). The resultant solution was stirred at room temperature for four hours. Aqueous ammonium chloride (50 mL) was then added, the organic and aqueous layers were separated and the aqueous layer was extracted twice with dichloromethane. The combined organic layers were then dried and concentrated, and the resultant viscous yellow oil was chromatographed over silica gel using a 5% methanol solution in dichloromethane as an eluant to afford the desired product, bis(3-nitrobenzenesulfonylaminopropyl)ether as a pale yellow oil in a yield of 3.3 g (66%). 1H NMR (CDCl3) δ 1.79 (qi, 4H, J=7.1 Hz), 3.16 (t, 4H, J=7.0 Hz), 3.47 (t, 4H, J=7.1 Hz), 5.82 (br s, 2H), 7.71 (m, 8H), 7.83 (d, 2H, J=6.6 Hz), 8.11 (d, 2H, J=6.6 Hz).
    Figure US20070060591A1-20070315-C00031
  • A solution of bis(3-nitrobenzenesulfonylaminopropyl)ether (190 mg, 0.38 mmol) and finely ground, freshly oven-dried cesium carbonate (320 mg, 1.14 mmol) in anhydrous DMF (60 mL) was heated to 80° C. Methanesulfonic acid, 5-methanesulfonyloxy-3-(4-{[(2-nitro-benzenesulfonyl)-pyridin-2-ylmethyl-amino]-methyl}-benzyl)-pentyl ester (250 mg, 0.38 mmol) in DMF (20 mL) was then added to the reaction over a 30 hour period via syringe pump. After complete addition, the reaction was stirred another 30 hours. The mixture was then cooled, diluted with 300 mL ethyl acetate and extracted repeatedly with water. The organic phase was then dried and concentrated, and the residue was purified by column chromatography on silica gel using 25% ethyl acetate in dichloromethane as eluant to afford the desired product (203 mg, 51%). 1H NMR (CDCl3) δ 1.47-1.71 (m, 9H), 2.42 (d, 2H, J=6.1 Hz), 3.15-3.32 (m, 12H), 4.53 (s, 2H), 4.55 (s, 2H), 6.94 (d, 2H, J=6.4 Hz), 7.03 (d, 2H, J=6.4 Hz), 7.05 (d, 1H, J=5.8 Hz), 7.52 (m, 4H), 7.64 (m, 6H), 7.91 (m, 5H), 8.44 (d, 1H, J=5.4 Hz).
    Figure US20070060591A1-20070315-C00032
  • A solution of the intermediate from above (203 mg, 0.203 mmol) in acetonitrile (8 mL) was treated with potassium carbonate (450 mg, 3.05 mmol) and thiophenol (0.25 mL, 2.44 mmol) and the mixture was stirred overnight at room temperature. Following standard work-up, the crude mixture was purified by chromatography on silica gel (85:12:3 dichloromethane:methanol:ammonium hydroxide), the desired product (47 mg, 57%) as a white foam. 1H NMR (CDCl3) δ 1.44-1.89 (m, 8H), 1.93 (m, 3H), 2.51 (m, 4H), 2.68 (m, 2H), 2.72 (m, 2H), 3.10 (br s, 2H(NH)), 3.54 (m, 4H), 3.81 (s, 2H), 3.91 (s, 2H), 7.09 (d, 2H, J=6.1 Hz), 7.13 (m, 1H), 7.29 (d, 2H, J=6.1 Hz), 7.32 (m, 1H) 7.64 (t, 1H, J=5.8 Hz), 8.54 (d, 1H, J=5.4 Hz).
  • The white foam was then converted to the corresponding hydrobromide salt to give AMD8922 (42 mg). 1H NMR (D2O) δ 1.73 (m, 4H), 2.00 (m, 5H), 2.65 (d, 2H, J=7.2 Hz); 3.04-3.19 (m, 8H), 3.51 (br s, 4H), 4.38 (s, 2H), 4.58 (s, 2H), 7.32 (d, 2H, J=7.5 Hz), 7.43 (d, 2H, J=7.5 Hz), 7.85 (t, 1H, J=6.9 Hz), 7.89 (d, 1H, J=8.4 Hz), 8.34 (d, 1H, J=7.5 Hz), 8.74 (d, 1H, J=5.1 Hz). 13C NMR (CDCl3) δ 24.01, 26.30, 34.51, 39.35, 41.88, 44.74, 47.47, 51.58, 69.66, 127.05, 127.16, 128.44, 130.47 (2C), 130.72 (2C), 141.74, 144.62, 146.00, 147.25. ES-MS m/z 411 (M+H); Anal. Calcd. for (C25H38N4O×4.0 HBr×3.5H2O): C,37.66; H, 6.19; N, 7.03; Br 40.09. Found: C, 37.67; H, 6.06; N, 6.92; Br, 40.24.
  • EXAMPLE 5 Preparation of N-[4-(1-oxa-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt). (AMD8779, FIG. 5)
  • Figure US20070060591A1-20070315-C00033
  • To a solution of bis(3-aminopropyl)ether (513 mg, 3.88 mmol) and triethylamine (1.7 ml, 11.78 mmol) in CH2Cl2 (15 ml) was added 2-nitrobenzenesulfonylchloride (97% pure, 1.95 g, 8.53 mmol) in CH2Cl2 (5 ml). Standard work-up followed by purification of the crude product by flash column chromatography on silica gel (10:90 EtOAc/CH2Cl2), gave the desired product (1.80 g, 92%) as a yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.79-1.87 (tt, J=6.0, 6.0 Hz, 4H), 3.20-3.26 (dt, J=6.0, 6.0 Hz, 4H), 3.49-3.53 (t, J=6.0 Hz, 4H), 5.75-5.79 (t, J=6.0 Hz, 2H), 7.29-7.76 (m, 4H), 7.86-7.88 (m, 2H), 8.13-8.16 (m, 2H).
    Figure US20070060591A1-20070315-C00034
  • The desired macrocycle was prepared using standard macrocyclization conditions; Bridger et al. J. Med. Chem. 1995, 38, 366-378): To a stirred solution of the intermediate from above (1.8 g, 3.59 mmol) in DMF (180 ml) containing anhydrous Cs2CO3 (3.6 g, 11.05 mmol) heated to 80° C. was added dropwise, a solution of N-(diethoxyphosphoryl)-O,O′-bis(2-methylsulfonyl)diethanolamine (Bridger, et al. J. Med. Chem. (1995) 38, 366-378) (1.9 g, 4.78 mmol) in DMF (20 ml). Evaporation of the solvent and purification of the residue by column chromatography on silica gel (30:70 ethyl acetate/CH2Cl2) gave the desired macrocycle (1.25 g, 55%) as a light yellow foam. 1H NMR (CDCl3, 300 MHz) δ 1.25-1.35 (m, 6H), 1.87-1.91 (m, 4H), 3.05-3.14 (m, 4H), 3.41-3.48 (m, 12H), 3.96-4.07 (m, 4H), 7.61-7.63 (m, 2H), 7.68-7.71 (m, 4H), 8.05-8.09 (m, 2H); 13C (CDCl3, 75.5 MHz) δ 16.51, 16.60, 30.63, 44.68, 44.74, 44.92, 47.24, 63.07, 63.14, 67.69, 124.52, 131.34, 132.26, 133.58, 134.11 148.32; exact mass m/z calcd. for C26H38N5O12PS2 707.17, found [M+H]+708.5.
    Figure US20070060591A1-20070315-C00035
  • A saturated solution of anhydrous hydrogen bromide (gas) in acetic acid (8 mL) was added to the macrocycle from above (1.25 g, 1.77 mmol) contained in round bottomed flask closed by glass stopper. The resulting solution was allowed to stir overnight at room temperature and 50 ml of diethyl ether was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the supernatant solution was decanted off. This white solid (HBr salt) was dissolved in 30 ml methanol and stirred with excess K2CO3 for 30 min and then concentrated. The residue was diluted with 300 ml ethyl acetate and the solids were filtered off by passing though a short celite column. Evaporation of the solvent and purification of the residue by column chromatography on silica gel (3:3:94 Methanol/NH4OH/CH2Cl2) gave the desired intermediate (736 mg, 74%) as a light yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.87-1.91 (b, 4H), 2.85-2.88 (b, 4H), 3.39-3.91 (t, J=6.0 Hz, 4H), 3.49-3.56 (m, 8H), 7.59-7.70 (m, 6H), 7.98-8.01 (m, 2H); 13C (CDCl3, 75.5 MHz) δ 29.61, 47.90, 48.71, 68.36, 124.37, 131.07, 132.17, 133.45, 133.77, 148.65; exact mass m/z calcd. for C22H29N5O9S2 571.14, found [M+H]+572.20.
    Figure US20070060591A1-20070315-C00036
  • Reaction of the intermediate from above (736 mg, 1.29 mmol) with N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (Bridger et al. U.S. Ser. No. 09/111,895) (980 mg, 2.27 mmol) and K2CO3 (560 mg, 4.06 mmol) in CH3CN (13 mL) followed by purification of the crude intermediate by column chromatography on silica gel (10:90 ethyl acetate/CH2Cl2) gave desired intermediate (799 mg, 64%) as a light yellow foam. 1H NMR (CDCl3, 300 MHz) δ 1.80-1.84 (b, 4H), 2.58-2.62 (t, J=7.1 Hz, 4H), 3.38-3.42 (m, 8H), 3.50-3.55 (m, 6H), 4.59 (b, 4H), 7.06 (b, 4H), 7.11-7.15 (m, 1H), 7.18-7.21 (d, J=7.8 Hz, 1H), 7.54-7.70 (m, 10H), 7.87-7.89 (d, J=7.8 Hz, 2H), 7.99-8.01 (d, J=7.8 Hz, 1H), 8.41 (d, 1H); 13C (CDCl3, 75.5 MHz) δ 29.93, 44.77, 46.17, 51.28, 52.00, 52.30, 59.46, 67.21, 122.57, 124.11, 128.46, 128.82, 130.48, 131.06, 131.71, 133.38, 133.53, 133.61, 134.07, 136.67, 138.20, 147.93, 149.26, 155.62; exact mass m/z calcd. for C42H46N8O13S3 966.23, found [M+H]+967.20.
  • AMD8779: N-[4-(11-oxa-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt)
  • Figure US20070060591A1-20070315-C00037
  • The intermediate from above (780 mg, 0.81 mmol) was reacted with anhydrous K2CO3 (1.36 g, 9.86 mmol) and thiophenol (644 mg, 5.84 mmol) in anhydrous DMF (8 ml). Purification of the crude material on a chromatron over silica gel (2 mm plate), using 3:3:94 CH3OH/NH4OH/CH2Cl2 gave the free amine (170 mg, 51%) as a light yellow oil. This free amine was re-dissolved in acetic acid (5 ml) saturated with anhydrous hydrogen bromide (gas), the resulting solution was allowed to stir for 30 min. at room temperature and then 50 ml of diethyl ether was added. The white precipitate which formed was allowed to settle to the bottom of the flask and the supernatant solution was decanted off. The white solid was washed by decantation with diethyl ether (5 times), and the remaining traces of solvent were removed by blowing nitrogen though the flask followed by drying in vacuo overnight at 50° C. to give AMD8779 (140 mg, 46%) as a white solid. 1H NMR (D2O, 300 MHz) δ 1.99-2.03 (b, 4H), 2.85-2.88 (b, 4H), 3.25-3.29 (b, 8H), 3.73-3.76 (t, J=5.3 Hz, 4H), 3.82 (s, 2H), 4.41 (s, 2H), 4.55 (s, 2H), 7.39-7.41 (d, J=8.1 Hz, 2H), 7.52-7.55 (d, J=8.1 Hz, 2H), 7.76-7.79 (m, 2H), 8.22-8.24 (m, 1H), 8.68-8.70 (m, 1H); 13C (D2O, 75.5 MHz) δ 25.58, 44.74, 46.92, 49.16, 49.98, 51.33, 55.08, 70.39, 126.57, 130.48, 130.95, 131.41, 137.27, 143.11, 147.01, 147.92; exact mass m/z calcd. for C24H37N5O 411.30, found [M+H]+412.30; Anal. (C24H37N5O.4.1 HBr.4.1H2O)C, 35.28; H, 6.08; N, 8.57; Br, 40.09. Found C, 35.42; H, 6.07; N, 8.50; Br, 39.92.
  • EXAMPLE 6 Preparation of N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt). (AMD8834, FIG. 6)
  • Figure US20070060591A1-20070315-C00038
  • Reaction of 3,3′-thiodipropionitrile (1.81 g, 12.91 mmol) in THF (10 ml) with BH3/Me2S (10 M, 4.5 ml, 45 mmol) followed by work-up with 6 N hydrochloride acid (30 ml) followed by 10 N NaOH (18 ml) and evaporation of the solvent gave the crude diamine (1.4 g, 73%). 1H NMR (CDCl3, 300 MHz) δ 1.18-1.26 (m, 4H), 1.68-1.79 (m, 4H), 2.55-2.60 (t, J=7.5 Hz, 4H), 2.77-2.81 (t, J=6.0 Hz, 4H). This material was used without further purification in the next step.
    Figure US20070060591A1-20070315-C00039
  • Reaction of the diamine from above (1.41 g, 9.51 mmol) with 2-nitrobenzenesulfonylchloride (97% pure, 4.7 g, 20.57 mmol) in CH2Cl2 (40 ml) containing triethylamine (4.2 ml, 29.01 mmol) followed by purification of the crude material by column chromatography on silica gel (10:90 EtOAc/CH2Cl2) gave the desired nosyl derivative (4.5 g, 91%) as a yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.78-1.87 (tt, J=6.8, 6.8 Hz, 4H), 2.52-2.57 (t, J=7.5 Hz, 4H), 3.18-3.25 (dt, J=6.0, 6.0 Hz, 4H), 5.45-5.49 (t, J=6.0 Hz, 2H), 7.75-7.78 (m, 4H), 7.78-7.86 (m, 2H), 8.13-8.16 (m, 2H).
    Figure US20070060591A1-20070315-C00040
  • Macrocyclization was performed as described in Example 5: A solution of the intermediate from above (2.6 g, 5.01 mmol) and anhydrous Cs2CO3 (4.9 g, 15.04 mmol) in DMF (250 ml) was reacted with the requisite dimesylate (2.4 g, 6.04 mmol) in DMF (20 ml). Evaporation of the solvent and purification of the crude product by column chromatography on silica gel (30:70 ethyl acetate/hexanes) gave the desired macrocycle (810 mg, 23%) as a yellow foam. 1H NMR (CDCl3, 300 MHz) δ 1.11-1.33 (m, 6H), 1.92-1.97 (m, 4H), 2.61-2.65 (t, J=6.0 Hz, 4H), 3.20-3.25 (m, 4H), 3.40-3.50 (m, 8H), 3.97-4.05 (m, 4H), 7.63-7.73 (m, 6H), 8.05-8.09 (m, 2H).
    Figure US20070060591A1-20070315-C00041
  • The intermediate from above (810 mg, 1.12 mmol) was dissolved in a saturated solution of anhydrous hydrogen bromide (gas) in acetic acid (10 mL). Evaporation of the solvent gave the crude amine (596 mg, 91%). 1H NMR (CDCl3, 300 MHz) δ 1.96-2.05 (m, 4H), 2.60-2.65 (t, J=7.5 Hz, 4H), 2.89-2.93 (t, J=6.0 Hz, 4H), 3.35-3.38 (t, J=4.5 Hz, 4H), 3.42-3.46 (t, J=6.0 Hz, 4H), 7.63-7.73 (m, 6H), 7.96-7.99 (m, 2H). This material was used directly in the next step.
    Figure US20070060591A1-20070315-C00042
  • The amine from above (596 mg, 1.01 mmol) was reacted with N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (Bridger, et al. U.S. Ser. No. 09/111,895) (770 mg, 1.78 mmol) and K2CO3 (500 mg, 3.62 mmol) in CH3CN (12 mL). Purification of the crude material by chromatography on silica gel (10:90 ethyl acetate/CH2Cl2) gave the desired intermediate (400 mg, 40%) as a light yellow oil. 1H NMR (CDCl3, 300 MHz) δ 1.94-2.05 (m, 4H), 2.58-2.62 (t, J=6.0 Hz, 4H), 2.68-2.73 (t, J=7.5 Hz, 4H), 3.31-3.36 (t, J=7.5 Hz, 4H), 3.43-3.48 (t, J=7.5 Hz, 4H), 3.56 (s, 2H), 4.60 (s, 2H), 4.75 (s, 2H), 7.10-7.20 (m, 7H), 7.58-7.71 (m, 9H), 7.82 (d, 2H), 8.0 (d, 1H), 8.45 (d, 1H).
  • AMD8834: N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt)
  • Figure US20070060591A1-20070315-C00043
  • The intermediate from above (400 mg, 0.41 mmol) was reacted with anhydrous K2CO3 (674 g, 4.88 mmol) and thiophenol (322 mg, 2.92 mmol) in anhydrous DMF (8 ml). Purification of the crude product by flash chromatography on a chromatron over silica gel (2 mm plate), using 3:3:94 CH3OH/NH4OH/CH2Cl2 gave the free amine (107 mg, 61%) as a light yellow oil. This free amine was re-dissolved in acetic acid saturated with anhydrous hydrogen bromide (5 ml) and the resulting solution was allowed to stir for 30 min. at room temperature and 50 ml of diethyl ether was then added. The white precipitate which formed was allowed to settle to the bottom of the flask and the supernatant solution was decanted off. The solid was washed by decantation with diethyl ether (5 times) and the remaining traces of solvent were removed by blowing nitrogen though the flask followed drying in vacuo at 50° C. to give AMD8834 (196 mg, 89%) as a white solid. 1H NMR (D2O, 300 MHz) δ 2.03-2.07 (m, 4H), 2.88 (m, 8H), 3.29-3.32 (m, 4H), 3.35-3.39 (t, J=6.2 Hz, 4H), 3.80 (s, 2H), 4.43 (s, 2H), 4.61-4.62 (d, J=3.0 Hz, 2H), 7.45-7.48 (d, J=7.8 Hz, 2H), 7.52-7.55 (d, J=7.8 Hz, 2H), 7.84-7.95 (m, 2H), 8.34-8.42 (m, 1H), 8.75 (m, 1H); 13C (D2O, 75.5 MHz) δ 24.30, 30.12, 45.09, 47.92, 48.41, 50.27, 51.55, 55.48, 127.27, 127.42, 130.33, 130.99, 131.60, 137.59, 145.10, 145.76, 146.96; exact mass m/z calcd. for C24H37N5S 427.28, found [M+H]+428.30; Anal. (C24H37N5S) 4.3 HBr 2.6H2O)C, 35.05; H, 5.70; N, 8.52; S, 3.90; Br, 41.78. Found C, 35.18; H, 5.52; N, 8.48; S, 3.89; Br, 41.53.
  • EXAMPLE 7 Preparation of N-[4-(11-sulfoxo-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine. (AMD9424, FIG. 7)
  • A solution of Oxone® (504 mg, 0.82 mmol) in water (15 mL) was added dropwise to a cooled (−10° C.) stirred solution of N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (350 mg, 0.82 mmol) in methanol (15 mL). The mixture was stirred at −10° C. for 10 min then poured into saturated NaHCO3 solution (40 mL) and extracted with CHCl3 (4×50 mL). The separated organic layers were dried (MgSO4) and concentrated in vacuo. Purification of the crude product by chromatography on silica gel (3% MeOH/CH2Cl2) gave AMD9424 (75 mg, 21%). 1H NMR (CDCl3, 300 MHz) δ 1.90-2.01 (m, 2H), 2.06-2.17 (m, 2H), 2.59-2.85 (m, 14H), 3.07-3.18 (m, 2H), 3.57 (s, 2H), 3.83 (s, 2H), 3.92 (s, 2H), 7.17 (dd, J=7.5, 4.7 Hz, 2H), 7.23 (d, J=7.7 Hz, 2H), 7.32 (d, J=7.7 Hz, 2H), 7.65 (td, J=7.5, 1.7 Hz, 1H), 8.56 (d, J=4.9 Hz, 1H); 13C (CDCl3, 75.5 MHz) δ 24.36 (2), 47.52 (2), 48.30 (2), 52.38 (2), 52.81 (2), 53.55, 54.87, 59.11, 122.35, 122.73, 128.67 (2), 129.23 (2), 136.85, 138.00, 139.43, 149.68, 160.07; exact mass calculated for C24H37N5OS: 443, found: m/z 444 [M+H]+; Anal calculated for (C24H37N5OS.0.6 C4H10O.0.4H2O): C, 64.02; H, 8.91, N, 14.14, found: C, 64.06, H, 8.72, N, 13.98.
  • EXAMPLE 8 Preparation of N-[4-(11-sulfono-1,7-diazacyclotetradecanyl) 1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (hydrobromide salt). (AMD9408, FIG. 8)
  • Di-tert-butyldicarbonate (278 mg, 1.3 mmol) was added to a solution of N-[4-(11-thia-1,7-diazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (136 mg, 0.32 mmol) in THF (3.5 mL) and H2O (0.1 mL) and the mixture was stirred at room temperature overnight, under a nitrogen atmosphere. The mixture was concentrated and the residue was partitioned between CH2Cl2 (20 mL) and H2O (10 mL). The separated organic layer was washed with H2O (3×10 mL) and saturated NaCl solution (10 mL), dried (MgSO4), and concentrated in vacuo. Flash chromatography on silica gel (3% MeOH/CH2Cl2) provided the desired BOC intermediate as a clear oil (240 mg, 100%). 1H NMR (CDCl3, 300 MHz) δ 1.31-1.58 (m, 27H), 1.82-1.91 (m, 4H), 2.58-2.80 (m, 8H), 3.29-3.44 (m, 8H), 3.62 (s, 2H), 4.43 (s, 2H), 4.51 (d, J=7.4 Hz, 2H), 7.12-7.23 (m, 6H), 7.65 (t, J=7.5, 1H), 8.54 (d, J=4.0 Hz, 1H).
  • A solution of Oxone® (197 mg, 0.32 mmol) in water (2 mL) was added dropwise to a cooled (−10° C.) stirred solution of the BOC intermediate from above (232 mg, 0.32 mmol) in methanol (2 mL). The mixture was stirred at −10° C. for 10 min then poured into saturated NaHCO3 solution (5 mL) and extracted with CHCl3 (4×10 mL). The separated organic layers were dried (MgSO4) and concentrated in vacuo to give a clear viscous oil. The crude material was purified by column chromatography on a silica gel column (3% MeOH/CH2Cl2) to give the desired sulfone (73.3 mg, 30%). 1H NMR (CDCl3, 300 MHz) δ 1.39-1.50 (m, 27H), 2.06-2.14 (m, 4H), 2.60-2.68 (m, 4H), 3.10 (t, J=7.2 Hz, 4H), 3.29-3.44 (m, 8H), 3.62 (s, 2H), 4.43 (s, 2H), 4.52 (d, J=4.8 Hz, 2H), 7.15-7.21 (m, 6H), 7.65 (t, J=7.5, 1H), 8.54 (d, J=4.4 Hz, 1H); exact mass calculated for C39H61N5O8S: 759, found: m/z 760 [M+H]+.
  • The sulfone (73 mg, 0.96 mmol) was dissolved in a minimum amount of acetic acid and treated with a saturated solution of HBr in acetic acid (5 mL). The mixture was stirred under nitrogen for 63 h at room temperature and diethyl ether (100 mL) was added. A precipitate formed and was allowed to settle to the bottom of the flask and the supernatant solution was decanted. The white solid was washed with diethyl ether (5×100 mL) and the remaining traces of solvent were removed by blowing nitrogen though the flask followed by drying under vacuum overnight at 55° C. to give AMD9408 (70 mg, 84%) as a white solid. 1H NMR (CDCl3, 300 MHz) δ 2.36-2.44 (m, 4H), 2.84-2.87 (m, 4H), 3.21-3.23 (m, 4H), 3.38 (t, J=5.4 Hz, 4H), 3.67-3.70 (m, 4H), 3.73 (s, 2H), 4.34 (s, 2H), 4.39 (s, 2H), 7.44-7.51 (m, 6H), 7.93 (td, J=7.8, 1.7 Hz, 1H), 8.58 (d, J=6.1 Hz, 1H); 13C (CDCl3, 75.5 MHz) δ 18.76 (2), 45.41 (2), 47.04 (2), 47.79, 50.57 (2), 51.83, 51.88 (2), 56.71, 127.90, 128.23, 130.07, 131.03 (2), 131.29 (2), 138.44, 144.62, 145.95, 146.77; exact mass calculated for C24H37N5OS: 459, found: m/z 460 [M+H]+; Anal calculated for (C24H37N5O2S.4 HBr 3H2O): C, 33.14; H, 5.49, N, 8.05, Br, 40.42, found: C, 33.36, H, 5.39, N, 7.70, Br, 40.25.
  • EXAMPLE 9 N-[4-(3-carboxo-1,4,7-triazacyclotetradecanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine
  • Procedures as used in the preceding examples were used to synthesize this compound. The structure of this compound in provided in FIG. 9.
  • EXAMPLE 10 Inhibition of Chemokine Induced Ca Flux Measured on a FLIPR (Molecular Devices)
  • Reagents:
  • Loading dye: Fluo-3, AM (Molecular Probes F-1241) is dissolved in anhydrous DMSO and stored frozen in aliquots. To increase the solubility of the dye in the loading medium, 10% (w/v) pluronic acid (Molecular Probes F-127) is added to the Fluo-3 stock solution immediately before use.
  • Flux Buffer:
  • HBSS+20 mM Hepes buffer+0.2% BSA, pH 7.4. HBSS 10× [(w/o phenol red and sodium bicarbonate (Gibco 14 065-049)]; Hepes buffer 1M (Gibco 15 630-056), BSA (Sigma A3675). The flux buffer is vacuum-filtered and stored refrigerated for a maximum of 5 days. Before use in the experiment, the buffer is warmed at 37° C. in a waterbath.
  • Antagonists:
  • The test compounds are diluted in flux buffer and added to 4 wells of a black microplate (4 parallel measurements per compound). The following control wells are used: 100% response control (no inhibition), flux buffer is added; 100% inhibition control: chemokine is added at 5-times the concentration required to induce a Ca flux.
  • Preparation of the Agonist (Chemokine) Plate:
  • The chemokines are diluted in flux buffer to concentrations that are 4-fold higher than the desired concentrations required for stimulation of the cells (i.e. 2.5 nM for SDF-1α). The chemokines were added to untreated 96-well Sero well compound plates (International Medical, Sterilin code 611F96). In the negative control well's (baseline monitoring), flux buffer is added instead of chemokine. As a positive control to check for dye loading efficiency, 20 μM digitonin (final concentration) is also included. The agonist plate is incubated in the FLIPR (37° C.) for 15-30 min.
  • Cell Loading Protocol for Measuring Inhibition of SDF-1α Induced Ca Flux in SUP-T1 Cells
  • SUP-T1 cells are centrifuged at room temperature (RT) and re-suspended in loading medium (RPMI-1640 containing 2% FBS and 4 μM Fluo-3, AM). The cells are incubate at room temperature for 45 min. then washed twice in flux buffer then incubated in flux buffer at room temperature for 10 min. The cells are centrifuged and re-suspended in flux buffer at a density of 3×106 cells per mL. A 100 μL aliquot of the cell suspension (3×105 cells) is added to each well of a black microplate (Costar 3603), which already contains 50 μL of a solution of the test compound (at concentrations that are 3-fold higher than the desired final compound concentrations). The microplate is then gently centrifuged at room temperature. Homogeneous spreading of the cells on the bottom of the microplate wells is then confirmed with a microscope and the microplate was incubated in the FLIPR (37° C.) for 10 min. prior to testing.
  • Fluorescence Measurements as a Function of Time on the FLIPR
  • The FLIPR settings (camera exposure time and laser power) are adjusted to obtain initial fluorescence values between 8,000 and 10,000 units. After monitoring a 20 second-baseline, the agonist (chemokine) (50 μL) is added by automatic pipettor with black pipette tips. Fluorescence is measured simultaneously in all wells of the microplate every 2 seconds (first 2 min) and thereafter every 6 seconds (additional 2 min). The average α-flux measured in each set of 4 identical wells (one test compound) was calculated by the FLIPR software.
  • The compounds of the current invention were tested for inhibition of SDF-1α induced Ca flux in SUP-T1 cells using the method described above. The compounds described in Examples 1-9 inhibited SDF-1α induced Ca flux greater than 50% at 25 μg/mL.
  • EXAMPLE 11 Assay for Inhibition of HIV-1 (NL4.3) Replication in MT-4 Cells
  • Inhibition of HIV-1 NL4.3 (or IIIB) replication assays were performed as previously described (Bridger, et al, J. Med. Chem. 42:3971-3981 (1999); De Clercq, et al., Proc. Natl. Acad. Sci. 89:5286-5290 (1992); De Clercq, et al., Antimicrob. Agents Chemother. 38:668-674 (1994); Bridger, et al. J. Med. Chem. 38:366-378 (1995)). Anti-HIV activity and cytotoxicity measurements were carried out in parallel. They were based on the viability of MT-4 cells that had been infected with HIV in the presence of various concentrations of the test compounds. After the MT-4 cells were allowed to proliferate for 5 days, the number of viable cells was quantified by a tetrazolium-based colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) procedure in 96-well microtrays. In all of these assays, viral input (viral multiplicity of infection, MOI) was 0.01, or 100 times the 50% cell culture infective dose (CCID50). The EC50 was defined as the concentration required to protect 50% of the virus-infected cells against viral cytopathicity.
  • The compounds of the current invention were tested as described above. The compounds described in Examples 1-9 inhibited HIV-1 replication with EC50's in the range 0.003-33.3 ug/mL.
  • EXAMPLE 12 Inhibition of Collagen-Induced Arthritis
  • A compound related to those of the invention demonstrated inhibition of collagen-induced arthritis (CIA) in a mutant mouse model.
  • Experimental Animal Treatment
  • The control group consisted of ten mice that were injected with collagen as discussed below. The treatment group consisted of eight mice which were also injected with collagen and were further treated by administering 1,1′-[1,4-phenylenebis(methylene))]bis-1,4,8,11-tetraazacyclotetradecane (AMD 3100; see FIG. 10) intravenously using osmotic pumps (200 μl, Alza, 0.5 μl/hr) at a concentration of 5 mg/ml over a 14-day period following collagen injection.
  • Mutant Mice
  • The generation and the basic characteristics of the mutant mouse strain (129/Sv/Ev) with a disruption in the gene coding for the α-chain of the IFN-γ receptor (IFN-γ RKO) have been described (Huang, S., et al., Science 259:1742 (1993)). These IFN-γ RKO mice were back-crossed with DBA/1 wild-type mice for 10 generations to obtain the DBA/1 IFN-γ RKO mice used in the present study. IFN-γ RKO and wild-type mice were bred in the Experimental Animal Centre of the University of Leuven. The experiments were performed in 8- to 12-week old mice, but in each experiment, the mutant and wild-type mice were age-matched with a 5 day limit. The male to female ratio was kept between 0.8 and 1.3 in each experimental group.
  • Induction of Collagen-Induced Arthritis and Clinical Assessment of Arthritis
  • Collagen-induced arthritis was carried out in the following manner (see: Vermeire, et al., Int. J. Immunol. 158:5507-5513, (1997)). Native chicken collagen type II (EPC, Owensvillle, Mo.) was dissolved in 0.05 M acetic acid at 2 mg/ml by stirring overnight at 6° C., and emulsified in an equal volume of incomplete (IFA) or complete Freund's adjuvant (CFA) containing 1.5 mg/ml heat killed Mycobacterium butyricum (Difco, Detroit, Mich.). Mice were sensitized with a single 100 μl intradermal injection of the emulsion at the base of the tail. Mice were examined daily for signs of arthritis. The disease severity was recorded following a scoring system for each limb. Score 0: normal; score 1: redness and/or swelling in one joint; score 2: redness and/or swelling in more than one joint; score 3: redness and/or welling in the entire paw; score 4: deformity and/or ankylosis.
  • Histological Examination
  • Spleens and fore and hind limbs were fixed in buffered saline—B5fixative (10% formalin with quicksilver). Alternatively, tissues were fixed in 10% formalin or pure methanol (see: Vermeire, et al., J. Immunol. 158:5507-5513 (1997)). Limbs were subsequently decalcified overnight with formic acid. Four-micron thick paraffin sections were stained with hematoxylin and eosin. Severity of arthritis was evaluated using three parameters: infiltration of mono- and polymorphonuclear cells, hyperplasia of the synovium and parmus formation. Each parameter was scored on a scale from 0 to 3 (absent; weak, moderate and severe).
  • In Vivo Antibody Treatments
  • Monoclonal antibodies were produced from hybridomas grown by intraperitoneal inoculation in Pristane-primed athymic nude mice (nu/nu of NMRI background). Neutralizing monoclonal antibody against MuIFN-γ (F3, rate IgG24) was purified by affinity chromatography on a mouse anti-rat K chain monoclonal antibody (Billiau, A., et al., J. Immunol. 140:1506 (1988)). The neutralizing titer (end-point dilution corresponding to 50% neutralization of the antiviral effect of 30 units/ml of mouse IFN-γ on mouse 1929 cells challenged with mengovirus) was 1053 U/ml (IgG content, 1.4 mg/ml). A neutralizing rate IgG24 antibody against murine IL-12 was produced using hybridoma C17.8 (kindly provided by Dr. G. Trinchieri, Wistar Institute, Philadelphia, Pa.). The antibody was purified by affinity chromatography on proteinG (Pharmacia, Uppsala, Sweden). Antibody against murine IL-6 was prepared from ascites fluid from thymus-less nude mice inoculated with the 20F3 (rat x mouse) hybridoma (American Type Culture Collection, Rockville, Md.). This rat IgG antibody was purified by affinity chromatography on an anti-rat K chain monoclonal antibody-Sepharose column. The neutralizing titer (endpoint dilution corresponding to 50% neutralization of the cell growth effect of 10 U of murine IL-6 per ml) was 1055 (IgG content: 2.9 mg/ml). Irrelevant rat IgG24 was used as an isotope control and was prepared from ascites fluid of a rat plasmocytoma (obtained through the courtesy of Dr. H. Bazin, University of Louvain, Medical School, Brussels, Belgium). The IgG was purified by anion exchange chromatography on Hiload Q Sepharose and gel filtration on Superdex 200 (Pharmacia). Batches of anti-IFN-γ, anti-IL-12, anti-IL-6 and irrelevant IgG24 were tested for endotoxin content by a chromogenic Limulus amoebocyte lysate assay (KabiVitrum, Stockholm, Sweden) and were found to contain less than 2 ng/ml endotoxin. Injections were given in 200 μl of pyrogen-free saline.
  • Following 14 days after treatment, 7 of the ten mice in the control group demonstrated arthritis, while only 1 of the 8 animals treated with AMD 3100 demonstrated disease. The single treated animal did not develop arthritic pathology until after 20 days post-treatment. Additionally, the treated animals compared with the control animals did not demonstrate any significant body weight loss. Further, the treated animals maintained body weight consist with healthy animals not injected with collagen.
  • EXAMPLE 13 Treatment of Glioblastoma
  • Compounds of the present invention may be used in the treatment of glioblastomas, fibromas, astrocytomas or myelomas affecting the central nervous system. The compounds may be used according to standard clinical practice and procedures, using dosages as provided in the foregoing examples and according to clinical end points, such as imaging, immunological and other methodologies.
  • For example, the etiology or association of chemokine receptor binding in the proliferation of glioblastoma tumor cells has been reported by Sehgal, et al., J. of Surg. Oncolo. 69:99-104 (1998) (“Sehgal I”) and Sehgal, et al., J. of Surg. Oncolo. 69:239-248 (1998) (“Sehgal II”). The role of CXCR4 of its binding to its receptor appears to play a significant role in the formation and/or proliferation of glioblastoma cells. The inhibition of the binding by CXCR4 to its natural receptor ligand by compounds of the present invention offer a new drug in the treatment tumors of central nervous system that are mediated or associated with chemokines, such as CXCR4.
  • EXAMPLE 14 Treatment of Non-Small Cell Lung Cancer
  • Compounds of the present invention may be used in the treatment of non-small cell lung cancer. The compounds may be used according to standard clinical practice and procedures, using dosages as provided in the foregoing examples and according to clinical end points, such as imaging, immunological and other methodologies.
  • For example, CXC chemokines have been found to regulate or are associated with the regulation of angiogenesis in non-small cell lung cancer (see: Arenberg, et al., J. of Leukocyte Biol. 62:554-562 (1997); and Moore, et al., TCM, vol. 8(2):51-58 (1998) Elsevier Science, Inc.). The role of CXC chemokines and the binding to their respective receptors appear to play a significant role in the formation and/or proliferation of non-small cell lung cancer promoted by an increase in angiogenic activity. The inhibition of the binding of these CXC chemokines to their natural receptor ligands by compounds of the present invention offer a new drug in the treatment tumors such as non-small cell lung cancer that are mediated or associated with increased levels of chemokines.
  • EXAMPLE 15 N-[1,4,8,11-Tetraazacyclotetradecanyl-1, 4-phenylenebis(methylene)]-2-(amino-methyl)pyridine hexahydrobromide (AMD 3465)
  • White solid: Mp 200-205° C. (dec); 1H NMR (D2O) δ 2.04 (m, 4H), 3.20-3.40 (m, 8H), 3.40-3.60 (m, 8H), 4.34 (s, 2H), 4.38 (s, 2H), 4.51 (s, 2H), 7.50 (m, 4H), 7.75 (t, 1H, J=6.6 Hz), 7.82 (d, 1H, J=7.9 Hz), 8.26 (t, 1H, J=7.9 Hz), 8.63 (d, 1H, J=5.3 Hz); 13C NMR (D2O) δ 18.30, 18.96, 37.04, 37.28, 37.40, 40.92, 41.13, 41.49, 44.26, 47.61, 48.01, 51.29, 58.88, 127.46, 127.75, 130.40, 131.05, 131.23, 131.47, 132.10, 132.44, 144.95, 145.81, 146.01; FAB MS m/z 493 (M+H81Br, 7), 491 (M+H79Br, 7), 411 (M+H, 100). Anal. (C24H38N6.6HBr); Calc. C, 32.36; H, 4.98; N, 9.44; Br, 53.21. Found C, 32.20; H, 5.00; N, 9.30; Br, 53.10.
  • EXAMPLE 16 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-N-methyl-2-(aminomethyl)pyridine hexahydrobromide hydrate (AMD 3538)
  • White solid: Mp 220-225° C. (dec); 1H NMR (D2O) δ 2.06 (m, 4H), 2.76 (s, 3H), 3.20-3.65 (m, 16H), 4.47 (bs, 4H), 4.65 (s, 2H), 7.54 (bs, 4H), 7.80 (t, 1H), 7.87 (d, 1H), 8.28 (t, 1H), 8.68 (d, 1H); 13C NMR (D2O) δ 18.14, 18.75, 18.89, 36.74, 37.04, 37.15, 37.62, 40.38, 40.72, 40.91, 41.28, 44.05, 47.50, 56.98, 58.88, 60.28, 127.60, 128.86, 130.78, 130.96, 132.16, 132.64, 144.91, 145.04, 146.12; FAB MS m/z 507 (M+H81Br, 27), 507 (M+H79Br, 22), 425 (M+H, 100). Anal. (C25H40N6.6HBr.1.5H2O); Calc. C, 32.04; H, 5.27; N, 8.97; Br, 51.16. Found C, 31.88; H, 5.30; N, 8.93; Br, 51.00.
  • EXAMPLE 17 N-[1,4,8,11-Tetraazacyclotetradecanyl-1, 4-phenylenebis(methylene)]-4-)amino-methyl)pyridine hexahydrobromide (AMD 3500)
  • White solid: mp 201-204° C. (dec); 1H NMR (D2O) δ 1.91-2.12 (m, 4H), 3.00-3.49 (m, 16H), 4.13 (s, 2H), 4.34 (s, 2H), 4.53 (s, 2H), 7.39-7.57 (m, 4H), 8.02 (d, 2H, J=6.3 Hz), 8.74 (d, 2H, J=6.3 Hz); 13C NMR (D2O) δ 18.26, 18.88, 36.94, 37.29, 37.36, 40.89, 41.06, 41.44, 44.21, 47.61, 49.17, 51.43, 59.02, 127.84, 130.21, 131.64, 132.15, 132.45, 142.19, 151.67; FAB MS m/z 493 (M+H81Br, 8), 491 (M+H79Br, 10), 411 (M+H, 83), 320 (37), 247 (58), 201 (100). Anal. (C24H38N6.6HBr); Calc. C, 32.17; H, 4.95; N, 9.34; Br, 53.50. Found C, 32.16; H, 5.03; N, 9.41; Br, 53.28.
  • EXAMPLE 18 N-[1,4,8,11-Tetraazacyclotetradecanyl-1, 4-phenylenebis(methylene)]-3-(amino-methyl)pyridine hexahydrobromide (AMD 3499)
  • White solid: mp 198-202° C. (dec); 1H NMR (D2O) δ 1.83-2.07 (m, 4H), 2.96-3.47 (m, 16H), 4.11 (s, 2H), 4.32 (s, 2H), 4.49 (s, 2H), 7.38-7.56 (m, 4H), 8.04 (t, 1H, J=6.4 Hz), 8.63 (d, 1H, J=8.3 Hz), 8.76 (d, 1H, J=5.6 Hz), 8.86 (s, 1H); 3C NMR (D2O) δ 18.23, 18.87, 36.92, 37.29 (2C), 40.88, 41.05, 41.43, 44.17, 47.22, 47.60, 51.18, 59.04, 128.29, 130.01, 131.49, 132.14, 132.66 (2C), 142.55, 142.76, 148.98; FAB MS m/z 493 (M+H81Br, 7), 491 (M+H79Br, 6), 411 (M+H, 100), 320 (33), 247 (24). Anal. (C24H38N6.6HBr); Calc. C, 32.17; H, 4.95; N, 9.34; Br, 53.50. Found C, 32.08; H, 5.02; N, 9.25; Br, 53.28.
  • EXAMPLE 19 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-(2-amino-methyl-5-methyl)pyrazine pentahydrobromide (AMD 3498)
  • White solid: mp 194-197° C. (dec); 1H NMR (D2O) δ 1.93-2.12 (m, 4H), 2.42 (s, 3H), 3.25 (s, 8H), 3.48 (s, 8H), 4.28 (s, 2H), 4.30 (s, 2H), 4.33 (s, 2H), 7.44 (s, 4H), 8.33 (s, 1H), 8.46 (s, 1H); 13C NMR (D2O) δ 18.01, 18.72, 19.80, 36.66, 37.05, 37.13, 40.70, 40.89, 41.27, 43.99, 47.47, 48.14, 50.61, 59.06, 129.97, 131.43, 132.04, 132.99, 140.93, 144.98, 146.49, 153.51; FAB MS m/z 509 (M+H81Br, 17), 507 (M+H79Br, 15), 426 (M+H, 100), 320 (21), 247 (20). Anal. (C24H39N7.5.5HBr); Calc. C, 33.10; H, 5.15; N, 11.26; Br, 50.47. Found C, 32.80; H, 5.41; N, 11.00; Br, 50.58.
  • EXAMPLE 20 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-ethyl)pyridine hexahydrobromide (AMD 3497)
  • White solid: mp 195-198° C. (dec); 1H NMR (D2O) δ 1.98-2.17 (m, 4H), 3.20-3.38 (m, 8H), 3.38-3.63 (m, 12H), 4.27 (s, 2H), 4.39 (s, 2H), 7.50 (s, 4H), 7.80-7.89 (m, 2H), 8.42 (m, 1H), 8.58 (d, 1H, J=5.8 Hz); 13C NMR (D2O) δ 18.51, 19.14, 29.85, 37.56 (3C), 41.21, 41.41, 41.82, 44.57, 45.27, 47.83, 51.10, 58.74, 126.35, 127.93, 130.66, 131.27, 131.99, 132.69, 141.89, 147.79, 150.91; FAB MS m/z 507 (M+H81Br, 40), 505 (M+H79Br, 34), 425 (M+H, 100). Anal. (C25H40N6.6HBr); Calc. C, 32.99; H, 5.09; N, 9.23; Br, 52.67. Found C, 32.79; H, 5.34; N, 9.11; Br, 52.45.
  • EXAMPLE 21 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-methyl)thiophene pentahydrobromide (AMD 3516)
  • White solid: mp 245-248° C. (dec); 1H NMR (D2O) δ 1.87-2.12 (m, 4H), 3.02-3.51 (m, 16H), 4.17 (s, 4H), 4.38 (s, 2H), 6.97 (t, 1H, J=3.9 Hz), 7.13 (d, 1H, J=3.1 Hz), 7.41 (s, 5H); 3C NMR (D2O) δ 18.80, 19.52, 38.03, (3C), 41.59 (2C), 42.21, 44.89 (2C), 48.15, 49.83, 58.52, 128.13, 129.12, 131.15, 131.47, 131.50, 131.90, 132.42, 132.87; FAB MS m/z498 (M+H81Br, 11), 496 (M+H79Br, 9), 416 (M+H, 53), 218 (100), 201 (64). Anal. (C23H37N5S.5HBr); Calc. C, 33.68; H, 5.16; N, 8.54; Br, 48.71. Found C, 33.85; H, 5.22; N, 8.50; Br, 48.52.
  • EXAMPLE 22 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-ethyl)mercaptan pentahydrobromide dihydrate (AMD 3530)
  • White solid: mp 234-236° C. (dec); 1H NMR (D2O) δ 1.75-2.05 (m, 4H), 2.75-3.45 (m, 20H), 4.05 (s, 2H), 4.15 (s, 2H), 7.35 (s, 4H); FAB MS m/z 462 (MH+H81Br, 15), 460 (MH+H79Br, 15), 380 (M+H, 100), 300 (64), 279 (47), 239 (49). Anal. (C20H37N5S.5HBr.2H2O.0.5HOAc) requires C, 29.67; H, 5.69; N, 8.24; Br, 46.99. Found C, 29.31; H, 5.72; N, 8.25; Br, 46.64.
  • EXAMPLE 23 N-[1,4,8,11-Tetraazacyclotetradecanyl-1, 4-phenylenebis(methylene)]-2-amino-benzylamine pentahydrobromide (AMD 3517)
  • White solid: mp 203-206° C. (dec); 1H NMR (D2O) δ 1.85-2.13 (m, 4H), 3.02-3.58 (m, 16H), 4.23 (s, 2H), 4.31 (s, 4H), 7.23-7.54 (m, 8H); 3C NMR (D2O) δ 18.03, 19.29, 37.78 (3C), 41.37 (2C), 42.00, 44.82, 46.25, 47.96, 51.16, 58.68, 124.04, 124.40, 129.40, 130.75, 131.21 (2C), 131.88, 131.96, 132.46, 132.83; FAB MS m/z 507 (M+H81Br, 15), 505 (M+H79Br, 18), 425 (M+H, 100), 320 (30), 201 (51). Anal. (C25H40N6.5.75HBr.0.5H2O). Calc. C, 33.42; H, 5.19; N, 9.35; Br, 51.14. Found C, 33.69; H, 5.35; N, 9.00; Br, 51.13.
  • EXAMPLE 24 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-Ihenylenebis(methylene)]-4-amino-benzylamine hexahydrobromide (AMD 3544)
  • Yellow solid: mp 120-125° C. (dec); 1H NMR (D2O) δ 1.8-2.0 (m, 4H), 2.9-3.4 (m, 16H), 4.1 (s, 2H), 4.18 (s, 4H), 7.2-7.5 (m, 8H); 13C NMR (D2O) δ 18.86, 19.57, 38.14, 41.76, 43.74, 45.14, 48.24, 50.14, 50.42, 51.49, 58.38, 124.13, 131.13, 131.30, 131.83, 131.92, 131.96, 132.67; FAB MS m/z 507 (M+H81Br, 5), 505 (M+H79Br, 5), 425 (M+H, 45), 201 (47), 155 (75), 106 (100). Anal. (C25H40N6.6HBr.HOAc) requires C, 33.43; H, 5.19; N, 8.66; Br, 49.42; O, 3.30. Found C, 33.42; H, 5.49; N, 8.62; Br, 49.23.
  • EXAMPLE 25 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-(amino-ethyl)imidazole hexahydrobromide (AMD 3543)
  • Off white solid: mp 135-140° C. (dec); 1H NMR (D2O) δ 1.75 (m, 2H), 190 (m, 2H), 2.70-3.27 (m, 20H), 3.77 (s, 2H), 4.14 (s, 2H), 7.18 (s, 1H), 7.25 (d, 2H, J=7.97 Hz), 7.37 (d, 2H, J=7.97 Hz), 8.48 (s, 1H); FAB MS m/z 496 (M+H81Br, 5), 494 (M+H79Br, 5), 414 (M+H, 17), 201 (15). Anal. (C23H39N7.6HBr) requires C, 30.73; H, 5.04; N, 10.91; Br, 53.32. Found C, 30.39; H, 5.41; N, 10.41; Br, 53.66.
  • EXAMPLE 26 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-benzylamine pentahydrobromide (AMD 3529)
  • Off white solid: mp 245-250° C. (dec); 1H NMR (D2O) δ 1.9-2.1 (m, 4H), 3.2-3.6 (m, 16H), 4.12 (s, 2H), 4.15 (s, 2H), 4.36 (s, 2H), 7.30 (s, 5H), 7.41 (d, 2H, J=8.3 Hz) 7.46 (d, 2H, J=8.3 Hz); 13C NMR (D2O) δ 18.43, 19.06, 37.29, 37.46, 37.63, 41.09, 41.32, 41.68, 44.46, 47.74, 50.18, 51.00, 58.79, 129.53, 129.97, 130.18, 130.35, 130.68, 131.18, 131.92, 133.14; FAB MS m/z 492 (M+H81Br, 13), 490 (M+H79Br, 13), 410 (M+H, 100), 201 (36). Anal. (C25H39N5.5HBr); requires C, 36.88; H, 5.45; N, 8.60; Br, 49.07. Found C, 36.79; H, 5.56; N, 8.48; Br, 48.79.
  • The compounds of the invention were tested in a screen by the MTT method (J. Virol. Methods 120:309-321 (1988)). MT-4 cells (2.5×104/well) were challenged with HIV-1 (HTLV-IIIB) or HIV-2 (LAV-2 ROD) at a concentration of 100 CCID50 and incubated in the presence of various concentrations of the test compounds, which were added immediately after challenge with the virus. After 5 days culture at 37° C. in a CO2 incubator, the number of viable cells was assessed by the MTT (tetrazolium) method. Antiviral activity and cytotoxicity of the compounds are expressed in Table 1 below as EC50 (μg/ml) and CC50 (μg/ml), respectively. The potential therapeutic usefulness was assessed by calculating a Selectivity Index (SI) corresponding to the ratio of CC50 to EC50.
    TABLE 1
    Anti-HIV activity data
    CC50 EC50 (μg/mL) SI
    Compound (μg/mL) HIV-1 (IIIB) HIV-2 HIV-1
     1 AMD3465 >250 0.008 0.032   3 × 104
     2 AMD3538 209 0.1 6.7 2.0 × 103
     3 AMD3500 >250 0.6 10.3 417
     4 AMD3499 >250 1.8 28.5 138
     5 AMD3498 >250 0.2 7.1 1.2 × 103
     6 AMD3497 >250 1.8 3.8 138
     7 AMD3516 158 0.7 9.8 225
     8 AMD3530 175 0.5 2.0 350
     9 AMD3517 153 0.8 10.6 191
    10 AMD3544 222 0.7 3.7 317
    11 AMD3543 239 0.2 1.0   1 × 103
    12 AMD3529 130 0.4 2.6 325
  • In this field of study, it is considered that any compound exhibiting a Selectivity Index of greater than 100 has the considerable potential for further study. HIV is one of the most challenging viruses to combat, and the results given above provide an indication of activity against other retroviruses and against other viruses in general.
  • EXAMPLE 27 N-[4-(1,4,7-Triazacyclotetra-decanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7049) N,N′-Bis(2-Nitrobenzenesulfonyl)-1,7-heptanediamine
  • To a stirred solution of 1,7-heptanediamine (5.01 g, 38.5 mmol) and Et3N (13.5 mL, 96.9 mmol) in CH2Cl2 (70 mL) was added a solution of 2-nitrobenzenesulfonyl chloride (18.80 g, 84.83 mmol) in CH2Cl2 (40 mL). The mixture was stirred at room temperature under nitrogen for 72 h and then concentrated in vacuo. The residue was stirred in diethyl ether (100 mL), and the precipitate was collected by filtration and washed with H2O (300 mL) followed by diethyl ether (300 mL) to give a gray solid (18.5 g, 96%): 1H NMR (DMF-d7) δ 1.21 (m, 6H), 1.49 (m, 4H), 3.04 (m, 4H), 7.87 (m, 2H), 7.95 (m, 4H), 8.04 (m, 2H), 8.15 (m, 2H).
  • General Procedure D 4-Diethoxyphosphoryl-1, 7-bis(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane
  • To a stirred solution of N,N′-bis(2-nitrobenzenesulfonyl)-1,7-heptanediamine (9.00 g, 18.0 mmol) and Cs2CO3 (17.8 g, 54.6 mmol) in DMF (500 mL) under nitrogen maintained at 80° C. was added dropwise a solution of N-(diethoxyphosphoryl)-O,O′-bis(2-methylsulfonyl)di-ethanolamine (Bridger et al., J. Med. Chem. 1995, 38, 366-378) (7.95 g, 20.0 mmol) in DMF (50 mL) over 8 h. Heating was continued for a further 17 h and the mixture was then allowed to cool and concentrated in vacuo. The residue was partitioned between CHCl3 (140 mL) and H2O (80 mL) and the aqueous layer was separated and extracted with CHCl3 (3×40 mL). The combined organic extracts were dried (MgSO4) and concentrated in vacuo and the residue was purified by column chromatography on silica gel (ethylacetate) to give the desired macrocycle as a yellow crystalline solid (2.85 g, contaminated with DMF).
  • To remove the unwanted DMF impurity, the residue was dissolved in EtOAc (75 mL), and the solution was washed sequentially with 5% NaHCO3 (2×10 mL) and brine (5×10 mL), dried (MgSO4) and evaporated to give a yellow amorphous solid (2.52 g, 20%): 1H NMR (CDCl3) δ 1.32 (t, 6H, J=7.1 Hz), 1.51 (m, 6H), 1.61 (m, 4H), 3.33 (m, 12H), 4.03 (m, 4H), 7.61 (m, 2H), 7.71 (m, 4H), 8.03 (m, 2H).
  • General Procedure E Synthesis of 1,7-Bis(2-nitrobenzenesulfonyl)-1, 4,7-triazacyclotetradecane
  • To a stirred suspension of the macrocycle from above (1.88 g, 2.66 mmol) in acetic acid (5 mL) was added a freshly prepared solution of saturated HBr(g) in acetic acid (20 mL) and the resulting homogeneous solution was stirred at room temperature for a further 22 h. Addition of diethyl ether (250 mL) to the reaction mixture gave a precipitate that was allowed to settle to the bottom of the flask and the supernatant solution was decanted. The precipitate was washed with ether by decantation (repeated 3×) and the residue was then partitioned between CH2Cl2 (40 mL) and 1N aqueous NaOH (25 mL). The separated aqueous layer was extracted with CH2Cl2 (2×20 mL) and the combined organic extracts were washed with brine (20 mL), then dried (MgSO4) and concentrated in vacuo to give a yellow amorphous solid (1.23 g, 81%): 1H NMR (CDCl3) δ 1.46-1.67 (m, 10H), 2.90 (m, 4H), 3.34 (m, 8H), 7.61 (m, 2H), 7.70 (m, 4H), 7.97 (m, 2H).
  • 4-Bromomethylbenzyl alcohol
  • To a solution of methyl 4-bromomethylbenzoate (5.73 g, 25 mmol) in dry CH2Cl2 (150 mL) cooled to −78° C. with stirring under nitrogen was added dropwise a solution of DIBAL-H (82.5 mL, 1.0 M solution in THF). Stirring was continued for 1.5 h at −78° C., and the reaction mixture was then allowed to warm to 0° C. and quenched with H2O. The organic layer was separated and the aqueous was extracted with CH2Cl2 (2×100 mL). The combined organic extracts were dried (MgSO4) and evaporated to give the desired alcohol (5.0 g, 100%) as a white solid: 1H NMR (CDCl3) δ 1.84 (br, 1H), 4.49 (s, 2H), 4.67 (s, 2H), 7.33 (d, 2H, J=8.2 Hz), 7.38 (d, 2H, J=8.2 Hz).
  • N-(2-Nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • A solution of 2-nitrobenzenesulfonylchloride (16.62 g, 0.075 mol) in dry CH2Cl2 (120 mL) was added dropwise via cannula to a stirred solution of 2-(aminomethyl)pyridine (5.41 g, 0.05 mol) and Et3N (13.9 mL, 0.10 mol) in dry CH2Cl2 (150 mL) under nitrogen. The reaction mixture was stirred for three hours at room temperature, and then quenched with water (20 mL). The aqueous layer was separated and extracted with EtOAc (5×80 mL). The combined organic extracts were dried (MgSO4) and evaporated to small volume to give a white precipitate which was collected by filtration and washed with cold CH2Cl2 to give the desired product (11.37 g, 78%) as a white solid: 1H NMR (Acetone-d6) δ 4.46 (s, 2H), 7.19 (dd, 1H, J=7.4, 4.5 Hz), 7.25-7.35 (br s, 1H), 7.39 (d, 1H, J=7.7 Hz), 7.68 (ddd, 1H, J=7.7, 7.5, 1.8 Hz), 7.76-7.88 (m, 2H), 7.94 (dd, 1H, J=7.7, 1.5 Hz), 8.04 (dd, 1H, J=7.5, 1.8 Hz), 8.38 (d, 1H, J=4.5 Hz).
  • N-[1-Methylene-4-(hydroxymethylene)phenylene]-N-(2-Nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • A mixture of N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (5.87 g, 20 mmol), 4-bromomethylbenzyl alcohol (4.02 g, 20 mmol) and K2CO3 (5.53 g, 40 mmol) in dry CH3CN (150 mL) were heated at 60° C. for 4 h with stirring under nitrogen. The mixture was then allowed to cool to room temperature, the solvent evaporated and the residue was partitioned between water and CH2Cl2. The separated aqueous phase was extracted with CH2Cl2, and the combined organic extracts were dried (MgSO4) and evaporated. The residue was suspended in ethyl acetate/hexane (1:1) and collected by filtration to give the desired product (6.87 g, 83%) as a white solid: 1H NMR (CDCl3) δ 1.78 (t, 1H, J=5.8 Hz), 4.58 (s, 2H) 4.60 (s, 2H), 4.64 (d, 2H, J=5.8 Hz), 7.13-7.26 (m, 6H), 7.54-7.59 (m, 2H), 7.66-7.68 (m, 2H), 7.98 (d, 1H, J=7.4 Hz), 8.40 (d, 1H, J=3.8 Hz).
  • N-[1-Methylene-4-(chloromethylene)phenylene]-N-(2-Nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • To a stirred solution of the alcohol from above (1.91 g, 4.62 mmol) and Et3N (2.0 mL, 14 mmol) in CH2Cl2 (20 mL) cooled in an ice bath under nitrogen, was added methanesulfonyl chloride (0.73 mL, 9.4 mmol) and the reaction mixture was then heated to reflux for a further 6 h. The solution was diluted with CH2Cl2 (60 mL) and washed with 10% aqueous HCl (2×20 mL), 5% aqueous NaHCO3 (20 mL), and H2O (25 mL) then dried (MgSO4) and concentrated in vacuo to give an orange oil (1.95 g, 98%): 1H NMR (CDCl3) δ 4.52 (s, 2H), 4.60 (s, 4H), 7.12-7.26 (m, 6H), 7.55 (m, 2H), 7.67 (d, 2H, J=4.0 Hz), 7.94 (d, 1H, J=8.0 Hz), 8.41 (d, 1H, J=4.8 Hz). This was used without further purification.
  • General Procedure F N-[4-[1,7-Bis(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetra-decanyl]-1,4-phenylenebis(methylene)]-N-(2-Nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • A mixture of 1,7-bis(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane (1.1 g, 1.9 mmol), the chloride from above (0.98 g, 2.3 mmol) and K2CO3 (0.85 g, 6.2 mmol) were heated to reflux in CH3CN (30 mL) under nitrogen for 62 h. The solvent was evaporated in vacuo and the residue was partitioned between CH2Cl2 (100 mL) and brine (70 mL). The aqueous phase was separated and extracted with CH2Cl2 (40 mL) and the combined organic extracts were dried (MgSO4) and concentrated in vacuo. The residue was purified by column chromatography on silica gel (3% MeOH/CH2Cl2) and the evaporated fractions containing the desired product were subjected to a second column purification on silica gel (ethyl acetate) to give a pale yellow amorphous solid (940 mg, 49%): 1H NMR (CDCl3) δ 1.44 (br s, 6H), 1.60 (br s, 4H), 2.75 (m, 4H), 3.23-3.33 (m, 8H), 3.59 (s, 2H), 4.58 (s, 2H), 4.59 (s, 2H), 7.08-7.20 (m, 6H), 7.55-7.70 (m, 10H), 7.82 (dd, 2H, J=7.6, 1.6 Hz), 7.99 (d, 1H, J=7.8 Hz), 8.40 (d, 1H, J=4.7 Hz).
  • N-[4-(1,4,7-Triazacyclotetra-decanyl)-1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Pentahydrobromide Dihydrate
  • The intermediate from above (870 mg, 0.90 mmol), K2CO3 (1.15 g, 8.32 mmol), and thiophenol (0.33 mL, 3.2 mmol) were stirred in DMF (12 mL) for 7.5 h at room temperature. The mixture was concentrated in vacuo and the residue was partitioned between CH2Cl2 (30 mL) and H2O (15 mL). The organic phase was separated, washed with 5% NaHCO3 (10 mL) then H2O (10 mL) then dried (MgSO4) and concentrated in vacuo. The yellow residue was purified by column chromatography on basic alumina (CH2Cl2, 1% MeOH/CH2Cl2, and 10% MeOH/CH2Cl2) to give the free base as a yellow oil (134 mg, 36%): 1H NMR (CDCl3) δ 1.48 (br s, 6H), 1.60 (br s, 4H), 2.61 (m, 12H), 3.56 (s, 2H), 3.83 (s, 2H), 3.92 (s, 2H), 7.16 (m, 1H), 7.24 (m, 2H), 7.32 (m, 3H), 7.79 (m, 1H), 8.56 (d, 1H, J=4.7 Hz).
  • The free base (134 mg, 0.33 mmol) was dissolved in EtOH (4 mL) and a freshly prepared solution of saturated HBr(g) in EtOH (9 mL) was added, giving a white precipitate. The mixture was stirred for 5 min and diethyl ether (15 mL) was added. The precipitate was allowed to settle to the bottom of the flask and the supernatant solution was decanted. The solid was then dissolved in MeOH (5 mL) and re-precipitated with a large volume of ether, washed with ether by decantation (15×) and finally, the last traces of ether were removed by evaporation at reduced pressure (room temperature). Drying the solid in vacuo at 40° C. for 16 h, gave the desired product as a white solid (178 mg, 63%): 1H NMR (DMSO-d6) δ 1.44 (br s, 6H), 1.75 (br s, 4H), 3.04 (br s, 8H), 3.37 (m, 4H), 4.06 (br s, 2H), 4.31 (s, 2H), 4.38 (s, 2H), 7.52-7.68 (m, 6H), 8.01 (m, 1H), 8.70 (d, 1H, J=5.0 Hz); FAB-MS m/z 492 (MH+H81Br), 490 (MH+H79Br), 410 (M+H). Anal. Calcd for C25H39N5.5HBr.0.1Et2O.2.3H2O: C, 35.35; H, 5.79; N, 8.11; Br, 46.29. Found: C, 35.55; H, 5.70; N, 8.18; Br, 46.17.
  • EXAMPLE 28 N-[7-(4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7050)
  • 2,6-Bis(2-aminoethyl)pyridine was prepared as described in Bridger et al. U.S. Pat. No. 5,698,546, which is hereby incorporated in its entirety by reference herein.
  • 2,6-Bis[N-(2-nitrobenzenesulfonyl)-2-aminoethyl]pyridine
  • To a stirred solution of 2,6-Bis(2-aminoethyl)pyridine (2.7 g, 16 mmol) and Et3N (5.7 mL, 41 mmol) in CH2Cl2 (35 mL) was added 2-nitrobenzenesulfonyl chloride (8.01 g, 36.1 mmol) in CH2Cl2 (20 mL) and the mixture was stirred at room temperature under nitrogen for 42 h. The mixture was washed with brine (25 mL) and the organic phase was dried (MgSO4) and concentrated in vacuo. The brown residue was purified by column chromatography on silica gel (50% then 60% THF/hexane) to give a pale yellow solid (5.2 g, 59%): 1H NMR (CDCl3) δ 3.01 (m, 4H), 3.52 (m, 4H), 6.38 (m, 2H), 6.94 (d, 2H, J=7.7, Hz), 7.47 (t, 1H, J=7.7 Hz), 7.72 (m, 4H), 7.82 (m, 2H), 8.13 (m, 2H).
  • 7-Diethoxyphosphoryl-4,10-Bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene
  • Using General Procedure D: Reaction of 2,6-bis[N-(2-nitrobenzenesulfonyl)-2-aminoethyl]pyridine (5.2 g, 9.7 mmol) and N-(diethoxyphosphoryl)-O,O′-bis(2-methylsulfonyl)di-ethanolamine (4.25 g, 10.7 mmol) followed by silica gel column purification (60% then 90% THF/hexane) of the reaction products gave the title compound as a yellow amorphous solid (1.48 g, 21%): 1H NMR (CDCl3) δ 1.23 (t, 6H, J=7.1 Hz), 2.60 (m, 4H), 2.98-3.08 (m, 8H), 3.84-3.94 (m, 8H), 7.11 (d, 2H, J=7.6 Hz), 7.56-7.74 (m, 7H), 8.07 (m, 2H).
  • 4,10-Bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene
  • Using General Procedure E: Reaction of 7-diethoxyphosphoryl-4,10-bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (1.04 g, 1.4 mmol) gave the title compound as a yellow amorphous solid (744 mg, 88%): 1H NMR (CDCl3) δ 2.81 (m, 4H), 3.08 (m, 4H), 3.33 (m, 4H), 3.88 (m, 4H), 7.07 (d, 2H, J=7.7 Hz), 7.54-7.71 (m, 7H), 8.02 (m, 2H).
  • N-[7-[4,10-Bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure F: Reaction of 4,10-bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (740 mg, 1.2 mmol) and N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (610 mg, 1.4 mmol) followed by silica gel column purification (50% then 80% THF/hexane) of the reaction products gave the title compound as a yellow amorphous solid (648 mg, 54%): 1H NMR (CDCl3) δ 2.26 (m, 4H), 3.03 (m, 8H), 3.37 (s, 2H), 3.94 (m, 4H), 4.56 (s, 2H), 4.57 (s, 2H), 6.95-7.17 (m, 8H), 7.52-7.72 (m, 11H), 7.85 (m, 2H), 7.98 (d, 1H, J=7.7 Hz), 8.39 (d, 1H, J=4.8 Hz).
  • General Procedure G N-[7-(4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Hexahydrobromide Trihydrate
  • To a solution of N-[7-[4,10-bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (640 mg, 0.64 mmol) in DMF (9 mL) containing K2CO3 (806 mg, 5.83 mmol) was added thiophenol (0.24 mL, 2.3 mmol) and the mixture was stirred at room temperature for 2 h. The mixture was concentrated in vacuo and the residue partitioned between ethyl acetate (30 mL) and water (10 mL). The organic phase was separated and extracted with 5% NaHCO3 (3×5 mL) then brine (5 mL). The combined aqueous phases were extracted with CH2Cl2 (3×10 mL). The combined organic extracts were dried (MgSO4) and evaporated and the residue was purified by column chromatography on alumina (CH2Cl2 followed by 10% MeOH/CH2Cl2) to give the free base of the title compound as a yellow oil (83 mg, 29%): 1H NMR (CDCl3) δ 2.57 (m, 8H), 3.01 (s, 8H), 3.36 (s, 2H), 3.78 (s, 2H), 3.92 (s, 2H), 6.64 (d, 2H, J=8.0 Hz), 7.07 (m, 4H), 7.18 (m, 1H), 7.33 (d, 1H, J=7.7 Hz), 7.67 (m, 2H), 8.58 (d, 1H, J=4.8 Hz).
  • The free base (74 mg, 0.17 mmol) was dissolved in MeOH (3 mL) and a freshly prepared solution of saturated HBr(g) in MeOH (7 mL) was added giving a white precipitate. The mixture was stirred for 5 min and diethyl ether was added (10 mL), the solid was allowed to settle to the bottom of the flask and the supernatant solution decanted. The solid was washed by decantation with MeOH (5×5 mL) then ether (10×5 mL) and the last traces of ether were removed by evaporation in vacuo followed by drying in vacuo at 40° C. for 17.5 h to give the title compound as a white solid (153 mg, 93%): 1H NMR (DMSO-d6) δ 2.81 (br s, 4H), 3.28 (m, 8H), 3.61 (br s, 4H), 3.85 (s, 2H), 4.27 (s, 2H), 4.36 (s, 2H), 7.29 (d, 2H, J=7.7 Hz), 7.36 (d, 2H, J=7.7 Hz), 7.53 (m, 3H), 7.63 (d, 1H, J=7.7 Hz), 7.80 (t, 1H, J=7.7 Hz), 7.99 (m, 1H), 8.69 (d, 1H, J=5.3 Hz); FAB-MS m/z 527 (MH+H81Br), 525 (MH+H79Br), 445 (M+H). Anal. Calcd for C27H36N6.6HBr3H2O: C, 32.95; H, 4.92; N, 8.54; Br, 48.72. Found: C, 32.75; H, 4.89; N, 8.39; Br, 48.61.
  • EXAMPLE 29 N-[7-(4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD7051) 1,3-Phenylenebis(ethylene)diamine
  • To a solution of 1,3-phenylenediacetonitrile (9.37 g, 60 mmol) in CH3OH (saturated with NH3, 150 mL) was added Raney-Ni (ca. 20 g, previously washed with CH3OH several times) and the mixture was hydrogenated at 45 psi on a Parr apparatus for 48 h. The reaction mixture was filtered through celite and the filtrate evaporated to give the crude product (9.45 g, 96%) as a light green oil: 1H NMR (CDCl3) δ 0.80-1.50 (br s, 4H), 2.70-2.76 (m, 4H), 2.94-2.99 (m, 4H), 7.01-7.07 (m, 3H), 7.18-7.26 (m, 1H). This was used in the next step without further purification.
  • N,N′-Bis(2-Nitrobenzenesulfonyl)-1,3-Phenylenebis(ethylene)diamine
  • A solution of 2-nitrobenzenesulfonylchloride (19.94 g, 0.090 mol) in dry CH2Cl2 (70 mL) was added dropwise via cannula to a stirred solution of 1,3-phenylenebis(ethylene)diamine (4.92 g, 0.030 mol) and Et3N (16.7 mL, 0.12 mol) in dry CH2Cl2 (80 mL) under nitrogen. The reaction mixture was stirred overnight at room temperature, and then quenched with water (20 mL). The precipitate was collected by filtration and washed with H2O, CH3OH, and Et2O to give the desired product (9.22 g, 58%) as a white solid: 1H NMR (DMSO-d6) δ 2.66 (t, 4H, J=7.7 Hz), 3.08-3.18 (br s, 4H), 6.94 (d, 2H, J=6.4 Hz), 6.98 (s, 1H), 7.12 (dd, 1H, J=6.4, 6.4 Hz), 7.78-7.84 (br m, 4H), 7.90-7.64 (br m, 4H), 8.16 (br s, 2H).
  • 7-Diethoxyphosphoryl-4, 10-bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1 (17),13,15-triene
  • Using General Procedure D: Reaction of N,N′-bis(2-nitrobenzenesulfonyl)-1,3-phenylenebis(ethylene)diamine (8.74 g, 16.4 mmol) with N-(diethoxyphosphoryl)-O,O′-bis(2-methylsulfonyl)di-ethanolamine (6.50 g, 16.4 mmol) followed by silica gel column purification of the reaction products (1:15:35 CH3OH-Et2O—CH2Cl2) gave the title compound (4.03 g, 33%) as a yellow foam: 1H NMR (CDCl3) δ 1.21 (t, 6H, J=6.4 Hz), 2.39-2.46 (br m, 4H), 2.83-2.97 (br m, 8H), 3.68-3.72 (m, 4H), 3.80-3.92 (m, 4H), 7.16 (d, 2H, J=6.5 Hz), 7.18 (s, 1H), 7.24 (dd, 1H, J=6.5, 6.5 Hz), 7.60-7.74 (m, 6H), 8.04-8.08 (m, 2H).
  • 4,10-Bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-triene
  • Using General Procedure E: Reaction of 7-diethoxyphosphoryl-4,10-bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (1.27 g, 1.72 mmol) followed by silica gel column purification of the reaction products (1:15:25 CH3OH-EtOAc-CH2Cl2 then 20% CH3OH in CH2Cl2) gave the title compound (574 mg, 57%) as a light yellow foam: 1H NMR (CDCl3) δ 1.42-1.50 (br, 1H), 2.01 (t, 4H, J=5.4 Hz), 2.90-3.10 (br m, 4H), 3.08 (t, 4H, J=5.4 Hz), 3.56-3.60 (br m, 4H), 7.16 (d, 2H, J=6.8 Hz), 7.31 (dd, 1H, J=6.8, 6.8 Hz), 7.36 (s, 1H), 7.61-7.63 (m, 2H), 7.70-7.73 (m, 4H), 8.01-8.04 (m, 2H).
  • N-[7-[4,10-Bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-Nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure F: Reaction of 4,10-bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (420 mg, 0.7 mmol) with N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (302 mg, 0.7 mmol) followed by silica gel column purification of the reaction products (1:3 Et2O—CH2Cl2) gave the title compound (491 mg, 70%) as a pale yellow solid: 1H NMR (CDCl3) δ 1.97-2.02 (br m, 4H), 2.73-2.78 (br m, 4H), 2.90-2.94 (br m, 4H), 3.32 (s, 2H), 3.64-3.67 (br m, 4H), 4.55 (s, 2H), 4.58 (s, 2H), 6.93 (d, 2H, J=8.0 Hz), 7.04 (d, 2H, J=8.0 Hz), 7.09-7.16 (br m, 4H), 7.23 (s, 1H), 7.29 (dd, 1H, J=7.9, 7.9 Hz), 7.51-7.72 (m, 10H), 7.80-7.83 (m, 2H), 7.98 (d, 1H, J=7.8 Hz), 8.39 (m, 1H).
  • N-[7-(4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Pentahydrobromide Dihydrate
  • Using General Procedure G: Reaction of N-[7-[4,10-bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17), 13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (380 mg, 0.38 mmol) followed by basic alumina column purification of the reaction products (1:20 CH3OH—CH2Cl2) gave the free base of the title compound.
  • Conversion of the free base to the hydrobromide salt using a saturated solution of HBr(g) in CH3OH followed by drying in vacuo overnight, gave the title compound (110 mg, 34% overall) as a white solid: 1H NMR (DMSO-d6) δ 2.80-2.88 (br s, 4H), 3.02-3.06 (br s, 4H), 3.10-3.16 (br s, 4H), 3.38-3.44 (br s, 4H), 3.80-3.86 (br s, 2H), 4.25-4.30 (br s, 2H), 4.33-4.37 (br s, 2H), 7.27-7.32 (br m, 4H), 7.42-7.63 (br m, 6H), 7.96 (dd, 1H, J=7.7, 7.7 Hz), 8.10-8.30 (br s, 3H), 8.69 (d, 1H, J=4.9 Hz), 9.45-9.62 (br s, 2H); FAB-MS m/z 526 (MH+H81Br), 524 (MH+H79Br), 444 (M+H, 100); Anal. Calcd for C28H42N5Br5.2H2O: C, 38.03; H, 5.24; N, 7.92; Br, 45.18. Found: C, 38.37; H, 5.28; N, 7.76; Br, 45.36.
  • EXAMPLE 30 N-[1-(1,4,7-Triazacyclotetra-decanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7059) General Procedure H 4-Diethoxyphosphoryl-7-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane
  • To a stirred solution of 4-diethoxyphosphoryl-1,7-bis(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane (1.32 g, 1.87 mmol) and K2CO3 (654 mg, 4.73 mmol) in DMF (11 mL) under nitrogen was added dropwise a solution of thiophenol (0.15 mL, 1.46 mmol) in DMF (8 mL) over 1 h. The mixture was stirred for an additional 3 h and then concentrated in vacuo. The residue was partitioned between CHCl3 (50 mL) and H2O (25 mL). The aqueous phase was separated and extracted with CHCl3 (3×20 mL) and the combined organic extracts were dried (MgSO4) and concentrated in vacuo. The residue was purified by column chromatography on basic alumina (CHCl3 then 3% MeOH/CHCl3) to give the title compound as a yellow oil (178 mg, 23%): 1H NMR (CDCl3) δ 1.31 (t, 6H, J=7.0 Hz), 1.40-1.67 (m, 10H), 2.65 (m, 2H), 2.78 (m, 2H), 3.12 (m, 2H), 3.26-3.37 (m, 4H), 3.48 (m, 2H), 3.97-4.09 (m, 4H), 7.61 (m, 1H), 7.68 (m, 2H), 8.06 (m, 1H).
  • N-[1-[4-Diethoxyphosphoryl-7-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetra-decanyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure F: Reaction of 4-diethoxyphosphoryl-7-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetradecane (236 mg, 0.453 mmol) and N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (238 mg, 0.551 mmol) followed by silica gel column purification of the reaction products (50% then 80% THF/hexane) gave the title compound as a yellow amorphous solid (305 mg, 73%): 1H NMR (CDCl3) δ 1.27 (t, 6H, J=7.1 Hz), 1.43 (br s, 8H), 1.63 (br s, 2H), 2.32 (br s, 2H), 2.55 (m, 2H), 3.13-3.41 (m, 8H), 3.49 (s, 2H), 3.85-4.02 (m, 4H), 4.57 (s, 2H), 4.58 (s, 2H), 7.07-7.22 (m, 6H), 7.51-7.71 (m, 7H), 7.98 (d, 1H, J=7.4 Hz), 8.04 (m, 1H), 8.41 (d, 1H, J=4.0 Hz).
  • N-[1-[7-(2-Nitrobenzenesulfonyl)-1,4,7-triazacyclotetra-decanyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure E: Reaction of N-[1-[4-diethoxyphosphoryl-7-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetra-decanyl]-1,4-phenylenebis(methylene)]-N-
  • (2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (300 mg, 0.328 mmol) gave the title compound as a yellow amorphous solid (214 mg, 84%): 1H NMR (CDCl3) δ 1.34-1.44 (m, 8H), 1.69 (br s, 2H), 2.34 (m, 2H), 2.52 (m, 2H), 2.62 (m, 2H), 2.82 (m, 2H), 3.42 (m, 6H), 4.58 (s, 2H), 4.59 (s, 2H), 7.08-7.24 (m, 6H), 7.52-7.71 (m, 7H), 8.01 (m, 2H), 8.42 (d, 1H, J=4.1 Hz).
  • N-[1-(1,4,7-Triazacyclotetra-decanyl)-1, 4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Pentahydrobromide Dihydrate
  • A mixture of N-[1-[7-(2-nitrobenzenesulfonyl)-1,4,7-triazacyclotetra-decanyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (209 mg, 0.268 mmol), K2CO3 (298 mg, 2.16 mmol), and thiophenol (0.17 mL, 1.7 mmol) in acetonitrile (3 mL) were heated to 50° C. for 16.5 h. The reaction mixture was diluted with CH2Cl2 (10 mL) and washed with brine (10 mL). The separated aqueous phase was extracted with CH2Cl2 (3×5 mL) and the combined organic extracts were dried (MgSO4) and evaporated. The residue was purified by column chromatography on basic alumina (CHCl3 then 10% MeOH/CHCl3) to give the free base of title compound as a yellow oil (92 mg, 84%): 1H NMR (CDCl3) δ 1.21-1.62 (m, 10H), 2.40-2.49 (m, 4H), 2.60 (m, 6H), 2.79 (m, 2H), 3.49 (s, 2H), 3.80 (s, 2H), 3.91 (s, 2H), 7.14 (m, 1H), 7.28 (m, 4H), 7.62 (m, 6H), 8.54 (d, 1H, J=4.4 Hz).
  • Conversion of the free base (86 mg, 0.21 mmol) to the hydrobromide salt using a saturated solution of HBr(g) in MeOH (See General Procedure G) followed by drying in vacuo at 40° C. for 15.5 h gave the title compound as a white solid (128 mg, 70%): 1H NMR (D2O) δ 1.48 (br s, 6H), 1.82 (m, 4H), 3.22-3.36 (m, 10H), 3.50 (br s, 2H), 4.48 (s, 4H), 4.64 (s, 2H), 7.62 (s, 4H), 7.88 (m, 1H), 7.94 (d, 1H, J=8.0 Hz), 8.38 (m, 1H), 8.77 (d, 1H, J=5.2 Hz); FAB-MS m/z 492 (MH+H81Br), 490 (M+H79Br), 410 (M+H). Anal. Calcd for C25H39N5.5HBr2.5H2O.0.1Et2O: C, 35.20; H, 5.82; N, 8.08; Br, 46.10. Found: C, 35.48; H, 5.66; N, 8.10; Br, 46.05.
  • EXAMPLE 31 N-[4-[4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7063) 7-Diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-(17),13,15-triene
  • Using General Procedure H: Reaction of 7-diethoxyphosphoryl-4,10-bis(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (1.48 g, 2.00 mmol) with thiophenol (with an additional heating of the reaction mixture to 50° C. for 1.5 h after the addition) followed by silica gel column purification of the reaction products (8% MeOH/CHCl3) gave the title compound as a light yellow oil (423 mg, 52%): 1H NMR (CDCl3) δ 1.23 (t, 6H, J=7.0 Hz), 2.50 (br s, 2H), 2.79 (br s, 2H), 3.02-3.15 (m, 10H), 3.82-3.98 (m, 6H), 7.06 (d, 2H, J=7.6 Hz), 7.54-7.63 (m, 2H), 7.70 (m, 2H), 8.01 (br s, 1H).
  • N-[4-[7-Diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1 (17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure F: Reaction of 7-diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (410 mg, 0.738 mmol) and N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (397 mg, 0.919 mmol) followed by silica gel column purification of the reaction products (50%, 80%, and 90% THF/hexane) gave the title compound as a white amorphous solid (441 mg, 63%): 1H.NMR (CDCl3) δ 1.15 (t, 6H, J=7.0 Hz), 2.42 (m, 4H), 2.77 (m, 2H), 2.92-3.02 (m, 6H), 3.10 (m, 2H), 3.59 (s, 2H), 3.66-3.91 (m, 6H), 4.58 (s, 2H), 4.59 (s, 2H), 6.94 (d, 1H, J=7.6 Hz), 7.07-7.14 (m, 6H), 7.22 (d, 1H, J=7.8 Hz), 7.51-7.72 (m, 8H), 8.00 (d, 1H, J=7.8 Hz), 8.04 (m, 1H), 8.42 (d, 1H, J=4.0 Hz).
  • N-[4-[7-Diethoxyphosphoryl-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-2-(aminomethyl)pyridine
  • A mixture of N-[4-[7-diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (434 mg, 0.456 mmol), K2CO3 (508 mg, 3.68 mmol), and thiophenol (0.28 mL, 2.7 mmol) were heated to 50° C. in CH3CN (3.5 mL) under nitrogen for 15 h. Upon cooling, the reaction mixture was partitioned between CHCl3 (15 mL) and brine (15 mL) and the aqueous layer was separated and extracted with CHCl3 (3×5 mL). The combined organic extracts were dried (MgSO4) and concentrated in vacuo and the residue was purified by column chromatography on basic alumina (CHCl3 then 10% MeOH/CHCl3) to give a yellow oil (218 mg, 82%): 1H NMR (CDCl3) δ 1.16 (t, 6H, J=7.1 Hz), 2.35 (m, 2H), 2.55 (m, 2H), 2.75 (m, 2H), 2.82 (m, 2H), 2.96-3.08 (m, 6H), 3.16 (m, 2H), 3.68 (s, 2H), 3.69-3.88 (m, 4H), 3.82 (s, 2H), 3.93 (s, 2H), 6.95 (d, 1H, J=7.6 Hz), 7.00 (d, 1H, J=7.5 Hz), 7.15-7.34 (m, 6H), 7.50 (m, 1H), 7.65 (m, 1H), 8.56 (d, 1H, J=4.7 Hz).
  • N-[4-[4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienlyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Hexahydrobromide Hydrate
  • To a stirred solution of N-[4-[7-diethoxyphosphoryl-4,7,10,17-tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-2-(aminomethyl)pyridine (211 mg, 0.36 mmol) in acetic acid (0.6 mL) was added a freshly prepared solution of saturated HBr(g) in acetic acid (6 mL) and the reaction mixture was allowed to stir at room temperature for 4 h. Addition of diethyl ether (10 mL) gave a white precipitate that was allowed to settle to the bottom of the flask and the supernatant solution was decanted. The solid was washed by decantation with MeOH (4×5 mL) and ether (6×5 mL) and the remaining traces of ether were removed by evaporation under reduced pressure. The product was dried in vacuo at 40° C. for 17 h, to give the title compound as a pale yellow solid (223 mg, 63%): 1H NMR (D2O) δ 3.14-3.36 (m, 10H), 3.55 (m, 4H), 3.75 (m, 2H), 4.45 (s, 2H), 4.50 (s, 2H), 4.64 (s, 2H), 7.22 (m, 2H), 7.53 (s, 4H), 7.70 (m, 1H), 7.95 (m, 1H), 8.00 (d, 1H, J=7.9 Hz), 8.46 (m, 1H), 8.79 (d, 1H, J=3.9 Hz); FAB-MS m/z 527 (MH+H81Br), 525 (MH+H79Br), 445 (M+H). Anal. Calcd for C27H36N6.6HBr.1.5H2O.0.2Et2O: C, 34.35; H, 4.87; N, 8.65; Br, 49.33. Found: C, 34.57; H, 5.04; N, 8.68; Br, 49.09.
  • EXAMPLE 32 N-[4-[4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD7058) 7-Diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1 (17),13,15-triene
  • Using General Procedure H: Reaction of 7-diethoxyphosphoryl-4,10-bis(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-triene
  • (1.11 g, 1.5 mmol) followed by silica gel column purification of the reaction products (2:5:20 CH3OH-Et2O—CH2Cl2 then 1:5 CH3OH—CH2Cl2) gave the title compound as a pale yellow oil (300 mg, 54%): 1H NMR (CDCl3) δ 1.21 (t, 6H, J=7.1 Hz), 1.78-1.92 (br s, 1H), 2.31-2.38 (br m, 2H), 2.56-2.60 (br m, 2H), 2.81-2.98 (br m, 10H), 3.60-3.64 (br m, 2H), 3.75-3.91 (m, 4H), 7.05-7.12 (m, 2H), 7.24-7.29 (m, 2H), 7.60-7.63 (m, 1H), 7.68-7.71 (m, 2H), 8.02-8.06 (m, 11H).
  • N-[4-[7-Diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure F: Reaction of 7-diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-triene (290 mg, 0.52 mmol) with N-[1-methylene-4-(chloromethylene)phenylene]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (271 mg, 0.63 mmol) followed by silica gel column purification of the reaction products (1:12:12 CH3OH-Et2O—CH2Cl2) gave the title compound (298 mg, 60%) as a pale yellow solid: 1H NMR (CDCl3) δ 1.17 (t, 6H, J=7.0 Hz), 2.29-2.45 (br m, 4H), 2.55-2.65 (br m, 2H), 2.71-2.75 (br s, 4H), 2.85-2.91 (br m, 2H), 2.96-2.98 (br m, 2H), 3.57 (s, 2H), 3.67-3.84 (br m, 6H), 4.57-4.61 (br s, 4H), 7.07-7.28 (br m, 10H), 7.55-7.71 (br m, 7H), 7.99-8.02 (m, 2H), 8.42-8.46 (m, 1H).
  • N-[4-[10-(2-Nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine
  • Using General Procedure E: Reaction of N-[4-[7-diethoxyphosphoryl-10-(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (290 mg, 0.31 mmol) gave the title compound (240 mg, 95%) as a white solid: 1H NMR (CDCl3) δ 1.65-1.79 (br s, 1H, coincide with H2O peak), 2.15-2.19 (br m, 4H), 2.44-2.48 (br m, 2H), 2.61-2.65 (br m, 2H), 2.67-2.71 (br m, 2H), 3.00-3.04 (br m, 2H), 3.10-3.14 (br m, 2H), 3.56-3.60 (br s, 4H), 4.55 (s, 2H), 4.61 (s, 2H), 6.96 (d, 1H, J=7.8 Hz), 7.02-7.10 (br m, 6H), 7.22-7.28 (br m, 3H), 7.52-7.72 (br m, 7H), 7.96-7.99 (m, 2H), 8.42-8.46 (m, 1H). This was used without further purification.
  • N-[4-[4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine Pentahydrobromide Dihydrate
  • Using General Procedure G: Reaction of N-[4-[10-(2-nitrobenzenesulfonyl)-4,7,10-triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-N-(2-nitrobenzenesulfonyl)-2-(aminomethyl)pyridine (236 mg, 0.29 mmol) followed by alumina column purification of the reaction products (1:99 CH3OH—CH2Cl2 then 1:10 CH3OH—CH2Cl2) gave the free base of the title compound (111 mg, 86%) as a pale yellow oil: 1H NMR (CDCl3) δ 2.24-2.28 (br s, 3H), 2.43-2.50 (br m, 4H), 2.58-2.62 (br m, 2H), 2.73-2.79 (br m, 8H), 2.95-2.98 (br m, 2H), 3.50 (s, 2H), 3.77 (s, 2H), 3.90 (s, 2H), 6.83-6.87 (br m, 3H), 7.05-7.33 (br m, 7H), 7.63-7.67 (m, 1H), 8.54-8.56 (m, 1H).
  • Conversion of the free base (104 mg, 0.23 mmol) to the hydrobromide salt using a saturated solution of HBr(g) in CH3OH followed by drying of the product in vacuo, gave the title compound (101 mg, 52%) as a white solid: 1H NMR (D2O) δ 2.90-2.94 (br m, 2H), 2.97-3.01 (br m, 2H), 3.12-3.16 (br m, 2H), 3.17-3.21 (br m, 2H), 3.24-3.28 (br m, 4H), 3.47-3.51 (br m, 2H), 3.57-3.61 (br m, 2H), 4.38-4.42 (m, 6H), 7.34-7.40 (m, 2H), 7.46-7.60 (m, 8H), 7.90-7.94 (m, 1H), 8.58-8.62 (m, 1H); FAB-MS m/z 526 (MH+H81Br), 524 (MH+H79Br), 444 (M+H, 100); Anal. Calcd. for C28H42N5Br5.2.5H2O: C, 37.65; H, 5.30; N, 7.84; Br, 44.73. Found: C, 37.53; H, 5.26; N, 7.79; Br, 44.75.
    TABLE 2
    Inhibition of mAb 12G5 binding
    Compound IC50a (ng/ml)
    AMD3100 27
    AMD3465 3
    AMD7049 52
    AMD7050 1
    AMD7051 7
    AMD7058 >1000
    AMD7059 >1000
    AMD7063 9
    SDF-1αb 270

    aInhibition of mAb 12G5 binding to CXCR-4 in SUP-T1 cells.

    bNatural ligand for CXCR-4 (Bleul et al. Nature, 382: 829-832 (1996); Oberlin et al., Nature, 382: 833-835 (1996)).
  • TABLE 3
    % Inhibition of Ca2+ flux (conc)
    Compound or IC50 a (ng/ml)
    AMD3100 5
    AMD3465 1
    AMD7049 100% (1 μg/ml)
    AMD7050 100% (1 μg/ml)
    AMD7051 100% (1 μg/ml)
    AMD7058  44% (1 μg/ml)
    AMD7059  36% (1 μg/ml)
    AMD7063 100% (1 μg/ml)

    aInhibition of Signal transduction (increasing intracellular Ca2+ flux) induced by SDF-1α binding to CXCR-4 on SUP-T1 cells.
  • Each of the following compounds, including AMD 3484 (see FIG. 28), were synthesized according to procedures in Bridger et al., J Med. Chem. 1995, 38, 366-378; J. Med. Chem. 1996, 39, 109-119 and U.S. Pat. No. 5,583,131, U.S. Pat. No. 5,698,546 and U.S. Pat. No. 5,817,807, which are each incorporated in their entirety by reference herein.
  • EXAMPLE 33 1-[2,6-Dimethoxypyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Tetrahydrobromide (AMD 7032)
  • 1H NMR (D2O) δ 1.78 (m, 2H), 1.88-1.92 (m, 2H), 2.59-3.03 (m, 16H), 3.60 (s, 2H), 3.91 (s, 6H), 6.44 (s, 2H); 13C NMR (D2O) δ 26.75, 27.91, 48.34, 49.21, 49.89, 50.96, 52.01, 52.86, 54.88, 57.15, 57.53, 59.42, 142.65, 157.42, 166.42; FAB MS m/z 352 (M+H); Anal. (C18H33N5O2 4.1 HBr 0.25H2O); Calc. C, 31.44; H, 5.51; N, 10.18; Br, 47.64. Found C, 31.17; H, 5.61; N, 9.92; Br, 47.54.
  • EXAMPLE 34 1-[2-Chloropyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Tetrahydrochloride Monohydrate (AMD 7048)
  • 1HNMR(D2O) δ 1.92 (m, 2H), 2.12 (m, 2H), 2.77-2.80 (m, 4H), 2.96-3.39 (m, 12H), 3.85 (s, 2H), 7.33 (d, 1H, J=5.4 Hz), 7.44 (s, 1H), 8.40 (d, 1H, J=5.4 Hz); 13C NMR (D2O) δ 24.75, 27.59, 47.40, 47.55, 49.11, 49.23, 52.12, 52.40, 53.81, 54.42, 56.98, 126.97, 128.30, 151.90, 152.34, 153.78; FAB MS m/z 326 (M+H); Anal. (C16H28N5Cl.4.2HCl.0.5HOAc.1.1H2O); Calc. C, 38.61; H, 6.94; N, 13.24; Cl, 34.86. Found C, 38.63; H, 6.94; N, 13.52; Cl, 34.61.
  • EXAMPLE 35 1-[2,6-Dimethylpyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Pentahydrobromide Dihydrate (AMD 7060)
  • 1H NMR (D2O) δ 1.77 (m, 2H), 1.93 (m, 2H), 2.48 (s, 6H), 2.61-3.00 (m, 16H), 3.61 (s, 2H), 7.07 (s, 2H); 13C NMR (D2O) δ 25.30, 26.22, 27.49, 47.75, 48.65, 49.43, 50.41, 51.58, 52.19, 54.09, 56.63, 58.46; FAB MS m/z 320 (M+H); Anal. (C18H33N5.5HBr.0.5HOAc.1.7H2O); Calc. C, 29.08; H, 5.57; N, 8.92; Br, 50.91. Found C, 29.04; H, 5.60; N, 8.73; Br, 50.87.
  • EXAMPLE 36 1-[2-Methylpyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Pentahydrobromide Dihydrate (AMD 7061)
  • 1H NMR (D2O) δ 2.01-2.08 (m, 2H), 2.22 (m, 2H), 2.70-2.72 (m, 2H), 2.77 (s, 3H), 2.91-2.92 (m, 2H), 3.33-3.52 (m, 12H), 4.00 (s, 2H), 7.86-7.89 (m, 2H), 8.56 (d, 1H, J=5.7 Hz); FAB MS m/z 306 (M+H); Anal. (C17H31N5.4.9HBr.0.3HOAc.2.1H2O); Calc. C, 27.9; H, 5.49; N, 9.24; Br, 51.67. Found C, 28.08; H, 5.50; N, 9.56; Br, 51.56.
  • EXAMPLE 37 1-[2,6-Dichloropyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Trihydrochloride Bishydrate (AMD 3451)
  • 1H NMR (D2O) δ 1.83-1.88 (m, 2H), 2.04-2.08 (m, 2H), 2.58-2.62 (m, 2H), 2.79-2.81 (m, 2H), 3.12-3.44 (m, 12H), 3.69 (s, 2H), 7.30 (s, 2H); 13C NMR (D2O) □36.26, 37.69, 55.26, 56.18, 58.33, 58.56, 58.92, 59.23, 63.57, 65.44, 70.72, 140.37, 166.14, 167.37; FAB MS m/z 360 (M+H). Anal. (C16H34N5Cl5O2): Calc. C, 38.00;H, 6.78; N, 13.85; Cl, 35.05. Found: C, 38.33; H, 6.42; N, 13.88; Cl, 35.43.
  • EXAMPLE 38 1-[2-Chloropyrid-5-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane Tetrahydrochloride Hemihydrate (AMD 3454)
  • 1H NMR (D2O) δ 1.96-2.09 (br m, 4H), 3.02-3.17 (m, 4H), 3.19-3.28 (br m, 8H), 3.40 (s, 4H), 4.10 (s, 2H), 7.40 (d, 1H, J=8.2 Hz), 7.80 (d, 1H, J=8.2 Hz), 8.27 (s, 1H); 13C NMR (D2O) δ 19.36, 19.47, 38.17, 38.64, 39.06, 41.74, 41.88, 42.18, 45.66, 48.29, 54.62, 125.59, 126.69, 142.79, 150.77, 151.75; FAB-MS m/z 326 (M+H). Anal. Calcd for C16H28N5Cl.4HCl.0.5H2O: C, 39.98; H, 6.92; N, 14.57; Cl, 36.87. Found: C, 40.36; H, 7.06; N, 14.56; Cl, 36.92.
  • General Procedures A, B and C were used to prepare the following compounds:
  • EXAMPLE 39 N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-purine Pentahydrobromide Dihydrate (AMD3472)
  • 1H NMR (D2O) δ 1.88-2.05 (br m, 4H), 3.06-3.22 (br m, 8H), 3.27-3.44 (br m, 8H), 4.22 (s, 2H), 5.59 (s, 2H), 7.29 (s, 4H), 8.80 (s, 1H), 9.11 (s, 1H), 9.28 (s, 1H); 13C NMR (D2O) δ 18.39, 19.25, 37.24, 37.55, 37.71, 41.13, 41.37, 41.71, 44.41, 47.73, 54.87, 129.45, 131.81, 132.53, 136.67, 140.96, 147.88, 152.46, 154.37; FAB-MS m/z 423 (M+H). Anal. Calcd for C23H34N8.5HBr.2H2O.0.5CH3CO2H: C, 32.27; H, 5.07; N, 12.54; Br, 44.73. Found: C, 32.66; H, 4.81; N, 12.41; Br, 44.58.
  • EXAMPLE 40 1-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-phenylpiperazine Pentahydrobromide Hydrate (AMD3526)
  • 1H NMR (D2O) δ 1.88-2.06 (br m, 4H), 3.11-3.53 (br m, 24H), 4.30 (s, 2H), 4.32 (s, 2H), 6.89-6.97 (m, 3H), 7.19-7.24 (m, 2H), 7.49 (s, 4H); 13C NMR (D2O) δ 18.74, 19.37, 37.34, 41.47, 41.76, 42.03, 44.31, 47.45, 48.26, 51.16, 58.48, 59.29, 118.18, 122.34, 129.99, 130.37, 131.53, 131.85, 132.62, 148.47; FAB-MS m/z 547 (M+H81Br), 545 (M+H79Br), 465 (M+H). Anal. Calcd for C28H44N6.5HBr.H2O: C, 37.90; H, 5.79; N, 9.47; Br, 45.03. Found: C, 37.72; H, 5.98; N, 9.38; Br, 46.78.
    TABLE 4
    Compound IC50 a (μg/mL)
    AMD3451 8.9
    AMD3472 45.4
    AMD3454 32.3
    AMD3526 82
    AMD3100 >100

    a50% Inhibitory Concentration (IC50)(μg/mL) exhibited by AMD compounds against infection of U87.CD4.CCR5 by HIV-1 BaL--

Claims (3)

1. A method to treat atherosclerosis comprising administering to a subject in need of such treatment a compound selected from the group consisting of:
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-methyl)pyridine (AMD 3465);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-N-methyl-2-(aminomethyl)pyridine (AMD. 3538);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-(amino-methyl)pyridine (AMD 3500);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-3-(amino-methyl)pyridine (AMD 3499);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-(2-amino-methyl-5-methyl)pyrazine (AMD 3498);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-ethyl)pyridine (AMD 3497);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-methyl) thiophene (AMD 3516);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-(amino-ethyl)mercaptan (AMD 3530);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-2-amino-benzylamine (AMD 3517);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-amino-benzylamine (AMD 3544);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-(amino-ethyl)imidazole (AMD 3543);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-benzylamine (AMD 3529);
N-[4-(1,4,7-Triazacyclotetra-decanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7049);
N-[7-(4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7050);
N-[7-(4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7051);
N-[4-[4,7,10-Triazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7058);
N-[1-(1,4,7-Triazacyclotetra-decanyl)-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7059);
N-[4-[4,7,10,17-Tetraazabicyclo[13.3.1]heptadeca-1(17),13,15-trienyl]-1,4-phenylenebis(methylene)]-2-(aminomethyl)pyridine (AMD 7063);
1-[2,6-Dichloropyrid-4-yl(methylene)]-1,4,8,11-tetraazacyclotetradecane (AMD 3451);
N-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-purine (AMD 3472); and
1-[1,4,8,11-Tetraazacyclotetradecanyl-1,4-phenylenebis(methylene)]-4-phenylpiperazine (AMD3526);
or a salt form thereof.
2. The method of claim 1, wherein said subject is a human.
3. The method of claim 1, wherein said subject is a mammal.
US11/599,066 1999-12-17 2006-11-13 Chemokine receptor binding heterocyclic compounds Abandoned US20070060591A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/599,066 US20070060591A1 (en) 1999-12-17 2006-11-13 Chemokine receptor binding heterocyclic compounds

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US17215399P 1999-12-17 1999-12-17
US09/740,050 US6667320B2 (en) 1999-12-17 2000-12-15 Chemokine receptor binding heterocyclic compounds
US10/703,781 US7022717B2 (en) 1999-12-17 2003-11-07 Chemokine receptor binding heterocyclic compounds
US11/281,296 US20060069129A1 (en) 1999-12-17 2005-11-16 Chemokine receptor binding heterocyclic compounds
US11/599,066 US20070060591A1 (en) 1999-12-17 2006-11-13 Chemokine receptor binding heterocyclic compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/281,296 Continuation US20060069129A1 (en) 1999-12-17 2005-11-16 Chemokine receptor binding heterocyclic compounds

Publications (1)

Publication Number Publication Date
US20070060591A1 true US20070060591A1 (en) 2007-03-15

Family

ID=22626583

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/740,050 Expired - Lifetime US6667320B2 (en) 1999-12-17 2000-12-15 Chemokine receptor binding heterocyclic compounds
US10/703,781 Expired - Lifetime US7022717B2 (en) 1999-12-17 2003-11-07 Chemokine receptor binding heterocyclic compounds
US11/281,296 Abandoned US20060069129A1 (en) 1999-12-17 2005-11-16 Chemokine receptor binding heterocyclic compounds
US11/599,066 Abandoned US20070060591A1 (en) 1999-12-17 2006-11-13 Chemokine receptor binding heterocyclic compounds

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/740,050 Expired - Lifetime US6667320B2 (en) 1999-12-17 2000-12-15 Chemokine receptor binding heterocyclic compounds
US10/703,781 Expired - Lifetime US7022717B2 (en) 1999-12-17 2003-11-07 Chemokine receptor binding heterocyclic compounds
US11/281,296 Abandoned US20060069129A1 (en) 1999-12-17 2005-11-16 Chemokine receptor binding heterocyclic compounds

Country Status (10)

Country Link
US (4) US6667320B2 (en)
EP (1) EP1244648B1 (en)
JP (1) JP2003516984A (en)
CN (1) CN100335478C (en)
AT (1) ATE428706T1 (en)
AU (1) AU2334301A (en)
CA (1) CA2389545C (en)
DE (1) DE60042030D1 (en)
HK (1) HK1046409A1 (en)
WO (1) WO2001044229A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012158707A1 (en) 2011-05-16 2012-11-22 Genzyme Corporation Use of cxcr4 antagonists

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL364673A1 (en) * 2001-07-31 2004-12-13 Anormed Inc. Methods to mobilize progenitor/stem cells
US7169750B2 (en) 2001-07-31 2007-01-30 Anormed, Inc. Methods to mobilize progenitor/stem cells
AU2003249999A1 (en) * 2002-07-10 2004-02-02 Hans Robert Kalbitzer 1,4,7,10-tetraazacyclododecanes as modulators of the guanine-binding protein for treating tumours
AU2005272653A1 (en) * 2004-08-13 2006-02-23 Anormed Inc. Chemokine combinations to mobilize progenitor/stem cells
FR2875410B1 (en) * 2004-09-23 2012-11-16 Guerbet Sa DIAGNOSTIC COMPOUNDS FOR THE TARGETING OF CHEMOKINE RECEPTOR
WO2007022523A2 (en) 2005-08-19 2007-02-22 Genzyme Corporation Methods to enhance chemotherapy
EP1993584B1 (en) 2006-02-02 2012-05-30 Allergan, Inc. Inhibitors of CXCR4 activity for use in the treatment of ocular disorders
CN101389329A (en) 2006-02-24 2009-03-18 健赞股份有限公司 Method for increasing blood and/or promoting organic regeneration
AU2007281090A1 (en) * 2006-08-02 2008-02-07 Genzyme Corporation Combination therapy
AR062271A1 (en) * 2006-08-07 2008-10-29 Genzyme Corp USE OF AN EFFECTIVE AMOUNT OF AT LEAST A CXCR4 INHIBITOR, AT LEAST A CXCR2 AND G-CSF AGONIST TO MOBILIZE PROGENITOR CELLS AND / OR MOTHER CELLS
US10933049B2 (en) 2015-06-03 2021-03-02 The University Of Queensland Mobilizing agents and uses therefor
US20180296551A1 (en) * 2015-10-19 2018-10-18 Ramot At Tel-Aviv University Ltd. Methods and compositions for treating neurodegenerative diseases
US11433048B2 (en) 2016-06-16 2022-09-06 Centre National De La Recherche Scientifique CXCR4 receptor-binding compounds useful for increasing interferon level
WO2019089833A1 (en) 2017-10-31 2019-05-09 Magenta Therapeutics Inc. Compositions and methods for hematopoietic stem and progenitor cell transplant therapy
WO2019089826A1 (en) 2017-10-31 2019-05-09 Magenta Therapeutics Inc. Compositions and methods for the expansion of hematopoietic stem and progenitor cells
WO2019113375A2 (en) 2017-12-06 2019-06-13 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progenitor cells
US10058573B1 (en) 2017-12-06 2018-08-28 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem cells
US11260079B2 (en) 2017-12-06 2022-03-01 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progenitor cells
CA3087527A1 (en) 2018-01-03 2019-07-11 Magenta Therapeutics, Inc. Compositions and methods for the expansion of hematopoietic stem and progenitor cells and treatment of inherited metabolic disorders
WO2021087406A1 (en) 2019-11-01 2021-05-06 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progentor cells
MX2022013493A (en) 2020-04-27 2023-02-22 Magenta Therapeutics Inc METHODS AND COMPOSITIONS FOR TRANSDUCING HEMATOPOIETIC STEM AND PROGENITOR CELLS <i>IN VIVO.
EP4308694A1 (en) 2021-03-16 2024-01-24 Magenta Therapeutics, Inc. Dosing regimens for hematopoietic stem cell mobilization for stem cell transplants in multiple myeloma patients

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5021409A (en) * 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
US5583131A (en) * 1991-12-16 1996-12-10 Johnson Matthey Public Limited Company Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity
US5698546A (en) * 1994-01-11 1997-12-16 Johnson Matthey Public Limted Company Cyclic polyamines
US5817807A (en) * 1995-06-06 1998-10-06 Anormed Inc. Antiviral compounds
US6365583B1 (en) * 1999-02-02 2002-04-02 Anormed, Inc. Methods to enhance white blood cell count
US20040209837A1 (en) * 1998-03-24 2004-10-21 Chugai Seiyaku Kabushiki Kaisha Vascularization inhibitors
US20060194776A1 (en) * 2003-06-30 2006-08-31 Caritas St. Elizabeth Medical Center Of Boston, Inc. Compositions and methods for treating tissue ischemia

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9105489D0 (en) 1991-03-15 1991-05-01 Johnson Matthey Plc Improvements in chemical compounds
US6506770B1 (en) 1996-06-06 2003-01-14 Anormed, Inc. Antiviral compounds
AU4692400A (en) * 1999-05-03 2000-11-17 Smithkline Beecham Corporation Cxcr-4 receptor antagonists - thrombopoietin mimetics

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5021409A (en) * 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
US5583131A (en) * 1991-12-16 1996-12-10 Johnson Matthey Public Limited Company Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity
US5698546A (en) * 1994-01-11 1997-12-16 Johnson Matthey Public Limted Company Cyclic polyamines
US5817807A (en) * 1995-06-06 1998-10-06 Anormed Inc. Antiviral compounds
US20040209837A1 (en) * 1998-03-24 2004-10-21 Chugai Seiyaku Kabushiki Kaisha Vascularization inhibitors
US6365583B1 (en) * 1999-02-02 2002-04-02 Anormed, Inc. Methods to enhance white blood cell count
US20060194776A1 (en) * 2003-06-30 2006-08-31 Caritas St. Elizabeth Medical Center Of Boston, Inc. Compositions and methods for treating tissue ischemia

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012158707A1 (en) 2011-05-16 2012-11-22 Genzyme Corporation Use of cxcr4 antagonists

Also Published As

Publication number Publication date
US20060069129A1 (en) 2006-03-30
US6667320B2 (en) 2003-12-23
CN100335478C (en) 2007-09-05
HK1046409A1 (en) 2003-01-10
CA2389545C (en) 2010-08-31
JP2003516984A (en) 2003-05-20
US20020077339A1 (en) 2002-06-20
DE60042030D1 (en) 2009-05-28
EP1244648A1 (en) 2002-10-02
CA2389545A1 (en) 2001-06-21
US7022717B2 (en) 2006-04-04
US20040102428A1 (en) 2004-05-27
AU2334301A (en) 2001-06-25
EP1244648B1 (en) 2009-04-15
ATE428706T1 (en) 2009-05-15
WO2001044229A1 (en) 2001-06-21
CN1411456A (en) 2003-04-16

Similar Documents

Publication Publication Date Title
US20070060591A1 (en) Chemokine receptor binding heterocyclic compounds
US7091217B2 (en) Chemokine receptor binding heterocyclic compounds
ES2325909T3 (en) HETEROCICLICAL COMPOUNDS THAT JOIN THE CHEMIOKIN RECEPTORS.
US7312234B2 (en) Chemokine receptor binding heterocyclic compounds
US20070066624A1 (en) Chemokine receptor binding compounds
AU2001294628A1 (en) Chemokine receptor binding heterocyclic compounds
AU2001291569A1 (en) Chemokine receptor binding heterocyclic compounds
US20050277668A1 (en) Chemokine receptor binding compounds
US20050059702A1 (en) Chemokine receptor binding heterocyclic compounds with enhanced efficacy
JP2004512336A (en) Chemokine receptor binding heterocyclic compounds
RU2297413C2 (en) Heterocyclic compounds modulating activity of chemokine receptor, their using and pharmaceutical composition containing thereof
RU2277092C2 (en) Heterocyclic compounds, their using and pharmaceutical composition for treatment of states mediated by cxcr4 and ccr5
MXPA03002327A (en) Chemokine receptor binding heterocyclic compounds.

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENZYME CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ANORMED CORPORATION;REEL/FRAME:020518/0053

Effective date: 20080109

Owner name: GENZYME CORPORATION,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ANORMED CORPORATION;REEL/FRAME:020518/0053

Effective date: 20080109

STCB Information on status: application discontinuation

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