USRE42152E1 - Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity - Google Patents

Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity Download PDF

Info

Publication number
USRE42152E1
USRE42152E1 US12/192,704 US19270492A USRE42152E US RE42152 E1 USRE42152 E1 US RE42152E1 US 19270492 A US19270492 A US 19270492A US RE42152 E USRE42152 E US RE42152E
Authority
US
United States
Prior art keywords
bis
methylene
tetraazacyclotetradecane
admixture
association
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.)
Expired - Lifetime, expires
Application number
US12/192,704
Inventor
Gary J. Bridger
Sreenivasan Padmanbhan
Renato Skerlj
David M. Thornton
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
Genzyme Corp
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=10706343&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE42152(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Genzyme Corp filed Critical Genzyme Corp
Application granted granted Critical
Publication of USRE42152E1 publication Critical patent/USRE42152E1/en
Assigned to GENZYME CORPORATION reassignment GENZYME CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANORMED CORPORATION
Assigned to ANORMED INC. reassignment ANORMED INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JOHNSON MATTHEY PUBLIC LIMITED COMPANY
Assigned to JOHNSON MATTHEY PUBLIC LIMITED COMPANY reassignment JOHNSON MATTHEY PUBLIC LIMITED COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THORNTON, DAVID MICHAEL, BRIDGER, GARY JAMES, PADMANAMHAN, SREENIVASAN, SKERLJ, RENATO TONY
Assigned to ANORMED CORPORATION reassignment ANORMED CORPORATION MERGER AND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ANORMED INC., DEMATAL CORP.
Adjusted expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • This invention concerns improvements in chemical compounds, more especially it concerns compounds and pharmaceutical compositions. In particular it concerns compositions and compounds having activity in in vitro tests on Human Immunodeficiency Virus-infected cells.
  • AIDS Acquired Immune Deficiency Syndrome
  • chemo-therapeutic treatments have been advocated, and some compounds have emerged as a potential basis for treatment, there is still a need for alternatives.
  • most treatments such as the compound known as AZT have a high toxicity to cells, and it would be desirable to find compounds which are less toxic. In man, the development of resistance to AZT has been identified as an additional clinical problem.
  • the present invention provides the use of compounds defined below, in phamaceutical compositions for treating HIV-infected patients.
  • the invention further provides pharmaceutical compositions comprising a said compound in combination or association with a pharmaceutically acceptable diluent or excipient, for the treatment of HIV-infected patents.
  • the invention may also be defined as the use of a said compound for the manufacture of a medicament for the treatment of HIV-infected patients.
  • the invention further provides a process for the production of a pharmaceutical composition for the treatment of a HIV-infected patient, comprising the combination of a compound as defined below with a pharmaceutically acceptable diluent or excipient, and formulating said composition into a form suitable far administration to said patient.
  • the invention also provides a method of treatment of an HIV-infected patient, comprising administering to said patient an effective dose of a said compound. It is to be understood that treatment includes prophylactic treatment of patients at risk, in view of the protective properties observed.
  • the use of the compounds may also be stated as a method of treating HIV-infected or HIV-challenged human cells to prevent or modulate the multiplication of the HIV, comprising administering to said cells an effective dose of a said compound. Whilst this description is especially directed to combating HIV, this invention includes other aspects in which other diseases may be treated, including for example microbial infections.
  • U.S. Pat. No. 4,156,683 discloses monocyclic and bicyclic macrocyclic compounds, which axe are said to have biological activity in regulating sodium, potassium and calcium levels in mammals. Additionally, a specific group of N-alkylated monocyclic compounds are said to possess activity against A 2 influenza viruses in a modified Hermann test on chick fibroblast tissue. It is also said that the preferred compounds, which form complexes of greater stability, are those having three bridging chains between bridgehead nitrogen atoms, that is fused bicyclic compounds.
  • EP-A-0296522 discloses certain functionally modified cyclic polyamines, including that known as “cyclam”, which complexes with rhodium and may be bound to an antibody or antibody fragment
  • cyclam which complexes with rhodium and may be bound to an antibody or antibody fragment
  • the aromatic-linked cyclic polyamines which form the subject of the present invention are not disclosed, nor is any anti-vital activity.
  • EP-A-0305320 also discloses several modified cyclic polyamines, but does not disclose identical cyclic polyamines linked together.
  • WO-A-9105762 discloses polyamines useful for their multi-point chelating activity, but does not disclose linked cyclic polyamines.
  • WO-A-9216494 is in the same name as the present applicants, and discloses long-chain polyamines, optionally linked to a cyclic polyamine, as agents active against HIV. No molecules having two cyclic polyamines, linked through an aromatic linker are disclosed in this prior art.
  • the present invention provides as active compounds linked cyclic compounds of the general formula I Z-R-A-R′-Y (I) in which Z and Y are identical cyclic polyamine moieties having from 9 to 20 ring members and from 3 to 6 amine nitrogens in the ring spaced by 2 or more carbon atoms from each other,
  • the cyclic polyamine moieties may be substituted or unsubstituted, and suitable substituents are alkyl and/or aryl groups, eg of up to 10 carbon atoms, and any other atoms or groups which do not substantially adversely affect the activity or toxicity of the compounds.
  • Preferred moieties are those of 10 to 15 ring members, and there are preferably 3 or 4 amine nitrogen atoms.
  • the aromatic or heteroaromatic moiety A tethers Y and Z through the linking groups R and R′.
  • Moiety A may be phenyl or fused aromatic such as napthyl, heterocyclic such as pyridyl or thiophenyl, fused heterocyclic or joined aromatic and/or joined heteroaromatic, for example biphenyl or bipyridyl respectively.
  • the moieties A may also be substituted at single or multiple non-linking positions with electron-donating groups, eg alkyl, thio, thioalkyl, hydroxyl, alkoxyl, amino and derivatives thereof, or electron-withdrawing groups or atoms, eg nitro, halogen, carboxy, carboxamido, sulfonic acid and derivatives thereof.
  • electron-donating groups eg alkyl, thio, thioalkyl, hydroxyl, alkoxyl, amino and derivatives thereof
  • electron-withdrawing groups or atoms eg nitro, halogen, carboxy, carboxamido, sulfonic acid and derivatives thereof.
  • the invention also includes what may be termed “prodrugs”, that is protected forms of the linked cyclic compounds, which release the compound after administration to a patient.
  • the compound may carry a protective group which is split off by hydrolysis in body fluids, eg e.g. in the bloodstream, thus releasing active compound.
  • a discussion of pro-drugs may be found in “Smith and Williams' Introduction to the Principles of Drug Design”, H. J. Smith, Wright, 2nd Edition, London 1988.
  • the invention further provides a method for the production of the compounds of formula la, which method comprises nucleophilic attack by cyclic polyamines Z′ and Y′ each having a single unprotected ring amine nitrogen, all other ring amine nitrogens being protected, on a compound of formula II X-R-A′-R′-X II wherein R, R′ and A′ are as defined above, and
  • the reaction is preferably carried out by reacting two equivalents of the protected polyamine with the compound of formula II in a solvent, such as acetonitrile or dimethylformamide, tetrahydrofuran or dioxane and in the presence of a base, for example sodium carbonate or potassium carbonate.
  • a solvent such as acetonitrile or dimethylformamide, tetrahydrofuran or dioxane
  • a base for example sodium carbonate or potassium carbonate.
  • the reaction generally takes place readily at room temperature to elevated temperature, to give a linked molecule having protected amine nitrogen atoms.
  • chromatography on silica gel is a particularly convenient method of separation.
  • the deprotection step is suitably carried out by refluxing the protected molecule in a mixture of aqueous HBr and acetic acid or in the case of diethylphosphoryl in the presence of hydrogen chloride (gas) in THF or dioxane.
  • the compounds are indicated for the treatment of vital infections, especially retrovirus infections and particularly HIV infections, and the compounds of formula I are to he be considered as active compounds for the pharmaceutical compositions, processes for making the same and methods of treatment mentioned above.
  • the compounds of formula I are to he be considered as active compounds for the pharmaceutical compositions, processes for making the same and methods of treatment mentioned above.
  • meso forms, enantiomers and resolved optically active forms of the compounds of formula I are included.
  • compounds of formula I diluted with non-toxic or other active substances.
  • Acid addition salts for example hydrochlorides, and non-toxic labile metal complexes of the compounds of formula I are also active compounds according to the present invention.
  • Non-toxic in the present context has to be considered with reference to the prognosis for the infected patient without treatment. Copper and zinc complexes are preferred although other metals such as nickel may be considered, whereas less labile metal atoms such as cobalt and rhodium are less preferred because of likely lower selectivity.
  • 1,1′-[2,3,5,6-Tetrafluoro-1,4-phenylenebis-(methylene)]-bis-tris-(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane 200 mg, 0.13 mmol was dissolved in a mixture of acetic acid and hydrobromic acid (48%) in a ratio of approximately 3:2 by volume (10 ml) and heated to 100° C. for 24 hours during which time a white solid precipitated.
  • 1,4-Dimethylnaphthalene-4,4′-dibromide gave 1,1′-[1,4-naphthylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate.
  • step b) The mono-deprotected tetraazacyclohexadecane macrocycle described in step b) was used as described in Example 1 steps c) and d), to prepare tetraazacyclohexadecane dimers.
  • 4,4′-Dibromo-m-xylene gave 1,1′-[1,3-phenylene-bis-(methylene)]-bis-1,5,9,13-tetraazacyclohexadecane octahydrobromide hexahydrate.
  • reaction mixture was allowed to cool to mom temperature and the solid was filtered off, washed with acetic acid followed by ether and dried in vacuo thus affording a white solid which was identified by 1 H NMR, 13 C NMR, FAB-MS and elemental analysis as 1,1′-[2,6-pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate (230 mg, 65%).
  • Mass spectrum (FAB); m/e (relative intensity); 591 (M+HBr, 26), 589 (M+HBr, 26), 509 (M+1, 22) 311 (23), 201 (71), 185 (100).
  • the compounds of the invention were tested in a screen by the MTT method (I Vixol 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
  • Anti-viral activity and cytotoxicity of the compounds are expressed in the table below as IC 50 ( ⁇ g/ml) and CC 50 ( ⁇ g/ml), respectively.
  • the potential therapeutic usefulness was assessed by calculating a Selectivity Index (SI) corresponding to the ratio of CC 50 to IC 50 .
  • SI Selectivity Index
  • the compounds according to the invention are highly active against HIV-1 and -2, with low toxicity, in the in vitro tests used.
  • the compound B which is the most preferred compound of the invention, was further tested for antiviral effects on different laboratory strains of HIV-1 in MT-4 cells, using the MIT assay.
  • Compound B was found to have an IC 50 in the range of 2-5 ng/ml against IIIb, RF, HE and NDK strains, showing that its high activity is remarkably strain-independent.
  • T4-lymphocytes and monocytes are targets for HIV-1 infection in vivo.
  • the following test method showed that compound B inhibits virus replication also in primary T4 cells and primary monocytes in culture.
  • T4 lymphocytes were purified from human spleens obtained from healthy donors by using a commercial kit (“Lympho-Kwik”) which combines reaction of cells with specific monoclonal antibodies and density gradient centrifugation to separate the cells. Preparations obtained by this procedure contained 60-80% CD4 positive cells as analysed by FACS. Cells were stimulated with 2 ⁇ g/ml PHA for 24 hours. Then they were spun down and infected with HIV-1, strain IIIb, by suspending the cells 10-fold concentrated in virus solution. Adsorption was allowed for 2 hours at 37° C. The inoculum was removed by centrifugation and the cells were re-suspended at their original concentration in fresh culture medium containing IL-2 (40 IE/ml).
  • Test compound was added after stimulation and virus adsorption. Every 3 to 4 days post infection half of the supernatant of the infected cultures was removed and replaced by fresh medium containing the test compound at the particular concentration, The concentration of vital p24 antigen was determined in the supernatent by means of a commercial ELISA kit (Coulter) and served as a parameter for virus production. Compound B does not interfere with the p24 Elisa test (highest concentration tested: 100 ⁇ g/ml).
  • Mononuclear cells were isolated from healthy, HIV-negative donors using Ficoll density separation. Cells (4 ⁇ 10 6 /ml) were incubated for 5 days in 48 well plates (Costar) in monocyte medium consisting of RPMII640, supplemented with 20% ECS and 10% human serum. On day 5 non-adherent cells were washed out four times with warm PBS containing 2% human serum. Preparations obtained by the procedure were >95% positive for non-specific esterase (Sigma) and cell viability (as determined by trypan blue exclusion) was always >95%.
  • the monocytotropic strain of HIV-1, BaL was used for the infection of these monocyte preparations (Pemo et al, 1 Exp Meal, 169, 933, 1989).
  • Adherent monocytes were exposed to 50 ⁇ g/well of a 1:30 dilution of HIV-1, BaL for 30 minutes subsequently, monocyte medium was added to 1 ml/well. Adsorption was allowed for 24 hours at 37° C. Then, the wells were washed twice in order to remove excess virus and were cultivated in the presence of different drug concentrations. Thus, test compounds were added after adsorption. Every 3 to 4 days post infection the supernatant of the infected cultures was removed and replaced by fresh medium containing the test compound at the particular concentration. The concentration of viral p24 antigen was determined as described above.
  • IC 50 and IC 90 values were calculated by comparing the p24 antigen concentrations in supernatent of treated, infected cells and uncreated, infected cells at days 11 and 14 post infection.
  • Table 2 shows that Compound B is a potent inhibitor of HIV-1 replication in both primary cell types, with IC 90 values of 1-2 ng/ml. At the highest concentration tested, 100 ng/ml, no cytotoxicity was observed.
  • Compound B was a strong inhibitor of vital replication in primary T4 cells infected with low-passage primary clinical isolates of HIV-1 from three different geographical locations (K31, Zaire, D370, California, and K6/2, Germany).
  • the low cytotoxicity of Compound B was also shown by incubation of exponentially growing cells with Compound B or with AZT and determining cell numbers 2, 3 and 4 days after seeding.
  • Compound B did not inhibit growth of MT4, MOLT4, HUT78, Jurkat cells (all T cell lines) nor the growth of the monocylic U937 cell line at concentrations below 300 ⁇ g/ml.
  • HUT78 cells AZT was in all cases more cytotoxic than Compound B with TC 50 values ( ⁇ g/ml) of 23, 37, 184 and 5 for MT4, MOLT4, Jurkat and U937 respectively.
  • the compounds of the invention do not block virus production from chronically infected cells, indicating that the antiviral target is in the early part of the infection process, before, or at, integration of the provirus.
  • a time-of-addition experiment was carried out on MT4 cells infected with HIV-1 strain IIIb at high virus multiplicity to ensure that the virus replicative steps would be synchronised in the whole cell populations.
  • Test compounds wife added 1, 2, 3, . . . 22, 23, 24 hours after infection, and vital p24 antigen production determined 29 hours after infection.
  • TIBO derivative R82913
  • Compound B was also found to inhibit fusion, which is the mechanism by which viruses enter cells and by which virus or infectious material is transmitted from cell to cell. Syncytium formation between chronically infected cells and uninfected cells reflects the gp120/41 mediated fusion process of vital entry.
  • the syncytium inhibition assay (Baba et al, J AIDS 3 493, 1990)) using HIV-1 IIIb infected HUT78 cells with MOLT4 cells indicates that Compound B is at least as potent as dextran sulphate in inhibition of fusion.
  • concentrations required (approximately 1 ⁇ g/ml) are considerably higher than the antiviral IC 50 values, but are well below cytotoxicity levels.
  • the compounds of Formula I axe therefore useful for the treatment and/or prophylaxis of HIV infection, alone or in combination with other active agents.
  • the appropriate dosage will, of course, vary depending upon, for example, the compound of Formula I employed, the host, the mode of administration and the nature and severity of the conditions being treated. However, in general, satisfactory results in humans are indicated to be obtained at daily dosages from about 0.01-20 mg/kg of body weight.
  • An indicated daily dosage in humans is in the range from about 0.7 mg to about 1400 mg of a compound of Formula I conveniently administered for example in divided doses up to four times a day.
  • the compounds of Formula I may be administered by any conventional route, particularly enterally, preferably orally, eg in the loan of tablets or capsules or in liquid form, eg as a syrup; or parenterally, eg in the form of solutions or suspensions for iv or sc administration.
  • Compound B is the preferred compound of Formula I. In view of its activity in the test methods as described above, it is indicated that Compound B may be administered to humans at daily dosages of from 2 to 200 mg, preferably 10 to 70 mg, by parentexal administration, eg by subcutaneous injection.
  • the compounds of Formula I may be administered in free base form or in pharmaceutically acceptable acid addition salt or metal complex form. Such salts and complexes may be prepaxed in conventional manner as described in the Examples, and exhibit the same order of activity as the free bases. Pharmaceutical compositions containing compounds of Formula I may be manufactured in conventional manner. Unit dosage forms contain for example from about 0.5 mg to about 100 mg of a compound of Formula I in free base or pharmaceutically acceptable acid addition salt form.

Abstract

Polyamine macrocyclic compounds, e.g. of 10 to 15 ring members and 3 to 6 ring amine nitrogens, linked through methylene groups to an aromatic moiety, show high selective activity against HIV.

Description

This application is the 356USC371 Nation 35 U.S.C. 371 National Stage of PCT/GB92/02334, filed Dec. 16, 1992, which claims the benefit of priority to GB 91/26677, filed Dec. 16, 1991.
This invention concerns improvements in chemical compounds, more especially it concerns compounds and pharmaceutical compositions. In particular it concerns compositions and compounds having activity in in vitro tests on Human Immunodeficiency Virus-infected cells.
The disease known as Acquired Immune Deficiency Syndrome (AIDS) caused by infection by HIV has attracted immense research effort because of the effects of the disease on infected individuals and the dangers of the disease spreading to a wider section of the population. In general, although various chemo-therapeutic treatments have been advocated, and some compounds have emerged as a potential basis for treatment, there is still a need for alternatives. In particular, most treatments such as the compound known as AZT have a high toxicity to cells, and it would be desirable to find compounds which are less toxic. In man, the development of resistance to AZT has been identified as an additional clinical problem.
We have found a group of compounds which show protective properties in vitro screens of cells challenged with HIV-1 and/or HIV-2, and are therefore useful for the treatment of AIDS and AIDS Related Complex and other vital and especially retrovital infections. Accordingly, the present invention provides the use of compounds defined below, in phamaceutical compositions for treating HIV-infected patients. The invention further provides pharmaceutical compositions comprising a said compound in combination or association with a pharmaceutically acceptable diluent or excipient, for the treatment of HIV-infected patents. The invention may also be defined as the use of a said compound for the manufacture of a medicament for the treatment of HIV-infected patients. The invention further provides a process for the production of a pharmaceutical composition for the treatment of a HIV-infected patient, comprising the combination of a compound as defined below with a pharmaceutically acceptable diluent or excipient, and formulating said composition into a form suitable far administration to said patient. The invention also provides a method of treatment of an HIV-infected patient, comprising administering to said patient an effective dose of a said compound. It is to be understood that treatment includes prophylactic treatment of patients at risk, in view of the protective properties observed. The use of the compounds may also be stated as a method of treating HIV-infected or HIV-challenged human cells to prevent or modulate the multiplication of the HIV, comprising administering to said cells an effective dose of a said compound. Whilst this description is especially directed to combating HIV, this invention includes other aspects in which other diseases may be treated, including for example microbial infections.
A 2,2′-dimer of cyclam has been reported as being isolated as a 2% by-product in the synthesis of cyclam (1,4,8,11-tetraaza-cyclotetradecane) (Barefield et al, J C S Chem Con (1981), 302). This compound was stated to be insoluble in water. We believe that the insoluble 2,2′-bicyclam is a mixture of the 2R,2′R and 2S,2′S enantiomers; we have characterised a soluble dimer which we believe to be the me, so 2R,2′S isomer. The 6,6′-bicyclam isomer has been reported by Fabbrizzi et al, Inorg Chem 25, 2671 (1986). Certain N,N′-linked bicyclic compounds have been reported by Ciampolini et al, Inorg Chem 26, 3527 (1987). No biological activity has been suggested for such compounds.
U.S. Pat. No. 4,156,683 discloses monocyclic and bicyclic macrocyclic compounds, which axe are said to have biological activity in regulating sodium, potassium and calcium levels in mammals. Additionally, a specific group of N-alkylated monocyclic compounds are said to possess activity against A2 influenza viruses in a modified Hermann test on chick fibroblast tissue. It is also said that the preferred compounds, which form complexes of greater stability, are those having three bridging chains between bridgehead nitrogen atoms, that is fused bicyclic compounds.
EP-A-0296522 discloses certain functionally modified cyclic polyamines, including that known as “cyclam”, which complexes with rhodium and may be bound to an antibody or antibody fragment The aromatic-linked cyclic polyamines which form the subject of the present invention are not disclosed, nor is any anti-vital activity.
EP-A-0305320 also discloses several modified cyclic polyamines, but does not disclose identical cyclic polyamines linked together.
WO-A-9105762 discloses polyamines useful for their multi-point chelating activity, but does not disclose linked cyclic polyamines.
WO-A-9216494 is in the same name as the present applicants, and discloses long-chain polyamines, optionally linked to a cyclic polyamine, as agents active against HIV. No molecules having two cyclic polyamines, linked through an aromatic linker are disclosed in this prior art.
Our U.S. Pat. No. 5,021,409 (equivalent to EP-A-0434385) describes linked cyclic compounds as being active against HIV-1 and HIV-2 in in vitro tests. We have now discovered that certain of the linked cyclic compounds exhibit surprisingly improved activity against HIV. Thus, the present invention concerns a selected group of the compounds taught in said USP, having activity of at least an order of magnitude greater than the compounds tested in said USP.
The present invention provides as active compounds linked cyclic compounds of the general formula I
Z-R-A-R′-Y  (I)
in which Z and Y are identical cyclic polyamine moieties having from 9 to 20 ring members and from 3 to 6 amine nitrogens in the ring spaced by 2 or more carbon atoms from each other,
    • A is an aromatic or heteroaromatic moiety other than quinoline, and
    • R and R′ are each methylene linked to an amine nitrogen atom in Z and in Y, the amine nitrogens being otherwise unsubstituted. The invention also encompasses acid addition salts and metal complexes of the compounds of formula I.
In the above formula, the cyclic polyamine moieties may be substituted or unsubstituted, and suitable substituents are alkyl and/or aryl groups, eg of up to 10 carbon atoms, and any other atoms or groups which do not substantially adversely affect the activity or toxicity of the compounds. Preferred moieties are those of 10 to 15 ring members, and there are preferably 3 or 4 amine nitrogen atoms.
The aromatic or heteroaromatic moiety A tethers Y and Z through the linking groups R and R′. Moiety A may be phenyl or fused aromatic such as napthyl, heterocyclic such as pyridyl or thiophenyl, fused heterocyclic or joined aromatic and/or joined heteroaromatic, for example biphenyl or bipyridyl respectively. The moieties A may also be substituted at single or multiple non-linking positions with electron-donating groups, eg alkyl, thio, thioalkyl, hydroxyl, alkoxyl, amino and derivatives thereof, or electron-withdrawing groups or atoms, eg nitro, halogen, carboxy, carboxamido, sulfonic acid and derivatives thereof.
The invention also includes what may be termed “prodrugs”, that is protected forms of the linked cyclic compounds, which release the compound after administration to a patient. For example, the compound may carry a protective group which is split off by hydrolysis in body fluids, eg e.g. in the bloodstream, thus releasing active compound. A discussion of pro-drugs may be found in “Smith and Williams' Introduction to the Principles of Drug Design”, H. J. Smith, Wright, 2nd Edition, London 1988.
A few of the active compounds according to the invention axe are known, (see Inorg Chem 26 (1987), p 3527-3533 and J Chem Soc, Chem Commun, (1991), 206, 207).
Accordingly, certain of the compounds of formula I are novel. The invention accordingly provides novel linked cyclic polyamine compounds of general formula Ia,
Z-R-A′-R′-Y  (Ia)
in which Z, Y, R and R′ are as defined above, with R and R′ linked to nitrogen atoms in Z and Y, and
    • A′ is an aromatic or heteroaromatic moiety which is unsubstituted or substituted, other than quinoline, provided that
    • A′ is not unsubstituted phenylene when Z and Y are 14-membered tetraaza rings,
      and their acid addition salts and metal complexes.
The invention further provides a method for the production of the compounds of formula la, which method comprises nucleophilic attack by cyclic polyamines Z′ and Y′ each having a single unprotected ring amine nitrogen, all other ring amine nitrogens being protected, on a compound of formula II
X-R-A′-R′-X  II
wherein R, R′ and A′ are as defined above, and
    • each X is an active substituent which can be displaced by the unprotected amine nitrogens of polyamines Z′ and Y′ and is preferably selected from Br, Cl, I, methanesulfonate, 4-tolylsulfonate and trifluoromethane sulfonate,
      and subsequently deprotecting the ring amine nitrogens.
It is well within the capabilities and knowledge of the skilled synthetic chemist to protect the amine nitrogens of cyclic polyamines, and it is preferred to use substitution by methanesulfonyl and/or 4-talylsulfonyl and/or diethylphosphoryl. The compounds of formula II are known.
The reaction is preferably carried out by reacting two equivalents of the protected polyamine with the compound of formula II in a solvent, such as acetonitrile or dimethylformamide, tetrahydrofuran or dioxane and in the presence of a base, for example sodium carbonate or potassium carbonate. The reaction generally takes place readily at room temperature to elevated temperature, to give a linked molecule having protected amine nitrogen atoms. In general, a mixture of products will be obtained, and we have found that chromatography on silica gel is a particularly convenient method of separation.
The deprotection step is suitably carried out by refluxing the protected molecule in a mixture of aqueous HBr and acetic acid or in the case of diethylphosphoryl in the presence of hydrogen chloride (gas) in THF or dioxane.
The compounds are indicated for the treatment of vital infections, especially retrovirus infections and particularly HIV infections, and the compounds of formula I are to he be considered as active compounds for the pharmaceutical compositions, processes for making the same and methods of treatment mentioned above. In these aspects of the invention, it is to be understood that meso forms, enantiomers and resolved optically active forms of the compounds of formula I are included. Also, it is to he be considered within the invention, compounds of formula I diluted with non-toxic or other active substances.
Acid addition salts, for example hydrochlorides, and non-toxic labile metal complexes of the compounds of formula I are also active compounds according to the present invention. Non-toxic in the present context has to be considered with reference to the prognosis for the infected patient without treatment. Copper and zinc complexes are preferred although other metals such as nickel may be considered, whereas less labile metal atoms such as cobalt and rhodium are less preferred because of likely lower selectivity.
The present invention will now be illustrated by the following preparative examples.
EXAMPLE 1
a) 2,3,5,6- Tetrafluoro-p-xylene-∝,∝′-diol
To a stirred solution of perfluoroterephthalic acid (1.0 g, 4.2 mmol) in anhydrous THF (10 ml) under an atmosphere of dry argon was added Borane. THF complex (1.0M solution in THF, 10 equivalents, 42 ml) dropwise, and the mixture stirred at room temperature overnight. The solution was evaporated under reduced pressure to give a colourless oil and the excess Borane destroyed by addition of anhydrous methanol (40 ml) and evaporation (repeated three times). The residue was treated with 5% aqueous hydrocMoric acid then the pH of the mixture was adjusted to pH9 with 1N aqueous sodium hydroxide solution and extracted with dichloromethane (3×50 ml). The combined organic extracts were dried (MgSO4) and evaporated to give 2,3,5,6-tetrafluoro-p-xylene-∝,∝′-diol (0.75 g, 86%) as a white solid. This was used without further purification.
b) 2,3,5,6- Tetrafluoro-p-xylene-∝,∝′-diol dimesylate
To a stirred solution of 2,3,5,6-tetrafluoro-p-xylene-∝,∝′-diol (0.72 g, 3.4 mmol) in dichloromethane (40 ml) containing triethylamine (1.2 ml, 2.5 equivalents) was added methanesulfonyl chloride (0.58 ml, 2.2 equivalents) dropwise at 0° C. and the mixture was allowed to warm to room temperature overnight. The solution was washed with saturated aqueous sodium bicarbonate solution (2×20 ml) and brine (2×20 ml) then dried (MgSO4) and evaporated under reduced pressure. The residue was suspended in ether and filtered giving 2,3,5,6-tetrafluoro-p-xylene-∝,∝′-diol dimesylate (0.9 g, 72%) as a white solid.
c) 1,1′-[2,3,5,6-Tetrafluoro-1,4-phenylenebis-(methylene)]-bis-tris(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane
2,3,5,6-Tetrafluoro-p-xylene-∝,∝′-diol dimesylate (150 mg, 0.4 mmol), tris-(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane monohydrate 826 mg, 1.2 mmol, 3.0 equivalents) and potassium carbonate (252 mg, 3.0 equivalents) in anhydrous acetonitrile (20 ml) were heated to reflux with stirring under argon for 48 hours until all the dimesylate starting material had been consumed; confirmed by TLC (silica gel, 2% methanol in dichloromethane as eluent). The mixture was evaporated under reduced pressure and the residue was dissolved in ethyl acetate (40 ml) and washed with saturated aqueous sodium bicarbonate solution (2×20 ml) and brine (2×20 ml) then dried (MgSO4) and evaporated. The residue was purified by column chromatography on silica gel during with 2% methanol in dichloromethane giving a white foam identified by 1H NMR and FAB-MS as 1,1′-[2,3,5,6-tetrafluoro-1,4-phenylene-bis-(methylene)]-bis-tris-(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane C70H86N8O12S6F4 requires C, 56.05; H, 5.78; N, 7.47; found C, 55.81; H, 5.73; N, 7.36.
d) 1,1′-[2,3,5,6-Tetrafluoro-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
1,1′-[2,3,5,6-Tetrafluoro-1,4-phenylenebis-(methylene)]-bis-tris-(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane (200 mg, 0.13 mmol) was dissolved in a mixture of acetic acid and hydrobromic acid (48%) in a ratio of approximately 3:2 by volume (10 ml) and heated to 100° C. for 24 hours during which time a white solid precipitated. The mixture was allowed to cool and the solid was filtered off and washed with acetic acid and ether and dried in vacuo giving a white solid identified by 1H NMR, FAB-MS and elemental analysis as 1,1′-[2,-3,5,6-tetra-fluoro-1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclo-tetradecane octahydrobromide dihydrate (65 mg, 40%).
C28H62N8O2Br8F4 requires C, 26.73; H, 4.96; N, 8.90; found C, 26.84; H, 5.05; N, 8.21.
The following compounds were prepared using analogous methods to those described above in steps b)-d):
5-Nitro-m-xylene∝,∝′-diol gave 1,1-[5-Nitro-1,3-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide dihydrate. C28H65N9O4Br8 requires C, 27.31; H, 5.31; N, 10.24; found C, 27.49;, H, 5.26; N, 9.75.
2,4,5,6-Tetrachloro-m-xylene-∝,∝′-diol gave 1,1′-[2,4,5,6-tetrachloro-1,3-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraaza-cyclotetradecane octahyclrobromide dihydrate.
C28H62O2Cl4Br8, requires C, 25.40; H, 4.71; N, 8.46; found C, 25.72; H, 4.76; N, 8.05.
EXAMPLE 2
a) ∝,∝′-Dibromo-1,4-dimethylnaphthalene
To a solution of 1,4-dimethylnaphthalene (0.5 g, 3.2 mmol) and benzoyl peroxide (0.08 equivalents, 62 mg) in carbon tetrachloride (20 ml) was added N-bromosuccinimide (1.14 g, 2.0 equivalents) and the mixture was heated to reflux for 24 hours during which time a white solid precipitated. The mixture was faltered hot (to remove the succinimide by-product) and then allowed to cool over several hours during which time a white cystalline solid precipitated. The solid was filtered off and dried giving 1,4-dimethylnaphthalene-∝,∝′-dibromide (473 mg, 50%).
The following compound was prepared using methods analogous to steps c) and d) of Example 1:
1,4-Dimethylnaphthalene-4,4′-dibromide gave 1,1′-[1,4-naphthylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate.
C32H72N8O4Br8 requires C, 30.20; H, 5.69;, N, 8.81; found C, 30.28; H, 5.52; N, 8.66.
EXAMPLE 3
a) 1-Benzyl-5,13-di-(p-toluenesulfonyl)-9-methanesulfonyl-1,5,9,13-tetraazacyclohexadecane.
To a solution of N,N-bis-[3-(p-toluenesulfonylamidopropyl)-]benzylamine hydrochloride (25 g) (NL Patent 6603655) in dry DMF (800 ml) under argon was added sodium hydride (10 equivalents) in small portions over 3 hours. When the addition was complete the solution was heated at 6° C. for 1 hour then allowed to cool and the excess sodium hydride was removed by filtration under argon. The filtrate was transferred to another dry flask and the solution was then heated to 100°-110° C. and bis-propanolamine-trimethanesulfonate [P Moore, J Chem Soc Dalton Trans 1985 (7) 1361-1364] (1.0 equivalent) in DMF (500 ml) was added dropwise over 8 hours with rapid stirring. The temperature was maintained at 100°-110° C. for a further 16 hours, allowed to cool then the mixture was poured into iced water (1500 ml) and the resulting off-white precipitate that formed was collected by filtration. The solid was dissolved in dichloromethane (250 ml) and the solution was washed with water (5×50 ml), then dried (MgSO4) and evaporated under reduced pressure to give a yellow oil. Trituration with ethanol (200 ml) gave a white crystalline solid which was filtered off, washed with a small volume of ethanol, then ether and dried in vacuo to give 1-benzyl-5,-13-di-(p-toluenesulphonyl)-9-methanesulphonyl-1,5,9,-13tetraazacyclohexadecane (45%), identified by 1H NMR and FAB-MS.
b) 1,9-Di-(p-toluenesulfonyl)-5-methanesulfonyl-1,5,9,13-tetraazacyciohexadecane
To a solution of 1-benzyl-5,13-di-(p-toluene-sulfonyl)-9-methanesulfonyl-1,5,9,13-tetraazacyclohexadecane in formic acid (20 ml) was added Palladium hydroxide on carbon (Pearlmans catalyst, 4.0 g) and the resulting suspension was heated to reflux for 72 hours with stirring. The mixture was allowed to cool, then filtered through celite and the filtrate was evaporated under reduced pressure. The colourless oil which remained was dissolved in dichloromethane (50 ml) and washed with 10% aqueous sodium hydroxide solution (2×20 ml), and water (2×20 ml) then dried (MgSO4) and evaporated under reduced pressure. The residue was purified by column chromatography on silica gel eluting with 3% methanol in dichloromethane giving a white solid identified by 1H NMR and FAB-MS as 1,9-di-(p-toluenesulphonyl-5-methanesulfonyl-1,5,9,13-tetraaza-cyclohexadecane.
The mono-deprotected tetraazacyclohexadecane macrocycle described in step b) was used as described in Example 1 steps c) and d), to prepare tetraazacyclohexadecane dimers.
The following compounds were prepared in this manner.
4,4′-Dibromo-m-xylene gave 1,1′-[1,3-phenylene-bis-(methylene)]-bis-1,5,9,13-tetraazacyclohexadecane octahydrobromide hexahydrate.
C32H72N8O6Br8 requires C, 29.2; H, 6.15; N; 8.54; found C, 29.37; H, 5.50; N, 7.90.
4,4′-Dibromo-p-xylenegave 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,5,9,13-tetraazacyclohexadecane octahydrobromide hexahydrate.
C32H76N8O6Br8 requires C, 29.29; H, 6.15; N, 8.54; found. C, 28.96; H, 5,47; N,7.96.
Other compounds which may be made according to the invention are:
  • 1,1′-[1,3-phenylenebis(methylene)]-bis-1,5,9,13-tetraazacyclohexadecane
  • 1,1′-[1,3-phenylenebis(methylene)]-bis-1,5,9-triazacyclododecane
  • 1,1′-[1,4-phenylenebis(methylene)]-bis-1,5,9-triazacyclododecane
EXAMPLE 4
Synthesis of Compound F
1,1′-[1,4-Phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane zinc dichloride monohydrate
To a stirred solution of 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazaeyclotetradecane (1 g) in methanol (25 ml) was added zinc(II) chloride (0.54 g, 2.0 eq) in methanol (5 ml). Towards the end of the addition a white precipitate formed. Sufficient methanol and water were added to give a homogenous solution and the mixture was then evaporated in vacuo. The solid residue was suspended in a mixture of methanol/ether and filtered giving 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane zinc dichloride monohydrate (1.45 g, 94%) as a white powder.
C28H56Cl4OZn2 requires; C, 42.38; H, 7.11; N, 14.12; Cl, 17.88: found; C, 42.64; H, 7.14; N, 14.18; Cl, 17.89.
EXAMPLE 5
Synthesis of Compound G
1,1′-[1,4-Phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane copper diacetate hexahydrate
To a stirred solution of 1,1′[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane (100 mg) was added copper(II) acetate (72 mg, 2.0 eq) in one portion. The solution became dark blue/purple in colour almost immediately. The mixture was stirred for one hour then triturated with ether to give a blue precipitate. The blue solid was filtered off and dried giving 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane copper diacetate hexahydrate (80 mg, 46%).
C36H80N8OCu2 requires; C, 43.58; H, 8.13; N, 11.29; found: C, 43.24; H, 7.88; N, 11.13.
EXAMPLE 6 1,1′-[3′-Biphenylene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11tetraazacyclotetradecane
A mixture of 3,3′-bis-(bromomethyl)-1,1′-biphenyl [W. Wenner, J. Org. Chem. (1952), 17, 525-528], (200 mg, 0.59 mmol), anhydrous potassium carbonate (325 mg, 2.35 mmol, 4 eq) and tris-(p-toluene-sulphonyl)-1,4,8,11-tetraazacyclotetradecane (801 mg, 1.18 mmol, 2 eq) in anhydrous acetonitrile (15 ml) was stirred at 50° C. under argon. After 6 hours the reaction mixture was allowed to cool; dichloromethane (75 ml) was added and the resulting solution filtered. The filtrate was evaporated in vacuo to yield a glassy white solid. Chromatography of the crude product on a column of silica gel (2.5 cm×20 cm), eluting with methanol/dichloromethane 1:160 v/v gave a white solid, identified by 1H NMR as 1,1′-[3,3′-biphenylene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (665 mg, 76%).
Synthesis of Compound J
1,1′-[3,3′-Biphenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate
The per-tosylated derivative from above (450 mg, 0.30 mmol) was dissolved in glacial acetic acid (9 ml). Hydrobromic acid (˜48% w/v, Aldrich, 3.5 ml) was added and the resulting mixture heated to reflux. After 24 hours the dark brown solution was cooled in an ice bath over 2 hours during which time an off-white precipitate formed. The precipitate was collected by centrifugation and washed with glacial acetic acid (3×10 ml) followed by diethyl ether (4×10 ml) then dried overnight in vacuo to give a white powder, identified by 1H NMR and elemental analysis as 1,1′-[3,3′-biphenylene-bis-(methylene)]-bis-1,4,8,11-tetraaza-cyclotetradecane octahydrobromide tetrahydrate (194 mg, 50%). C34H74N8Br8O4 requires; C, 31.46; H, 5.70; N, 8.63; found; C, 31.30; H, 5.68; N, 8.60.
EXAMPLE 7 1,1′-[4,4′-(2,2′-Bipyridine)-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacydotetradecane
A mixture of 4,4′-bis-(bromomethyl)-2,2′-bipyridine [T J Meyer, Inorg. Clam. (1991), 30, 2942-2949], (200 mg, 0.57 mol), anhydrous potassium carbonate (314 mg, 2.27 mmol, 4 eq) and tris-(p-toluenesulfonyl)-1,4,8,11-tetraazacyclotetradecane (774 mg, 1.14 mmol, 2 eq) in anhydrous acetonitrile (20 ml) was stirred at 50° C. under argon for 2 hours. The mixture was allowed to cool and dichloromethane (100 ml) was added and the resulting solution filtered through celite. The filtrate was evaporated in vacuo to give a yellow glassy solid which was purified by column chromatography on silica gel (3×20 cm column) using triethylamine/methanol/dichloromethane 1:1:100 v/v as eluent. A glassy white solid was obtained, identified by 1H NMR as 1,1′-[4,4′-(2,2′-bipyridine)-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (600 mg, 70%).
Synthesis of Compound K
1,1′-[4,4′-(2,2′-Bipyridine)-bis-(methyl)]-bis-1,4,8,11-tetraazacyclotetradecane decahydrobromide pentahydrate
The per-tosylate derivative from above (570 mg, 0.38 mmol) was dissolved in glacial acetic acid (6.5 ml). Hydrobromic acid (˜48% w/v, Aldrich, 3.0 mmol) was added and the mixture heated to reflux for 24 hours. The resulting dark brown solution was cooled in an ice bath over 2 hours) during which time an off-white precipitate formed. The precipitate was collected by centrifugation and washed with glacial acetic acid (3×10 ml) followed by diethyl ether (5×10 ml) and dried overnight in vacuo to give a white powder identified by 1H NMR and elemental analysis as 1,1′-[4,4′-(2,2′-bipyridine)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane decahydrobromide pentahydrate (450 mg, 81%).
C32H76N10Br10O5 requires; C, 25.97; H, 5.17; N, 9.46; found; C, 26.07; H, 4.57; N, 9.47.
EXAMPLE 8 1,1′-[2,9-(1,10-Phenanthroline)-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane
A mixture of 2,9-bis-(bromomethyl)-1,10-phenanthroline [C J Chandler, J. Heterocycl. Chem. (1981), 18, 599-601], (200 mg, 0.54 mmol), anhydrous potassium carbonate (300 mg,. 2.17 mmol, 4 eq) and axis-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (740 mg, 1.09 mmol, 2 eq) in anhydrous acetonitrile (20 ml) were stirred at 50° C. under argon for 3 hours. The mixture was allowed to cool and dichloromethane (100 ml) was added and the resulting solution filtered through celite. The filtrate was evaporated in vacuo to give a yellow glassy solid which was purified by column chromatography on silica gel (3×2 cm column) using triethylamine/methanol/dichloromethane 1:3:100 v/v eluent. A pale yellow solid was obtained, identified by 1H NMP as 1,1′-[2,9-(1,10-phenamhroline)-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (575 mg, 69%).
Synthesis of Compound L
1,1′-[2,9-(1,10-Phenanthroline)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane decahydrobromide trihydrate
The per-tosylated derivative from above (400 mg, 0.26 mmol) was dissolved in glacial acetic acid (8 ml). Hydrobromic acid (48% w/v, Aldrich, 3.5 ml) was added and the mixture was heated to reflux for 16 hours. The resulting dark brown solution was cooled in an ice bath over 2 hours during which time an off-white precipitate formed. The precipitate was collected by centrifugation then purified by re-precipitation from a mixture of hydrobromic acid (˜48% w/v, 2 ml) and water (2 ml) with glacial acetic acid (5 ml). The white solid was again collected by centrifugation, washed with glacial acetic acid (3×10 ml) and diethyl ether (4×10 ml) and finally dried overnight in vacuo to give a white powder, identified by 1H NMR and elemental analysis as 1,1′-[2,9-(1,10-phenanthroline)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane decahydrobromide trihydrate (80 mg, 21%).
C34H72N10Br10O3 requires; C, 27.82; H, 4.94; N, 9.54; found; C, 27.81; H, 4.97; N, 9.17.
EXAMPLE 9
This compound and corresponding intermediates axe described by T. A. Kaden, Helv. Chim. Acta., (1985), 69, 53-61. An alternative procedure is given below.
11,11′-[1,4-Phenylene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane
A mixture of α,α′-dibromo-p-xylene (249 mg, 0.94 mmol), anhydrous potassium carbonate (652 mg, 4.71 mmol, 5 eq) and tris(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane [T. A. Kaden, Heir. Chim. Acta., (1983), 66, 861-870] (1.25 g, 1.89 mmol, 2 eq) in anhydrous acetonitrile (15 ml) was heated at 50° C. with stirring under argon for 18 hours. The reaction mixture was allowed to cool and dichloromethane (50 ml) was added and the resulting solution filtered through celite. The filtrate was evaporated in vacuo to give a white foam which was purified by column chromatography on silica gel using methanol/dichloromethane (1:40 v/v) as eluent. A white solid was obtained, identified by 1H NMR as 11,11′-[1,4-phenylene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane (1.0 g, 74%)
Synthesis of Compound M
11,11′-[1,4-Phenylene-bis-(methylene)]-bis-1,4,7,11-tetraazacyclotetradecane octahydrobromide dihydrate
The per-tosylated derivative from above (500 mg, 0.35 mmol) was dissolved in glacial acetic acid (7 ml). Hydrobromic acid (˜48% w/v, 4 ml) was added and the resulting mixture heated to reflux for 20 hours. Further glacial are fie acid (10 ml) was added and the solution was cooled in an ice bath over 1 hour during which time a white precipitate formed. The solid was collected by centrifugation and washed with glacial acetic acid (2×10 ml) followed by diethyl ether (4×10 ml) and dried overnight in vacuo to give a white powder, identified by 1H NMR and elemental analysis as 11,11′-[1,4-phenylene-bis-(methylene)]-bis-1,4,7,11-tetraazacyclotetradecane octahydrobromide dihydrate (280 mg, 67%).
C28H66N8Br8O2 requires; C, 28.35; H, 5.61; N, 9.45; found; C, 28.34; H, 5.42; N, 9.02.
EXAMPLE 10 11[(1, Methylene-4-bromomethylene)-phenylene]-tris-(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane
A mixture of α,α′-dibromo-p-xylene (3.98 g, 15.1 mmol, 10 eq), and anhydrous potassium carbonate (417 mg, 3.02 mmol, 2 eq) in anhydrous acetonitrile (20 ml) was heated to 50° C. With rapid stirring a solution of tris-(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane (1.0 g, 1.51 mmol) in anhydrous acetonitrile (20 ml) was added dropwise over 4 hours. After a further 1 hour the reaction mixture was allowed to cool and the solvent evaporated in vacuo. The residue was purified by column chromatography on silica gel (5×20 cm), eluting with a gradient of dichloromethane to methanol/dichloromethane 1:20 v/v over 2 liters total elution volume. To the resulting colourless glass was added dry hexane (150 ml) and the mixture was heated to reflux then allowed to cool to room temperature. The precipitate which formed was filtered, washed with hexane (3×10 ml) followed by diethyl ether (20 ml) and dried overnight in vacuo to give the title compound as a white powder (710 mg, 53%).
1,11′-[1,4-Phenylene-bis-(methylene)]-tris-(p-tohenesulphonyl)-1,4,8,11-tetraazacyclotetradecane)-tris-(p-toluenesulphonyl)-1,4,7,11-tetraazacyclotetradecane
A mixture of 11-[(1-methylene-4-bromomethylene)-phenylene]-tris-(p-tohenesulphonyl)-1,4,7,11-tetraacyclotetradecane (350 mg, 0.41 mmol), anhydrous potassium carbonate (230 mg, 1.66 mmol, 4 eq) and tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (422 mg, 0.62 mmol, 1.5 eq) in anhydrous acetonitrile (20 ml) were heated with stirring at 50° C. under argon for 7 hours. The reaction mixture was allowed to cool and the solvent was evaporated in vacuo. The residue was purified by column chromatography on silica gel (2.5×25 cm column) using methanol/dichloromethane (1:60 v/v) as eluent, followed by preparative thin layer chromatography on silica gel (eluent methanol/dichloromethane 1:40 v/v, 20 mg/plate) to give a colourless glass, identified by 1H NMR as the title compound (130 mg, 30%).
Synthesis of Compound N
1,11′-[1,4-Phenylene-bis-(methylene)]-1,4,8,11-tetraazacyclotetradecane-1,4,7,11-tetraazacydotetradecane octahydrobromide hexahydrate
The per-tosylated derivative from above (115 mg, 0.08 mmol) was dissolved in glacial acetic acid (3 ml). Hydrobromic acid (˜48%, Aldrich, 1.5 mmol) was added and the mixture was heated to reflux for 48 hour. The resulting dark brown solution was cooled in an ice bath and a white precipitate formed. The solid was collected by centrifugation and washed with glacial acetic acid (3×10 ml) followed by diethyl ether (5×10 ml) and dried overnight in vacuo to give a white powder, identified by 1H NMR and elemental analysis as 1,11′-[1,4-phenylene-bis-(methylene)]-1,4,8,11-tetraazacyclotetradecane-1,4,7,11-tetraaza-cyclotetradecane octahydrobromide hexahydrate. (71 mg, 75%). C29H74N8Br8O6 requires; C, 26.73; H, 5.93; N, 8.91; found; C, 26.50; H. 5.69; N, 9.31.
EXAMPLE 11 1,1′-[2,6-Pyridinebis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane
A stirred solution of 2,6-bis(bromomethyl)pyridine hydrobromide [M. E. Haeg, B. J. Whitlock and H. W. Whitlock Jr, J. Ant. Chem. Soc., (1989), 111, 692], (131 mg, 0.378 mmol), tris-(p-toluene-sulphonyl)-1,4,8,11-tetraazacyclotetradecane (500 mg, 0.75 mmol) and potassium carbonate (400 mg, 2.88 mmol) in anhydrous acetonitrile (15 ml) was heated at 80° C. for 22 hours under an atmosphere of arson. The reaction mixture was allowed to cool to room temperature and concentrated in vacuo. The residue was purified by column chromatography on silica gel using 3% methanol in dichloromethane as eluent thus affording a pale white solid which was identified by 1H NMP, and FAB-MS as 1,1′-[2,6-pyridinebis-(methylene)]-bis-tris(p-toluene-sulphonyl)-1,4,8,11-tetraacyclotetradecane (500 ml, 93%).
Mass spectrum (FAB); m/e (relative intensity); 1428 (M+1, 100), 1272 (35)
Synthesis of Compound O
1,1′-[2,6-Pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate
To a stirred solution of 1,1′-[2,6-pyridinebis-(methylene)]-bis-tris(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (500 mg, 0.35 mmol) in acetic acid (16 ml) was added 48% hydrobromic add (12 ml) and the solution heated to 110° C. for 48 hours during which time a white solid precipitated. The reaction mixture was allowed to cool to mom temperature and the solid was filtered off, washed with acetic acid followed by ether and dried in vacuo thus affording a white solid which was identified by 1H NMR, 13C NMR, FAB-MS and elemental analysis as 1,1′-[2,6-pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide tetrahydrate (230 mg, 65%).
C27H69N9O4Br8 requires; C, 26.50; H, 5.64; N, 10.31; Br, 52.29; found C, 26.91; H, 5.31; N, 10.08; Br, 51.99. Mass spectrum (FAB); m/e (relative intensity); 586 (M+HBr, 48), 584 (M+HBr, 50), 504 (M+1, 100), 201 (60).
EXAMPLE 12 1,1′-[3,5-Pyridine-bis-(methylene)]-bis-tris(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane
A stirred solution of 3.5-bis(bromomethyl)pyridine hydrobromide (M. Momenteau, J. Mispelter, B. Loock and J. M. Lhoste, J. Chem. Soc. Perkin Trans. 1, (1985), 61], (131 mg, 0.37 mmol) , tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (500 mg, 0.755 mmol) and potassium carbonate (400 mg, 2.88 mmol) in anhydrous dimethylformamide (15 ml) were heated at 70° C. for 21 hours under an atmosphere of argon. The reaction mixture was allowed to cool to room temperature and concentrated in vacuo. The residue was purified by column chromatography on silica gel using 2% methanol in dichloromethane as eluent thus affording a white foamy solid which was identified by 1H NMR and FAB-MS as 1,1′-[3,5-pyridinebis-(methylene)]-bis-tris(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (320 mg, 78%).
Mass spectrum (FAB); m/e (relative intensity); 1428 (M+1, 100), 1272 (45).
Synthesis of Compound P
1,1′[3,5-Pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane nonahydrobromide dihydrate
To a stirred solution of 1,1′-[3,5-pyridinebis-(methylene)-bis-tris(p-tohenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (320 mg, 0.224 mmol) in acetic acid (12 ml) was added 48% hydrobromic acid (8 ml) and the solution heated to 100° C. for 48 hours during which time a white solid precipitated. The reaction mixture was allowed to cool to room temperature and the solid was filtered off, washed with acetic add followed by ether and dried in vacuo thus affording a white solid which was identified by 1H NMR, 13C NMR, FAB-MS and elemental analysis as 1,1′-[3,5-pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane nonahydrobramide dihydrate (150 mg, 53%).
C27H66N9O2Br9 requires; C, 25.56; H, 5.21; N, 9.94; Br, 56.74; found C, 25.71; H, 5.25; N, 9.76; Br, 56.28. Mass spectrum (FAB); m/e (relative intensity); 586 (M+HBr, 39), 584 (M+HBr, 41), 504 (M+1, 60), 201 (100).
EXAMPLE 13 1,1′-[1,3-Phenylenebis-(methylene)]-bis-tris(p-toluenesulphonyl)-1,4,7,10-tetraazacyclododecane
A stirred solution of α,α′-dibromo-m-xylene (125 mg, 0.472 mmol), tris-(p-tohenesulphonyl)-1,4,7,10-tetraazacyclododecane [M. F. Tweedle et al, Inorg. Chem., (1992), 30, 1265], (600 mg, 0,945 mmol) and potassium carbonate (400 mg, 2.88 mmol) in anhydrous acetonitrile (15 ml) were heated to reflux for 6 hours under an atmosphere of argon. The resulting cloudy white solution was allowed to cool to room temperature and the solids collected by filtration and washed with acetonitrile. The solid residue was dissolved in a mixture of dichloromethane (100 ml) and water (15 ml). The organic phase was separated and washed with water (15 ml), dried (MgSO4) and concentrated under reduced pressure. The residue was dried in vacuo thus affording a white foamy solid which was identified by 1H NMR as 1,1′-[1,3-phenylenebis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,7,10-tetraazacyclododecane (330 mg, 51%).
Synthesis of Compound Q
1,1′-[1,3-Phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane hexahydrobromide
To a stirred solution of 1,1′-[1,3-phenylenebis-(methylene)]-bis-tris-(p-toluenesulfonyl)-1,4,7,10-tetraazacyclododecane (330 mg, 0.24 mmol) in anhydrous methanol/tetrahydrofuran (1:2, 15 ml) was added 3% sodium amalgam (20 g) and dibasic sodium phosphate (400 mg). The reaction mixture was vigorously stirred under argon at 70° C. for 41 hours. The reaction mixture was allowed to cool to room temperature and the supernatent solution was separated from the solids by decantation then concentrated in vacuo. Chloroform (50 ml) and water (5 ml) were added to the residue and the aqueous phase was extracted with chloroform (3×50 ml). Concentration of the combined organic fractions afforded quantitatively a viscous oil which was identified by 1H NMR as 1,1′-[1,3-phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane.
Into a stirred solution of 1,1′-[1,3-phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane in ethanol (20 ml, 95%) was bubbled HBr gas for 15 minutes resulting in an immediate white precipitate. The white solid was filtered off, washed with ethanol and ether and immediately dried in vacuo for 48 hours thus affording a white solid which was identified by 1H NMR, 13C NMR, FAB-MS and elemental analysis as 1,1′-[1,3-phenylene-(methylene)]-bis-1,4,7,10-tetraazacyclo-dodecane hexahydrobromide (130 mg, 63%).
C27H52N8Br6 requires C, 30.92; H, 5.62; N, 12.02; Br, 51.43; found C, 31.09; H, 5.80; N, 11.90; Br, 51.17. Mass spectrum (FAB); m/e (relative intensity); 529 (M+HBr, 53), 527 (M+HBr, 55), 447 (M+I, 100), 277 (40), 185 (35).
EXAMPLE 14 1,1′-[1,4-Phenylenebis-(methylene)]-bis-tris(p-toluenesulphonyl)-1,4,7,10-tetraazacyclododecane
A stirred solution of α,α′-dibromo-p-xylene (99 mg, 0.374 mmol), tris-(p-toluenesulphonyl)-1,4,7,10-tetraazacyclododecane (475 mg, 0.748 mmol) and potassium carbonate (320 mg, 2.24 mmol) in anhydrous acetonitrile (15 ml) was heated at reflux for 1.4 hours under an atmosphere of argon. The resulting cloudy white solution was allowed to cool to room temperature and the solids collected by filtration and washed with acetonitrile. The solid residue was dissolved in a mixture of dichloromethane (120 ml) and water (15 ml). The organic phase was separated and washed with water (15 ml), dried (MgSO4) and concentrated under reduced pressure. The residue was dried in vacuo thus affording a white solid which was identified by 1H NMR as 1,1′-[1,4-phenylenebis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,7,10-tetraazacyclododecane (360 mg, 70%).
Mass spectrum (FAB); m/e (relative intensity); 1371 (IVI+1, 12), 1217 (8).
Synthesis of Compound R
1,1′-[1,4-Phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane hexahydrobromide
To a stirred solution of 1,1′-[1,4-phenylenebis-(methylene)-bis-tris(p-toluensulphonyl)-1,4,7,10-tetraazacycledodecane (360 mg, 0.262 mmol) in anhydrous methanol/dimethylsulphoxide (1:5, 18 ml) was added 3% sodium amalgam (23 g) and dibasic sodium phosphate (400 mg). The reaction mixture was vigorously stirred under argon at 100° C. for 4 hours then allowed to cool to room temperature and the supernatent solution was separated from the solids by decantation and concentrated in vacuo. Chloroform (50 ml) and water (5 ml) were added to the residue and the aqueous phase was extracted with chloroform (3×50 ml). Concentration of the combined organic fractions afforded quantitatively a foamy white solid which was identified by 1H NMR as 1,1′-[1,4-phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane.
Into a stirred solution of 1,1′-[1,4-phenylenebis-(methylene)]-bis-1,4,7,10-tetraazacyclododecane in ethanol (15 ml, 95%) was bubbled HBr gas for 15 minutes resulting in an immediate white precipitate. The white solid was filtered off, washed with ethanol and ether and immediate dried in vacuo for 48 hours thus affording a white solid which was identified by 1H NMR, 13C NMR, FAB-MS and elemental analysis as 1,1′-[1,4-phenylenebis-(methylene)]-bis-1,4,7,10-tetra-azacyclododecane hexahydrobromide (115 mg, 44%).
C27H52N8Br6 requires C, 30.92; H, 5.62; N, 12.02; Br 51.43, found C, 30.90; H, 5.83; N, 11.83; Br, 51.19. Mass spectrum (FAB); m/e (relative intensity); 529 (M+HBr, 40), 527 (M+HBr, 40), 447 (M+1, 58), 185 (100).
EXAMPLE 15 1,1′-[2,5-Thiophene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane
To a solution of tris-p-toluenesulphonyl-1,4,8,11-tetraazacyclotetradecane monohydrate (1.0 g, 1.5 mmol) and potassium carbonate (300 mg, 2.2 mmol) in acetonitrile (20 ml) was added 2,5-dichloromethyl thiophene [J. M. Griffing, L. F. Salisbury, J. Am. Chem. Soc., (1948), 70, 3416-3419], (137 mg, 0.76 mmol) and the mixture was heated to reflux overnight with rapid stirring. The mixture was allowed to cool and the solid was filtered off. The filtrate was evaporated in vacuo and the residue partitioned between methylene chloride (50 ml) and water (25 ml). The organic layer was separated, dried (Na2SO4) and evaporated in vacuo to give the crude product as a light brown solid. Column chromatography [silica gel; methylene chloride/methanol(40/1)] was used to isolate a white solid identified by 1H NMR and FAB-MS as 1,1′-[2,5-thiophene-bis-(methylene)]-bis-tris-(p-tolueno-sulphonyl)-1,4,8,11-tetraazacyclotetradecane (315 mg, 29% ).
Mass spectrum (FAB); M/e (relative intensity); 1434 (M+1, 49), 1277 (31). 772 (100), 616 (30), 508 (24).
Synthesis of Compound T
1,1′-[2,5-Thophenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane octahydrobromide
To a solution of 1.1′-[2,5-thiophene-bis-(methylene)]-bis-tris-(p-toluenesulphonyl)-1,4,8,11-tetraazacyclotetradecane (177 mg, 0.12 mmol) in acetic acid (6 ml) was added hydrobromic acid (Aldrich 48% aqueous, 4 ml) and the mixture was heated to a reflux with stirring for 16 hours during which time a light brown solid precipitated from a dark brown solution. On cooling, a further portion of acetic acid was added (10 ml) and the solids were filtered off, washed with acetic acid (10 ml) and ether (20 ml) and dried in vacuo giving a white solid identified by 1H NMR and FAB-MS as 1,1′-[2,5-thiophene-bis-(methylene)-bis-1,4,8,11-tetraaza-cyclotetradecane octahyarobromide (82 mg, 97%),
Mass spectrum (FAB); m/e (relative intensity); 591 (M+HBr, 26), 589 (M+HBr, 26), 509 (M+1, 22) 311 (23), 201 (71), 185 (100).
The compounds of the invention were tested in a screen by the MTT method (I Vixol 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. Anti-viral activity and cytotoxicity of the compounds are expressed in the table below as IC50 (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 IC50. A control test was performed using the known anti-HIV treatment AZT.
In Table 1 below, the compounds screened were:
    • AZT: known anti-HIV compound
    • A: 1,1′-[1,3-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane
    • B: 1,1′-[1,4-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane
    • C: 1,1′-[5-nitro-1,3-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
    • D: 1,1′-[2,3,5,6-tetrafluoro-1,3-phenylene-bis-(methylene)]bis-1,4,8,11-tetraazacyclotetradecane
    • E: 1,1′-[1,4-naphthylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane
    • F-V: See preceding preparative Examples.
    • W: 1,1′-[2,5-dimethyl-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
    • X: 1,1′-[2,5-dichloro-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
    • Y: 1,1′-[2-bromo-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
    • Z: 1,1′-[6-phenyl-2,4-pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
TABLE 1
HIV-1 (IIIB) HIV-2 (ROD)
IC50 CC50 IC50 CC50
CMPD μg/ml μg/ml SI μg/ml μg/ml SI
AZT <0.008 >1 >125
Com-
parison
A 0.03 >500 >1.5 × 104 <0.01 >500   >5 × 104
B 0.006 >500 >8.3 × 104 <0.01 >500   >5 × 104
C 0.05 55 1100 0.07 55 756
D 0.01 60 6000 0.01 60 6000
E 0.07 71 1014 0.05 71 1420
F 0.0026 >200 >7.6 × 104 0.0019 >200   >1 × 105
G 0.018 >200 >1.1 × 104 0.027 >200 >7.4 × 103
J 0.16 >200 >1250 0.22 >200 >900
K 0.38 117 300 0.35 117 334
L 0.29 >200 >690 0.32 >200 >625
M 0.03 >500 >1.6 × 104 0.07 >500 >7.1 × 103
N 0.01 >500   >5 × 104 0.07 >500 >7.1 × 103
O 0.03 >500 >1.6 × 104 0.08 >500 >6.2 × 103
P 0.04 >500 >1.2 × 104 0.09 >500 >5.5 × 103
Q 0.07 19 271 0.5 19 38
R 0.3 51 170 2.2 51 23
T 0.01 >500 >5.0 × 104 0.02 >500 >2.5 × 104
W 0.0076 >250 >3.2 × 104 0.0013 >250 >1.9 × 105
X 0.0131 71.87 5461 0.0030 72.66   2.4 × 104
Y 0.0075 >250 >3.2 × 104 0.0043 >250 >5.7 × 104
Z 0.0489 >250 5112 0.0246 >250   1.0 × 104
It can readily be seen that the compounds according to the invention are highly active against HIV-1 and -2, with low toxicity, in the in vitro tests used.
The compound B, which is the most preferred compound of the invention, was further tested for antiviral effects on different laboratory strains of HIV-1 in MT-4 cells, using the MIT assay. Compound B was found to have an IC50 in the range of 2-5 ng/ml against IIIb, RF, HE and NDK strains, showing that its high activity is remarkably strain-independent.
T4-lymphocytes and monocytes are targets for HIV-1 infection in vivo. The following test method showed that compound B inhibits virus replication also in primary T4 cells and primary monocytes in culture.
Primary T4 lymphocytes were purified from human spleens obtained from healthy donors by using a commercial kit (“Lympho-Kwik”) which combines reaction of cells with specific monoclonal antibodies and density gradient centrifugation to separate the cells. Preparations obtained by this procedure contained 60-80% CD4 positive cells as analysed by FACS. Cells were stimulated with 2 μg/ml PHA for 24 hours. Then they were spun down and infected with HIV-1, strain IIIb, by suspending the cells 10-fold concentrated in virus solution. Adsorption was allowed for 2 hours at 37° C. The inoculum was removed by centrifugation and the cells were re-suspended at their original concentration in fresh culture medium containing IL-2 (40 IE/ml). Test compound was added after stimulation and virus adsorption. Every 3 to 4 days post infection half of the supernatant of the infected cultures was removed and replaced by fresh medium containing the test compound at the particular concentration, The concentration of vital p24 antigen was determined in the supernatent by means of a commercial ELISA kit (Coulter) and served as a parameter for virus production. Compound B does not interfere with the p24 Elisa test (highest concentration tested: 100 μg/ml).
Mononuclear cells were isolated from healthy, HIV-negative donors using Ficoll density separation. Cells (4×106/ml) were incubated for 5 days in 48 well plates (Costar) in monocyte medium consisting of RPMII640, supplemented with 20% ECS and 10% human serum. On day 5 non-adherent cells were washed out four times with warm PBS containing 2% human serum. Preparations obtained by the procedure were >95% positive for non-specific esterase (Sigma) and cell viability (as determined by trypan blue exclusion) was always >95%.
The monocytotropic strain of HIV-1, BaL, was used for the infection of these monocyte preparations (Pemo et al, 1 Exp Meal, 169, 933, 1989).
Adherent monocytes were exposed to 50 μg/well of a 1:30 dilution of HIV-1, BaL for 30 minutes subsequently, monocyte medium was added to 1 ml/well. Adsorption was allowed for 24 hours at 37° C. Then, the wells were washed twice in order to remove excess virus and were cultivated in the presence of different drug concentrations. Thus, test compounds were added after adsorption. Every 3 to 4 days post infection the supernatant of the infected cultures was removed and replaced by fresh medium containing the test compound at the particular concentration. The concentration of viral p24 antigen was determined as described above.
IC50 and IC90 values were calculated by comparing the p24 antigen concentrations in supernatent of treated, infected cells and uncreated, infected cells at days 11 and 14 post infection.
Table 2 shows that Compound B is a potent inhibitor of HIV-1 replication in both primary cell types, with IC90 values of 1-2 ng/ml. At the highest concentration tested, 100 ng/ml, no cytotoxicity was observed.
TABLE 2
Activity of Compound B and AZT against HIV-a, IIIb,
replication in primary T4 lymphocytes and against HIV-1, BaL,
replication in primary monocytes
IC50 (μg/ml) IC90 (μg/ml)
Compound Cell Type day 11 day 14 day 11 day 14
B Lymphocytes <0.001 <0.001 <0.001 0.0010
AZT Lymphocytes 0.00045 0.00043 0.0022 0.0011
B Monocytes <0.001 0.0011 0.0019 0.0021
AZT Monocytes 0.0010 0.0010 0.0015 0.0017
Using the same methods, it was also shown that Compound B was a strong inhibitor of vital replication in primary T4 cells infected with low-passage primary clinical isolates of HIV-1 from three different geographical locations (K31, Zaire, D370, California, and K6/2, Germany).
The low cytotoxicity of Compound B was also shown by incubation of exponentially growing cells with Compound B or with AZT and determining cell numbers 2, 3 and 4 days after seeding. Compound B did not inhibit growth of MT4, MOLT4, HUT78, Jurkat cells (all T cell lines) nor the growth of the monocylic U937 cell line at concentrations below 300 μg/ml. With the exception of the HUT78 cells, AZT was in all cases more cytotoxic than Compound B with TC50 values (μg/ml) of 23, 37, 184 and 5 for MT4, MOLT4, Jurkat and U937 respectively.
In contrast to HIV-protease inhibitors, the compounds of the invention do not block virus production from chronically infected cells, indicating that the antiviral target is in the early part of the infection process, before, or at, integration of the provirus. To pinpoint the stage at which the compounds interact with the HIV replicative cycle, a time-of-addition experiment was carried out on MT4 cells infected with HIV-1 strain IIIb at high virus multiplicity to ensure that the virus replicative steps would be synchronised in the whole cell populations. Test compounds wife added 1, 2, 3, . . . 22, 23, 24 hours after infection, and vital p24 antigen production determined 29 hours after infection.
Depending on the stage at which compounds interact and the need for intracellular metabolism, addition of the compounds could be delayed for n hours without loss of activity. Daxtran sulphate, which acts at the virus adsorption step, must be added together with the virus (n=0) to be active. For AZT, which, following its intercellular phosphorylation, acts at the reverse transcriptase step, addition to the cells could be delayed until ca 4 hours (n=4) after infection. For the TIBO derivative (R82913), which does not need intracellular transformation before it can interact with reverse transcriptase the addition could be delayed by another 2 hours (n=6). The protease inhibitor Ro31-8959 which interacts with a late event in the virus cycle (assembly of mature virus) was still effective if added as late as 12 hours after infection (n=12). From the time-of-addition experiment appeared that for Compound B, n=1 or 2, so that the compound must interact with a process following virus absorption but preceding reverse transcription, for example, virus-cell fusion and/or uncoating.
To obtain further evidence for the inhibitory effect of Compound B on HIV uncoating (or fusion), experiments were designed whereby the viral RNA harvested from cells that had just been infected was monitored for its sensitivity to degradation by RNase. It was reasoned that if tincoating (fusion) was hampered, the viral capsid (or envelope) proteins would remain associated with the viral RNA genome and thus the RNA should be protected against RNase attack. When MT4 cells were exposed to radiolabelled viral particles at a very high multiple of infection and then treated with different concentrations of Compound B, viral RNA harvested from the cells 4 hours after infection showed resistance to degradation by RNaseA. Vital RNA harvested from HIV-infected cells treated with other anti-HIV agents (ie AZT, DDI, R82913, or Ro31-8959) did not show this increased resistance to degradation by RNase.
In addition, Compound B was also found to inhibit fusion, which is the mechanism by which viruses enter cells and by which virus or infectious material is transmitted from cell to cell. Syncytium formation between chronically infected cells and uninfected cells reflects the gp120/41 mediated fusion process of vital entry. The syncytium inhibition assay (Baba et al, J AIDS 3 493, 1990)) using HIV-1 IIIb infected HUT78 cells with MOLT4 cells indicates that Compound B is at least as potent as dextran sulphate in inhibition of fusion. The concentrations required (approximately 1 μg/ml) are considerably higher than the antiviral IC50 values, but are well below cytotoxicity levels.
These results strongly indicate that the compounds of the invention inhibit primarily the uncoating step and also to some extent the fusion step of the vital replicative cycle. This is a unique mode of action for anti-HIV agents, and the involvement of two distinct target steps makes it less likely that resistance to the drug will develop rapidly in treated patients.
Although no suitable animal models exist for the testing of in vivo efficacy of anti-HIV agents, testing of drug serum levels in the rabbit was carried out, and after sc administration of 10 mg/kg of Compound B, samples of rabbit serum were taken. Measurement of anti-HIV activity in the sen showed levels of the drug exceeding the in vitro IC50 level by a factor of a hundred for at least 6 hours after administration. This indicates that the compound would have anti-HIV activity in humans or an animal susceptible to infection by HIV.
The compounds of Formula I axe therefore useful for the treatment and/or prophylaxis of HIV infection, alone or in combination with other active agents. The appropriate dosage will, of course, vary depending upon, for example, the compound of Formula I employed, the host, the mode of administration and the nature and severity of the conditions being treated. However, in general, satisfactory results in humans are indicated to be obtained at daily dosages from about 0.01-20 mg/kg of body weight. An indicated daily dosage in humans is in the range from about 0.7 mg to about 1400 mg of a compound of Formula I conveniently administered for example in divided doses up to four times a day.
The compounds of Formula I may be administered by any conventional route, particularly enterally, preferably orally, eg in the loan of tablets or capsules or in liquid form, eg as a syrup; or parenterally, eg in the form of solutions or suspensions for iv or sc administration.
Compound B is the preferred compound of Formula I. In view of its activity in the test methods as described above, it is indicated that Compound B may be administered to humans at daily dosages of from 2 to 200 mg, preferably 10 to 70 mg, by parentexal administration, eg by subcutaneous injection.
The compounds of Formula I may be administered in free base form or in pharmaceutically acceptable acid addition salt or metal complex form. Such salts and complexes may be prepaxed in conventional manner as described in the Examples, and exhibit the same order of activity as the free bases. Pharmaceutical compositions containing compounds of Formula I may be manufactured in conventional manner. Unit dosage forms contain for example from about 0.5 mg to about 100 mg of a compound of Formula I in free base or pharmaceutically acceptable acid addition salt form.

Claims (52)

We claim:
1. A pharmaceutical composition active against HIV comprising as an active ingredient a linked cyclic compound of formula I,

Z-R-A-R′-Y  (I)
in which Z and Y are identical cyclic polyamine moieties having from 10 to 15 ring members and from 3 to 6 4 amine nitrogens in the ring spaced by 2 or more carbon atoms from each other, said amine nitrogens being the only ring heteroatoms,
A is an unsubstituted aromatic or heteroaromatic moiety other than quinoline,
R and R′ are each methylene linked to nitrogen atoms in Z and Y,
the amine nitrogen atoms being otherwise unsubstituted, in admixture or association with a pharmaceutically acceptable diluent or carrier.
2. A composition according to claim 1, wherein in the compound of formula I, each moiety Z and Y has 14 ring members and 4 amine nitrogens in the ring.
3. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[1,3-phenylenebis(methylene)]-bis-1,4,8,11-tetra-azacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
4. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[1,4-phenylenebis(methylene)]-bis-1,4,8,11-tetra-azacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
5. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is a bis-zinc complex of 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
6. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is a bis-copper complex of 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
7. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[3,3′-biphenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
8. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 11,11′-[1,4-phenylene-bis-(methylene)]-bis-1,4,7,11-tetraazacyclotetradecane 11,11′-[1,4 -phenylene-bis-(methylene)]-bis- 1,4,7,11 -tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
9. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,11′-[1,4-phenylene-bis-(methylene)]-1,4,8,11-tetraazacyclotetradecane-1,4,7,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
10. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[2,6-pyridine-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
11. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1-[3,5-pyridine-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
12. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[2,5-thiophene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
13. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[4,4′-(2,2′-bipyridine)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
14. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[2,9-(1,10-phenanthroline)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
15. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[1,3-phenylene-bis-(methylene)]-bis-1,4,7,10-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
16. A pharmaceutical composition according to claim 1, wherein the active against HIV comprising as an active ingredient is 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,7,10-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
17. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[5-nitro-1,3-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
18. The compound of claim 1, which is 1′1′-[2,4,5,6-tetrachloro-1,3-phenyleneis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane A pharmaceutical composition active against HIV comprising as an active ingredient 11′-[2,4,5,6 -tetrachloro- 1,3 -phenylenebis(methylene)]bis- 1,4,8,11 -tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
19. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,3,5,6-tetrafluoro-1,4-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
20. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4-naphthylene-bis-(methylene)]bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
21. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,3-phenylenebis-(methylene)]bis-1,5,9-triazacyclododecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
22. The compound of claim 1, which is 1,1′-[1,4-phenylene-bis-(methylene)]-1,5,9-triazacyclododecane A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4 -phenylene-bis-(methylene)]-bis- 1,5,9 -triazacyclododecane in acid addition salt form in admixture or association with a pharmaceutically acceptable diluent or carrier.
23. The compound of claim 1, which is a bis-zinc complex of 1,1′[1,4-phenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane.
24. The compound of claim 1, which is 1,1′-[3,3′-biphenylene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
25. The compound of claim 1, which is 1,1′-[2,6-pyridine-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
26. The compound of claim 1, which is 1,1′-[3,5-pyridine-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
27. The compound of claim 1, which is 1,1′-[2,5-thiophene-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
28. The compound of claim 1, which is 1,1′-[4,4′-(2,2′-bipyridine)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
29. The compound of claim 1, which is 1,1′-[2,9-(1,10-phenanthroline)-bis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in acid addition salt form.
30. The compound of claim 1, which is 1,1′-[1,3-phenylene-bis-(methylene)]-bis-1,4,7,10-tetraazacyclotetradecane in acid addition salt form.
31. The compound of claim 1, which is 1,1′-[1,4-phenylene-bis-(methylene)]-bis-1,4,7,10-tetraazacyclotetradecane in acid addition salt form.
32. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,5-dimethyl-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
33. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,5-dichloro-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
34. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2-bromo-1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
35. The compound of claim 1, which is A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[6-phenyl-2,4-pyridinebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
36. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,3 -phenylenebis(methylene)]-bis- 1,4,8,11 -tetra-azacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
37. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4 -phenylenebis(methylene)]-bis- 1,4,8,11 -tetra-azacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
38. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[3,3-biphenylene-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
39. A pharmaceutical composition active against HIV comprising as an active ingredient 11,11′-[1,4 -phenylene-bis-(methylene)]-bis- 1,4,7,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
40. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,6 -pyridine-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
41. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1-[3,5 -pyridine-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
42. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,5 -thiophene-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
43. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[4,4-( 2,2-bipyridine)-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
44. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,9 -( 1,10 -phenanthroline)-bis-(methylene)]-bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
45. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,3 -phenylene-bis-(methylene)]-bis- 1,4,7,10 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
46. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4 -phenylene-bis-(methylene)]-bis- 1,4,7,10 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
47. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[5 -nitro- 1,3 -phenylenebis(methylene)]bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
48. A pharmaceutical composition active against HIV comprising as an active ingredient 11′-[2,4,5,6 -tetrachloro- 1,3 -phenylenebis(methylene)]bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
49. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[2,3,5,6 -tetra-fluoro- 1,4 -phenylenebis(methylene)]bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
50. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4 -naphthylene-bis-(methylene)]bis- 1,4,8,11 -tetraazacyclotetradecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
51. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,3 -phenylenebis-(methylene)]bis- 1,5,9 -triazacyclododecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
52. A pharmaceutical composition active against HIV comprising as an active ingredient 1,1′-[1,4 -phenylene-bis-(methylene)]-bis- 1,5,9 -triazacyclododecane in admixture or association with a pharmaceutically acceptable diluent or carrier.
US12/192,704 1991-12-16 1992-12-16 Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity Expired - Lifetime USRE42152E1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB919126677A GB9126677D0 (en) 1991-12-16 1991-12-16 Improvements in chemical compounds
PCT/GB1992/002334 WO1993012096A1 (en) 1991-12-16 1992-12-16 Linked cyclic polyamines with activity against hiv

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/244,863 Reissue US5583131A (en) 1991-12-16 1992-12-16 Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity

Publications (1)

Publication Number Publication Date
USRE42152E1 true USRE42152E1 (en) 2011-02-15

Family

ID=10706343

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/244,863 Ceased US5583131A (en) 1991-12-16 1992-12-16 Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity
US12/192,704 Expired - Lifetime USRE42152E1 (en) 1991-12-16 1992-12-16 Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/244,863 Ceased US5583131A (en) 1991-12-16 1992-12-16 Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity

Country Status (26)

Country Link
US (2) US5583131A (en)
EP (2) EP0619813B1 (en)
JP (1) JP3375961B2 (en)
KR (1) KR100286235B1 (en)
AT (1) ATE273964T1 (en)
AU (1) AU661086B2 (en)
CA (1) CA2125978C (en)
CZ (1) CZ286928B6 (en)
DE (2) DE122010000001I1 (en)
DK (1) DK0619813T3 (en)
ES (1) ES2224096T3 (en)
FI (1) FI942849A (en)
GB (1) GB9126677D0 (en)
HK (2) HK1014368A1 (en)
HU (1) HU224013B1 (en)
IL (1) IL103984A (en)
MX (1) MX9207316A (en)
MY (1) MY110897A (en)
NL (1) NL300425I1 (en)
NO (2) NO305984B1 (en)
NZ (1) NZ246179A (en)
PL (1) PL173643B1 (en)
PT (1) PT619813E (en)
RU (1) RU94031208A (en)
WO (1) WO1993012096A1 (en)
ZA (1) ZA929632B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080300165A1 (en) * 2004-11-05 2008-12-04 The General Hospital Corporation Purposeful Movement Of Human Migratory Cells Away From An Agent Source
WO2012158707A1 (en) 2011-05-16 2012-11-22 Genzyme Corporation Use of cxcr4 antagonists
WO2015031722A1 (en) 2013-08-30 2015-03-05 Ramot At Tel-Aviv University Ltd. Method for treating amyotrophic lateral sclerosis by inhibition of cxcr4/cxcl12 signaling

Families Citing this family (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9126677D0 (en) 1991-12-16 1992-02-12 Johnson Matthey Plc Improvements in chemical compounds
GB9318550D0 (en) * 1993-09-07 1993-10-20 Nycomed Salutar Inc Chelants
GB9400411D0 (en) * 1994-01-11 1994-03-09 Johnson Matthey Plc Improvements in chemical compounds
US5663161A (en) 1995-02-17 1997-09-02 The Research Foundation Of State University Of New York Anti-viral triaza compounds
US5612478A (en) * 1995-03-30 1997-03-18 Johnson Matthey Plc Process for preparing 1,1'-[1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
US5606053A (en) * 1995-05-02 1997-02-25 Johnson Matthey Plc Process for preparing 1,1'-[1,4-phenylenebis-(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane
GB9511357D0 (en) * 1995-06-06 1995-08-02 Johnson Matthey Plc Improved antiviral compounds
US6506770B1 (en) * 1996-06-06 2003-01-14 Anormed, Inc. Antiviral compounds
US5608061A (en) * 1995-08-02 1997-03-04 Johnson Matthey Plc Process for preparing 1,4,8,11-tetraazacyclotetradecane
US7087648B1 (en) * 1997-10-27 2006-08-08 The Regents Of The University Of California Methods for modulating macrophage proliferation using polyamine analogs
ATE315390T1 (en) 1998-11-17 2006-02-15 Smithkline Beecham Corp CYCLIC POLYAMINES FOR THE TREATMENT OF THROMBOCYTOPENIA
US6365583B1 (en) 1999-02-02 2002-04-02 Anormed, Inc. Methods to enhance white blood cell count
US6750348B1 (en) 1999-03-24 2004-06-15 Anormed, Inc. Chemokine receptor binding heterocyclic compounds
AU775123B2 (en) * 1999-03-24 2004-07-15 Anormed Inc. Chemokine recpetor binding heterocyclic compounds
WO2000066112A1 (en) * 1999-05-03 2000-11-09 Smithkline Beecham Corporation Cxcr-4 receptor antagonists - thrombopoietin mimetics
WO2001044229A1 (en) * 1999-12-17 2001-06-21 Anormed Inc. Chemokine receptor binding heterocyclic compounds
CA2400157A1 (en) * 2000-02-22 2001-08-30 Ram W. Sabnis Organic polymeric antireflective coatings deposited by chemical vapor deposition
PT1317451E (en) 2000-09-15 2006-12-29 Anormed Inc Chemokine receptor binding heterocyclic compounds
MXPA03002325A (en) * 2000-09-15 2003-06-24 Anormed Inc Chemokine receptor binding heterocyclic compounds.
CA2419224A1 (en) 2000-09-15 2002-03-21 Anormed Inc. Chemokine receptor binding heterocyclic compounds
PL359292A1 (en) 2000-09-29 2004-08-23 Anormed Inc. Process for preparation of n-1 protected n ring nitrogen containing cyclic polyamines and products thereof
DE10117206A1 (en) * 2001-04-06 2002-10-10 Bayer Ag Process for the preparation of halogen-substituted dibenzyl alcohols, these halogen-substituted dibenzyl alcohols and their use
US7169750B2 (en) * 2001-07-31 2007-01-30 Anormed, Inc. Methods to mobilize progenitor/stem cells
KR100996378B1 (en) 2001-07-31 2010-11-25 아노르메드 인코포레이티드 A pharmaceutical composition for use in transplantation by mobilizing progenitor/stem cells
CN101353324A (en) * 2001-09-12 2009-01-28 阿诺麦德股份有限公司 Synthesis of enantiomerically pure amino-substituted fused bicyclic rings
IL161784A0 (en) * 2001-12-21 2005-11-20 Anormed Inc Chemokine receptor binding heterocyclic compounds with enhanced efficacy
US7354932B2 (en) * 2001-12-21 2008-04-08 Anormed, Inc. Chemokine receptor binding heterocyclic compounds with enhanced efficacy
KR101010905B1 (en) * 2002-01-18 2011-01-25 아스텔라스세이야쿠 가부시키가이샤 2-acylaminothiazole derivavtive or salt thereof
EP1613613B1 (en) 2003-04-11 2021-06-02 Genzyme Corporation Cxcr4 chemokine receptor binding compounds
US7501518B2 (en) * 2003-04-22 2009-03-10 Genzyme Corporation Methods of making 2,6-diaryl piperidine derivatives
ES2393188T3 (en) * 2003-04-22 2012-12-19 Genzyme Corporation Heterocyclic compounds that bind to chemokine receptors and have improved efficacy
US7498346B2 (en) * 2003-12-11 2009-03-03 Genzyme Corporation Chemokine receptor binding compounds
WO2005059107A2 (en) * 2003-12-11 2005-06-30 Anormed Inc. Chemokine receptor binding compounds
JP4870660B2 (en) * 2004-03-15 2012-02-08 アノーメッド インコーポレイティド CXCR4 antagonist synthesis process
ATE492523T1 (en) * 2004-08-05 2011-01-15 Sumitomo Chemical Co METHOD FOR PRODUCING HALOGEN-SUBSTITUTED BENZENEDIMETHANOL
EP1796716A4 (en) * 2004-08-13 2010-09-08 Anormed Inc Chemokine combinations to mobilize progenitor/stem cells
TW200619206A (en) * 2004-09-29 2006-06-16 Anormed Inc Chemokine-binding heterocyclic compound salts, and methods of use thereof
JP2008543858A (en) * 2005-06-15 2008-12-04 ジェンザイム・コーポレーション Chemokine receptor binding compounds
WO2007022371A2 (en) * 2005-08-16 2007-02-22 Genzyme Corporation Chemokine receptor binding compounds
CN101365336B (en) * 2005-08-19 2013-05-29 健赞股份有限公司 Methods to enhance chemotherapy
CN101355968A (en) * 2005-11-08 2009-01-28 安斯泰来制药有限公司 Compositions and methods for treating thrombocytopenia
ES2366745T3 (en) 2006-01-24 2011-10-25 Sumitomo Chemical Company Limited METHOD FOR PRODUCING BENCENDIMETHANOL REPLACED WITH HALOGEN.
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
BRPI0714799A2 (en) * 2006-08-02 2013-05-21 Genzyme Corp combination therapy
EP2056853A1 (en) * 2006-08-07 2009-05-13 Genzyme Corporation Combination therapy
AU2007284644B2 (en) * 2006-08-08 2011-09-01 Akarx, Inc. Compositions and methods for increasing blood platelet levels in humans
US20080214509A1 (en) * 2007-03-02 2008-09-04 Robert Kerbel Methods for enhancing the efficacy of vascular disrupting agents
EP2164953A4 (en) 2007-06-18 2010-06-30 Childrens Hosp & Res Ct Oak Method of isolating stem and progenitor cells from placenta
US9822364B2 (en) 2008-03-11 2017-11-21 Yale University Compositions and methods for controlled delivery of inhibitory ribonucleic acids
US9241898B2 (en) 2008-03-11 2016-01-26 Yale University Compositions and methods for controlled delivery of inhibitory ribonucleic acids
EP2149567A1 (en) * 2008-07-18 2010-02-03 Bayer Schering Pharma Aktiengesellschaft Cyclic polyamines for binding phosphatidylserine
US8771677B2 (en) * 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
US9895451B2 (en) 2011-12-02 2018-02-20 Yale University Formulations for targeted release of agents to low pH tissue environments or cellular compartments and methods of use thereof
US10465042B2 (en) 2011-12-02 2019-11-05 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
WO2013082529A1 (en) 2011-12-02 2013-06-06 Yale University Enzymatic synthesis of poly(amine-co-esters) and methods of use thereof for gene delivery
WO2014125499A1 (en) 2013-02-13 2014-08-21 Natco Pharma Limited Improved and commercially viable process for the preparation of high pure plerixafor base
PT107018A (en) * 2013-06-20 2014-12-22 Inst Superior Técnico SYNTHESIS AND USE OF CYCLAMS WITH ANTIBACTERIAL ACTIVITY
EP3721875B1 (en) 2014-05-09 2023-11-08 Yale University Particles coated with hyperbranched polyglycerol and methods for their preparation
US11918695B2 (en) 2014-05-09 2024-03-05 Yale University Topical formulation of hyperbranched polymer-coated particles
WO2016081621A1 (en) 2014-11-18 2016-05-26 Yale University Formulations for targeted release of agents under low ph conditions and methods of use thereof
US10682422B2 (en) 2014-11-18 2020-06-16 Yale University Formulations for targeted release of agents under low pH conditions and methods of use thereof
EP3050574B1 (en) 2015-01-28 2019-10-09 Universite De Bordeaux Use of plerixafor for treating and/or preventing acute exacerbations of chronic obstructive pulmonary disease
KR20180021685A (en) 2015-04-25 2018-03-05 더 제너럴 하스피털 코포레이션 Antitumor agent and anticancer drug combination therapy and composition for cancer treatment
AU2016269839B2 (en) 2015-06-03 2021-07-08 The University Of Queensland Mobilizing agents and uses therefor
US10544109B2 (en) 2015-09-02 2020-01-28 Fresenius Kabi Oncology Ltd. Process for the preparation of xylene linked cyclam compounds
CN108348606A (en) 2015-09-18 2018-07-31 综合医院公司以麻省总医院名义经营 Local delivery of anti-fugetactic agents for the treatment of cancer
US11357742B2 (en) 2015-12-14 2022-06-14 X4 Pharmaceuticals, Inc. Methods for treating cancer
EP3389634B1 (en) 2015-12-14 2021-10-06 X4 Pharmaceuticals, Inc. Methods for treating cancer
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
WO2017112894A1 (en) 2015-12-22 2017-06-29 X4 Pharmaceuticals, Inc. Methods for treating immunodeficiency disease
US20200308590A1 (en) 2016-02-16 2020-10-01 Yale University Compositions and methods for treatment of cystic fibrosis
AU2017221405A1 (en) 2016-02-16 2018-09-20 Carnegie Mellon University Compositions for enhancing targeted gene editing and methods of use thereof
CA3019394A1 (en) 2016-04-08 2017-10-12 X4 Pharmaceuticals, Inc. Methods for treating cancer
WO2017197128A1 (en) 2016-05-11 2017-11-16 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
EP3808748A1 (en) 2016-06-21 2021-04-21 X4 Pharmaceuticals, Inc. Substituted piperidines as cxcr4-inhibitors
WO2017223229A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
CN109641838A (en) 2016-06-21 2019-04-16 X4 制药有限公司 CXCR4 inhibitor and application thereof
WO2018049120A1 (en) 2016-09-09 2018-03-15 The General Hospital Corporation Ex vivo antigen-presenting cells or activated cd-positive t cells for treatment of cancer
US11453706B2 (en) 2016-09-09 2022-09-27 The General Hospital Corporation HSP fusion protein with anti-chemorepellant agent for treatment of infectious disease
WO2018049118A1 (en) 2016-09-09 2018-03-15 The General Hospital Corporation Hsp fusion protein with anti-chemorepellant agent for treatment of cancer
WO2018053270A1 (en) 2016-09-16 2018-03-22 The General Hospital Corporation Modified natural killer cells for the treatment of cancer
WO2018106738A1 (en) 2016-12-05 2018-06-14 Massachusetts Institute Of Technology Brush-arm star polymers, conjugates and particles, and uses thereof
MX2019011324A (en) 2017-03-23 2020-01-21 Massachusetts Gen Hospital Cxcr4/cxcr7 blockade and treatment of human papilloma virus-associated disease.
US20200113821A1 (en) 2017-04-04 2020-04-16 Yale University Compositions and methods for in utero delivery
EP3703715A1 (en) 2017-10-31 2020-09-09 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
KR20200096942A (en) 2017-12-06 2020-08-14 마젠타 테라퓨틱스 인코포레이티드 Dosing regimen for mobilization of hematopoietic stem and progeny 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
US20200338132A1 (en) 2018-01-03 2020-10-29 Magenta Therapeutics Inc. Compositions and methods for the expansion of hematopoietic stem and progenitor cells and treatment of inherited metabolic disorders
US20210189431A1 (en) 2018-08-10 2021-06-24 Yale University Compositions and methods for embryonic gene editing in vitro
US10548889B1 (en) 2018-08-31 2020-02-04 X4 Pharmaceuticals, Inc. Compositions of CXCR4 inhibitors and methods of preparation and use
US11814464B2 (en) 2019-04-29 2023-11-14 Yale University Poly(amine-co-ester) polymers and polyplexes with modified end groups and methods of use thereof
WO2020257776A1 (en) 2019-06-21 2020-12-24 Yale University Peptide nucleic acid compositions with modified hoogsteen binding segments and methods of use thereof
WO2020257779A1 (en) 2019-06-21 2020-12-24 Yale University Hydroxymethyl-modified gamma-pna compositions and methods of use thereof
US20220280656A1 (en) 2019-07-31 2022-09-08 Yale University Compositions and methods for treating sickle cell disease
US20220401481A1 (en) 2019-11-01 2022-12-22 Magenta Therapeutics, Inc. Dosing regimens for the mobilization of hematopoietic stem and progenitor cells
WO2021222313A1 (en) 2020-04-27 2021-11-04 Magenta Therapeutics, Inc. Methods and compositions for transducing hematopoietic stem and progenitor cells in vivo
EP4168471A1 (en) 2020-06-19 2023-04-26 Yale University Poly(amine-co-ester) polymers with modified end groups and enhanced pulmonary delivery
US20220031633A1 (en) 2020-07-28 2022-02-03 Yale University Poly(amine-co-ester) polymeric particles for selective pulmonary delivery
WO2022197776A1 (en) 2021-03-16 2022-09-22 Magenta Therapeutics, Inc. Dosing regimens for hematopoietic stem cell mobilization for stem cell transplants in multiple myeloma patients
WO2023056444A1 (en) 2021-10-01 2023-04-06 Janssen Pharmaceutica N.V. Methods of increasing progenitor cell production
US20230293448A1 (en) 2021-12-08 2023-09-21 Yale University Nanoparticle immunogenic compositions and vaccination methods
WO2023159189A1 (en) 2022-02-18 2023-08-24 Yale University Branched poly(amine-co-ester) polymers for more efficient nucleic expression

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0296522A2 (en) 1987-06-24 1988-12-28 The Dow Chemical Company Functionalized polyamine chelants and rhodium complexes thereof and process for their preparation
WO1991005762A1 (en) 1989-10-23 1991-05-02 Cockbain, Julian, Roderick, Michaelson Multi-site metal chelating agents
US5021409A (en) 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
US5047527A (en) 1989-03-20 1991-09-10 Centre National De La Recherche Scientifique (Cnrs) Process for the preparation of monofunctionalized cyclic tetramines
US5047572A (en) * 1988-11-18 1991-09-10 Ici Americas Inc. Trisubstituted benzoic acid intermediates
GB9126677D0 (en) 1991-12-16 1992-02-12 Johnson Matthey Plc Improvements in chemical compounds
EP0305320B1 (en) 1987-08-24 1992-09-23 Schering Aktiengesellschaft Multinucleic substituted complexants, complexes and complex salts, process for their preparation and pharmaceutical agents containing them
WO1992016494A1 (en) 1991-03-15 1992-10-01 Johnson Matthey Plc Long chain antiviral compounds
US5374416A (en) 1991-01-24 1994-12-20 Guerbet S.A. Nitrogenous macrocyclic ligands, polymetallic complexes and diagnostic and therapeutic composition

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0296522A2 (en) 1987-06-24 1988-12-28 The Dow Chemical Company Functionalized polyamine chelants and rhodium complexes thereof and process for their preparation
EP0305320B1 (en) 1987-08-24 1992-09-23 Schering Aktiengesellschaft Multinucleic substituted complexants, complexes and complex salts, process for their preparation and pharmaceutical agents containing them
US5047572A (en) * 1988-11-18 1991-09-10 Ici Americas Inc. Trisubstituted benzoic acid intermediates
US5047527A (en) 1989-03-20 1991-09-10 Centre National De La Recherche Scientifique (Cnrs) Process for the preparation of monofunctionalized cyclic tetramines
WO1991005762A1 (en) 1989-10-23 1991-05-02 Cockbain, Julian, Roderick, Michaelson Multi-site metal chelating agents
US5021409A (en) 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
EP0434385A2 (en) 1989-12-21 1991-06-26 Johnson Matthey Public Limited Company Use of macrocyclic nitrogen containing compounds for the treatment of retroviral infections
US5374416A (en) 1991-01-24 1994-12-20 Guerbet S.A. Nitrogenous macrocyclic ligands, polymetallic complexes and diagnostic and therapeutic composition
WO1992016494A1 (en) 1991-03-15 1992-10-01 Johnson Matthey Plc Long chain antiviral compounds
GB9126677D0 (en) 1991-12-16 1992-02-12 Johnson Matthey Plc Improvements in chemical compounds
WO1993012096A1 (en) 1991-12-16 1993-06-24 Johnson Matthey Public Limited Company Linked cyclic polyamines with activity against hiv

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
Alcock et al., "Studies of Pendant-arm Macrocyclic Ligands. Part 4. Two Penta-aza Macrocycles based on 1-(2'-Dimenthylaminoethyl)-1,5,9,13-tetra-azacyclohexadecane and its Complexes with Bivalent Metal Ions," J. Chem. Soc. Dalton Trans., pp. 1361-1364 (1985).
Baba et al., "Sulfated Polysaccharides as Potent Inhibitors of HIV-Induced Syncytium formation: A New Strategy Towards AIDS Chemotherapy," J. AIDS 3:493-499 (1990).
Barefield et al., "Characterization of 2,2'-Bi-(1,4,8,11-tetra-azacyclotetradecane): X-Ray Structure and Properties of the Dinuclear Complex [Ni2(C20H46N8)][CIO4]4," J.C.S. Chem. Comm., pp. 302-304 (1981).
Barefield et al., "Characterization of 2,2′-Bi-(1,4,8,11-tetra-azacyclotetradecane): X-Ray Structure and Properties of the Dinuclear Complex [Ni2(C20H46N8)][CIO4]4," J.C.S. Chem. Comm., pp. 302-304 (1981).
Bridger et al., "Synthesis and Structure-Activity Relationships of Phenylenebis(methylene)-Linked Bis-Tetraazamacrocycles That Inhibit HIV Replication. Effects of Macrocyclic Ring Size and Substituents on the Aromatic Linker," J. Med. Chem. 38:366-378 (1995).
Chandler et al., "Synthesis of some 2,9-Disubstituted-1,10-phenanthrolines," J. Heterocycl. Chem. 18(3):599-601 (1981).
Ciampolini, M. et al. "Dinickel and Dicopper Complexes with N,N-Linked Bis(cyclam) Ligands. An Ideal system for the Investigation of Electrostatic Effects on the Redox Behavior of Pairs of Metal Ions" Inorganic Chem. 26(21):3527-3533 (1987).
De Clercq et al.; Potent and selective inhibition of human immunodeficiency virus (HIV)-1 and HIV-2 replication by a class of bicyclams interacting with a viral uncoating event; Jun. 1992; Proc. Natl. Acad. Sci. USA; 89: 5286-5290. *
De Clercq, "The Bicyclam AMD3100 Story," Nature Reviews 2:581-587 (2003).
Diril, "Synthesis and Characterization of Binucleating Ligands and Their Manganese Complexes as Models for the Oxidation of Water to Molecular Oxygen," Dissertation, Rutgers, The State University of New Jersey (1988).
Diril, et al.; "Simulation strategies for unusual EPR spectra of binuclear mixed-valence manganese complexes: synthesis, properties, and x-ray structures of the MnIIMnIII complexes [Mn2(bpmp)(.mu.-OAc2](CIO4)2.cntdot.H2O and [Mn2(bcmp)(.mu.-OAc2](CIO4)2.cntdot.CH2CI2"; Journal of the American Chemical Society; 1989; 111 (14):5102-5114. *
Dischino et al., "Synthesis of Nonionic Gadolinium Chelates Useful as Contrast Agents for Magnetic Resonance Imaging. 1,4,7-Tris(carboxymethyl) -10-substituted-1,4,7,10-tetraazacyclododecanes and Their Corresponding Gadolinium Chelates," Inorg. Chem. 30:1265-1269 (1991).
Fabbrizzi et al., "Ditopic Receptors for Transition-Metal Ions: A Heterobimetallic Nickel(II)-Copper(II) Bis(macrocyclic) Complex and Its Stepwise Oxidation to the Tervalent State," Inorg. Chem. 25:2671-2672 (1986).
Griffing et al., "2,5-bis-(Chloromethyl)-thiophene and Some of its Derivatives," J. Am. Chem. Soc. 70:3416-3419 (1948).
Haeg et al., "Anthraquinone-Based Cyclophane Hosts: Synthesis and Complexation Studies," J. Am. Chem. Soc. 111(2):692-696 (1989).
Joao et al., "Quantitative Structural Activity Relationship Study of Bis-Tetraazacyclic Compounds. A Novel Series of HIV-1 and HIV-2 Inhibitors," J. Med. Chem. 38:3865-3873 (1995).
Mirriam-Webster's Collegiate Dictionary, 10th Edition p. 794 (2000).
Momenteau et al., "Both-faxes Hindered Porphyrins. Part 2. Synthesis and Characterization of internally Five-co-ordinated Iron (II) Basket-handle Porphyrins derived from 5,10,15,20-Tetrakis (o-hydroxyphenyl)porphyrin," J. Chem. Soc. Perkin Trans., pp. 61-70 (1985).
Pauwels et al., "Rapid and automated tetrazolium-based colorimetric assay for the detection of anti-HIV compounds," J. Virol. Methods 20:309-321 (1988).
Schneider et al., "Synthesis of Two Bis-tetraaza-macrocycles and Study of the Structures, Electrochemistry, VIS and EPR Spectra of their Binuclear Cu2+ and Ni2+ complexes," Helv. Chim. Acta. 68:53-61 (1985).
Schneider, R. et al. "Metal Complexes with Macrocyclic Ligands" Helvitica Chimica Acta 69(1):53-61 (1986).
U.S. Appl. No. 08/244,863, filed Aug. 18, 1994.
Wieghardt et al., "A Novel Mixed-valent MnIII-MnIV-Dimer,[L2Mn2(mu-O)2(mu-MeCO2)][BPh4]2 MeCN: Crystal Structure, Magnetic Properties, and E.S.R. Spectrum (L=1,4,7-triazacyclononane)," J. Chem. Soc. Chem. Commun., pp. 651-653 (1987).
Wieghardt et al., "Zweikernige Mangan(II,III,IV)-Modellkomplexe fuer das active Zentrum des Metalloproteins Photosystem II: Darstellung, Magnetismus und Kristallstruktur von [LMnIII(mu-CH3CO2)2MnIVL][CIO4]3(L=N,N',N''-Trimethyl-1,4,7-triazacyclononan)," Angew. Chem. 98:1026-1027 (1986).
Wieghardt et al., "A Novel Mixed-valent MnIII-MnIV-Dimer,[L2Mn2(μ-O)2(μ-MeCO2)][BPh4]2 MeCN: Crystal Structure, Magnetic Properties, and E.S.R. Spectrum (L=1,4,7-triazacyclononane)," J. Chem. Soc. Chem. Commun., pp. 651-653 (1987).
Wieghardt et al., "Zweikernige Mangan(II,III,IV)-Modellkomplexe fuer das active Zentrum des Metalloproteins Photosystem II: Darstellung, Magnetismus und Kristallstruktur von [LMnIII(μ-CH3CO2)2MnIVL][CIO4]3(L=N,N′,N″-Trimethyl-1,4,7-triazacyclononan)," Angew. Chem. 98:1026-1027 (1986).
Yaouanc, J-J. et al. "Mono N-Functionalization of Cyclic and Linear Tetraamines via Their Tridentate Tricarbonylchromium Complexes" J. Chem. Soc. Chem. Commun. 206-207 (1991).

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080300165A1 (en) * 2004-11-05 2008-12-04 The General Hospital Corporation Purposeful Movement Of Human Migratory Cells Away From An Agent Source
US9789171B2 (en) 2004-11-05 2017-10-17 The General Hospital Corporation Anti-fugetactic agents for the treatment of ovarian cancer
US10406217B2 (en) 2004-11-05 2019-09-10 The General Hospital Corporation Antifugetactic agents for the treatment of cancers
WO2012158707A1 (en) 2011-05-16 2012-11-22 Genzyme Corporation Use of cxcr4 antagonists
WO2015031722A1 (en) 2013-08-30 2015-03-05 Ramot At Tel-Aviv University Ltd. Method for treating amyotrophic lateral sclerosis by inhibition of cxcr4/cxcl12 signaling

Also Published As

Publication number Publication date
EP0619813A1 (en) 1994-10-19
NO942254L (en) 1994-06-15
MX9207316A (en) 1993-06-01
DE69233401T2 (en) 2005-10-13
HK1014368A1 (en) 1999-09-24
NO305984B1 (en) 1999-08-30
NO2010001I1 (en) 2010-03-08
HUT67544A (en) 1995-04-28
DK0619813T3 (en) 2004-12-20
NL300425I1 (en) 2010-01-04
NO2010001I2 (en) 2012-08-27
RU94031208A (en) 1996-09-10
JPH07501816A (en) 1995-02-23
CA2125978C (en) 2005-09-20
GB9126677D0 (en) 1992-02-12
US5583131A (en) 1996-12-10
IL103984A (en) 1998-10-30
EP0619813B1 (en) 2004-08-18
NO942254D0 (en) 1994-06-15
NZ246179A (en) 1996-03-26
HU9401786D0 (en) 1994-09-28
WO1993012096A1 (en) 1993-06-24
ZA929632B (en) 1993-06-18
AU661086B2 (en) 1995-07-13
IL103984A0 (en) 1993-05-13
DE122010000001I1 (en) 2010-08-26
FI942849A0 (en) 1994-06-15
MY110897A (en) 1999-06-30
FI942849A (en) 1994-06-15
CZ118894A3 (en) 1995-10-18
EP1223166A1 (en) 2002-07-17
CZ286928B6 (en) 2000-08-16
ES2224096T3 (en) 2005-03-01
KR100286235B1 (en) 2001-04-16
PL173643B1 (en) 1998-04-30
PT619813E (en) 2004-12-31
DE69233401D1 (en) 2004-09-23
CA2125978A1 (en) 1993-06-24
HK1049328A1 (en) 2003-05-09
JP3375961B2 (en) 2003-02-10
AU3165593A (en) 1993-07-19
HU224013B1 (en) 2005-04-28
ATE273964T1 (en) 2004-09-15

Similar Documents

Publication Publication Date Title
USRE42152E1 (en) Aromatic-linked polyamine macrocyclic compounds with anti-HIV activity
KR100420725B1 (en) The improved antiviral compound
EP0739345B1 (en) Cyclic polyamines
CA2130754C (en) Amine derivatives of oxo- and hydroxy-substituted hydrocarbons
AU2005249363A1 (en) Phosphonate analogs of HIV integrase inhibitor compounds
JP3480498B2 (en) Gallium (III) complex, method for producing the same, and pharmaceutical composition containing the same
JP2006505571A (en) Substituted indoles and their use as HCV inhibitors
CS331091A3 (en) Synergistic combination of compounds inhibiting hiv-reverse transcriptase and a pharmaceutical based thereon
US5681832A (en) Aroylaniline compounds, pharmaceutical compositions, and methods of using same to inhibit viral activity
JPH11500130A (en) Antiviral triaza compounds
WO1992016494A1 (en) Long chain antiviral compounds
ES2298077B1 (en) NEW POLYNITROGEN SYSTEMS AS ANTI-HIV AGENTS.
AU681342B2 (en) Amine derivatives of oxo- and hydroxy-substitued hydrocarbons
AU2003200066A1 (en) Amine Derivatives of Oxo- and Hydroxy-substituted Hydrocarbons
EP3423053A1 (en) Use of 2-oxo-2h-pyrrol-1(5h)-carboxamide derivatives as anti-hiv agents and process for the production thereof
ZA200210104B (en) Imidazopyridine and imidazopyrimidine antiviral agents.

Legal Events

Date Code Title Description
PTEF Application for a patent term extension

Free format text: PRODUCT NAME: MOZOBIL (PLERIXAFOR); REQUESTED FOR 5 YEARS

Filing date: 20090206

Expiry date: 20131210

PTEG Grant of a patent term extension

Free format text: PRODUCT NAME: MOZOBIL (PLERIXAFOR)

Filing date: 20090206

Expiry date: 20131210

AS Assignment

Owner name: JOHNSON MATTHEY PUBLIC LIMITED COMPANY, UNITED KIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRIDGER, GARY JAMES;PADMANAMHAN, SREENIVASAN;SKERLJ, RENATO TONY;AND OTHERS;SIGNING DATES FROM 19950831 TO 19950913;REEL/FRAME:030726/0699

Owner name: ANORMED INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JOHNSON MATTHEY PUBLIC LIMITED COMPANY;REEL/FRAME:030726/0837

Effective date: 19981125

Owner name: GENZYME CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ANORMED CORPORATION;REEL/FRAME:030727/0017

Effective date: 20080109

AS Assignment

Owner name: ANORMED CORPORATION, CANADA

Free format text: MERGER AND CHANGE OF NAME;ASSIGNORS:ANORMED INC.;DEMATAL CORP.;REEL/FRAME:033782/0221

Effective date: 20061117