WO2006124904A2 - Cancer treatment by combined inhibition of telomerase and hsp90 activities - Google Patents

Cancer treatment by combined inhibition of telomerase and hsp90 activities Download PDF

Info

Publication number
WO2006124904A2
WO2006124904A2 PCT/US2006/018927 US2006018927W WO2006124904A2 WO 2006124904 A2 WO2006124904 A2 WO 2006124904A2 US 2006018927 W US2006018927 W US 2006018927W WO 2006124904 A2 WO2006124904 A2 WO 2006124904A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
oligonucleotide
telomerase
cancer
subject
Prior art date
Application number
PCT/US2006/018927
Other languages
French (fr)
Other versions
WO2006124904A3 (en
Inventor
Nikhil C. Munshi
Masood A. Shammas
Sergei Gryaznov
Original Assignee
Geron Corporation
Dana-Farber Cancer Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Geron Corporation, Dana-Farber Cancer Institute filed Critical Geron Corporation
Publication of WO2006124904A2 publication Critical patent/WO2006124904A2/en
Publication of WO2006124904A3 publication Critical patent/WO2006124904A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/314Phosphoramidates
    • C12N2310/3145Phosphoramidates with the nitrogen in 3' or 5'-position
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • the invention is directed to anticancer treatment, and in particular to inhibition of tumor growth or cancer-cell proliferation, by treatment with a telomerase inhibitor in combination with an HSP90 inhibitor.
  • chemotherapeutic agents have been identified over the past few decades, and these are generally grouped into several categories on the basis of their mechanism of action. Combined-therapy treatments have become more common, in view of the perceived advantage of attacking the disease via multiple avenues. In practice, however, many such combinations do not provide even simple additivity of therapeutic effects.
  • a combined-drug approach for cancer treatment should provide a significant boost in efficacy and/or a significant reduction in undesired side effects, due to a reduced dose of the more toxic component and/or a reduction in the development of drug-resistance in the cancer being treated.
  • Particularly desirable are combination therapies which produce therapeutic results that are supraadditive or synergistic in nature relative to the effects of the individual agents, with minimal exacerbation of side effects.
  • the invention includes a method for inhibiting the proliferation of cancer cells, by (a) exposing the cells to an HSP90 inhibitor, and (b) either proceeding, following, or concomitantly with step (a), exposing the cells to a telomerase inhibitor.
  • the amount of HSP90 inhibitor to which the cells are exposed is effective, by itself, to inhibit proliferation of the cancer cells.
  • the amount of each inhibitor is effective, by itself, to inhibit proliferation of the cancer cells.
  • the combination provides a enhanced inhibiting effect relative to either component alone; more preferably, the combination provides a supraadditive or synergistic effect relative to the combined or additive effects of the components.
  • the telomerase inhibitor may include an oligonucleotide having nuclease- resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
  • the intern u cleoside linkages in the oligonucleotide may be selected from N3'->P5' phosphoramidate and N3'->P5' thiophosphoramidate linkages.
  • the telomerase inhibitor may include a lipid moiety, such as a fatty acid, sterol, or derivative thereof, which is attached covalently at one end of the oligonucleotide.
  • the oligonucleotide may be 10-20 bases in length, preferably 13-20 bases in length, and may have the sequence identified by SEQ ID NO:12 ( ⁇ '-TAGGGTTAGACAA-S 1 ).
  • One exemplary telomerase inhibitor is the compound designated herein as GRN163L.
  • the HSP90 inhibitor may include a benzoquinone ansamycin antibiotic, such as galdanamycin or an analogue thereof, as exemplified by 17-AAG (17-N- allylamino-17-demethoxygaldanamycin), or the closely related compounds herbimycin A, B, or C, and radicicol, or a pyrrolopyrimidine or pyrazolopyrimidine HSP90 inhibitor, such as disclosed in U.S. Patent Publications 2005/0107343A1 , and 2005/0119282 A1 , respectively.
  • the method may be used for use in treating a subject having a cancer, where, preferably, exposing step (a) includes administering the HSP90 inhibitor to the subject in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
  • each exposing step (a) and (b) includes administering the respective inhibitor to the subject in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
  • the method may be used in treating a variety of cancers in a subject.
  • the method is used in treating a subject diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain or myelomas.
  • telomerase inhibitor is the compound GRN163L
  • it may be administered to the subject by intravenous infusion, under infusion conditions effective to produce a blood concentration of the inhibitor of between 1 nM and 10O uM.
  • the invention is directed to a method for enhancing the anti-cancer treatment efficacy of an HSP90 inhibitor in a subject.
  • the method includes administering to the subject, before, during, or after administering the HSP90 inhibitor, an oligonucleotide telomerase inhibitor of the type composed of an oligonucleotide having nuclease-resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
  • the amount of the telomerase inhibitor is effective to inhibit the proliferation of cancer cells in the subject when the telomerase inhibitor is administered alone.
  • the enhancement of treatment efficacy may be evidenced, for example, by an increased survival time of the subject, or by an inhibition of tumor growth in the subject, relative to treatment with the HSP90 inhibitor alone.
  • the method may be used to treat a subject diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non- small-cell lung cancer, head, neck, and gastrointestinal cancers, prostate and testicular cancers, and melanomas.
  • breast cancer including metastatic breast cancer, ovarian cancer, lung cancer, including non- small-cell lung cancer, head, neck, and gastrointestinal cancers, prostate and testicular cancers, and melanomas.
  • the oligonucleotide may be 10-20 bases in length.
  • the oligonucleotide is 13-20 bases in length and includes the sequence identified by SEQ ID NO: 12 ( ⁇ '-TAGGGTTAGACAA-S 1 ).
  • An exemplary telomerase inhibitor is the compound identified as GRN163L, or an analog thereof.
  • This compound has (i) N3'->P5' thiophosphoramidate intemucleoside linkages in the oligonucleotide; (ii) the sequence identified as SEQ ID NO:12; and (iii) a palmitoyl (C16) moiety linked to the 5' end of the oligonucleotide through a glycerol or aminoglycerol linker.
  • One preferred HSP90 inhibitor is galdanamycin or an analog thereof, such as 17-AAG.
  • the HSP90 and telomerase inhibitors may be administered as a composition containing both inhibitors.
  • the invention may also be practiced to identify cancer patients who are candidates for effective anti-cancer treatment with a telomerase inhibitor.
  • the candidate patients are those whose cancer is responding to treatment with an HSP90 inhibitor, such as 17-AAG, but for whom combined treatment with a telomerase inhibitor is desired to enhance the anti-tumor efficacy of the HSP90 inhibitor alone.
  • kits for use in cancer therapy comprising (a) a dose of an HSP90 inhibitor, in an amount of the inhibitor effective, when administered alone, to inhibit the proliferation of cancer cells in the subject, and (b) a dose of an oligonucleotide telomerase inhibitor having nuclease-resistant intersubunit linkages, and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
  • the telomerase inhibitor is also provided in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
  • HSP90 inhibitor is 17-AAG
  • the telomerase inhibitor is the compound identified as GRN163L, or an analog thereof.
  • the latter compound has (i) N3'->P5' thiophosphoramidate internucleoside linkages in the oligonucleotide;(ii) the sequence identified as SEQ ID NO: 12; and (iii) a palmitoyl (C16) moiety linked to the 5' end of the oligonucleotide through a glycerol or aminoglycerol linker.
  • kits comprising a telomerase inhibitor and an HSP90 inhibitor, such as galdanamycin or an analog thereof, for use in therapy, such as the treatment of cancer.
  • therapy preferably comprises administering the HSP90 inhibitor to a subject, either preceding, following, or concomitantly with administration of said telomerase inhibitor.
  • the telomerase inhibitor is preferably a nuclease-resistant oligonucleotide which binds in a sequence-specific manner to the template region of hTR.
  • Preferred and/or exemplary inhibitors and cancer indications are as set out above.
  • the invention provides a kit containing a dose of an oligonucleotide telomerase inhibitor having nuclease-resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR, preferably in an amount effective to inhibit proliferation of cancer cells in a subject.
  • the kit may optionally include an insert that provides a user with one set of instructions for using the inhibitor in monotherapy and a separate set of instructions for using the inhibitor in combination with 17-AAG or other HSP90 inhibitor.
  • the set of instructions for the combination therapy may recommend (i) a lower dose of the telomerase inhibitor, when used in combination with the HSP90 inhibitor, (ii) a lower dose of the HSP90 inhibitor, when used in combination with the HSP90 inhibitor, and/or (iii) a different dosing regimen for one or both inhibitors, when used together, than would normally be recommended for the inhibitor(s) when used alone.
  • telomerase inhibitor for preparation of a medicament for use in treatment of cancer, wherein said treatment comprises administering said telomerase inhibitor to a subject in combination with an HSP90 inhibitor, such as galdanamycin or an analogue thereof.
  • Said treatment may comprise administering the HSP90 inhibitor to the subject either preceding, following, or concomitantly with the telomerase inhibitor, which is preferably a nuclease-resistant oligonucleotide which binds in a sequence-specific manner to the template region of hTR.
  • the invention provides the use of an HSP90 inhibitor and a telomerase inhibitor in the manufacture of a medicament for treating cancer in a subject.
  • Preferred and/or exemplary inhibitors and cancer indications are as set out above.
  • telomerase inhibitor in the manufacture of a medicament for treating cancer in a subject who is being treated with an HSP90 inhibitor, for the purpose of enhancing the anti-cancer efficacy of an HSP90 inhibitor in the subject.
  • Preferred and/or exemplary inhibitors and cancer indications are as set out above.
  • Figs. 1 -2 show enhancement of telomerase inhibiting activity of an NPS oligonucleotide hTR template inhibitor (GRN163) by conjugation to a lipid (to produce GRN163L), in human myeloma tumor xenografts (Fig. 1) and liver tissue (Fig. 2), respectively, in mice; and
  • GRN163 NPS oligonucleotide hTR template inhibitor
  • Fig. 3 illustrates the enhanced effect on inhibition of cancer cells achieved by co-administration of an HSP90 inhibitor, exemplified by 17-AAG and the telomerase inhibitor GRN163L, in INA6 myeloma cells.
  • a "polynucleotide” or “oligonucleotide” refers to a ribose and/or deoxyribose nucleoside subunit polymer or oligomer having between about 2 and about 200 contiguous subunits.
  • the nucleoside subunits can be joined by a variety of intersubunit linkages, including, but not limited to, phosphodiester, phosphodiester, methylphosphonate, P3'->N5' phosphoramidate, N3'->P5' phosphoramidate, N3'->'P5' thiophosphoramidate, and phosphorothioate linkages.
  • each linkage may be formed using the same chemistry, or a mixture of linkage chemistries may be used.
  • ATGUCCTG an oligonucleotide
  • the nucleotides are in 5'->3' order from left to right. Representation of the base sequence of the oligonucleotide in this manner does not imply the use of any particular type of internucleoside subunit in the oligonucleotide.
  • nucleoside includes the natural nucleosides, including 2'-deoxy and 2'-hydroxyl forms, e.g., as described in Komberg and Baker, DNA Replication, 2nd Ed. (Freeman, San Francisco, 1992), and analogs.
  • Aminalogs in reference to nucleosides, includes synthetic nucleosides having modified nucleobase moieties (see definition of "nucleobase” below) and/or modified sugar moieties, e.g., described generally by Scheit, Nucleotide Analogs (John Wiley, New York, 1980).
  • Such analogs include synthetic nucleosides designed to enhance binding properties, e.g., stability, specificity, or the like, such as disclosed by Uhlmann and Peyman (Chemical Reviews 90:543-584, 1990).
  • An oligonucleotide containing such nucleosides, and which typically contains synthetic nuclease-resistant internucleoside linkages, may itself be referred to as an "analog".
  • nucleobase includes (i) native DNA and RNA nucleobases (uracil, thymine, adenine, guanine, and cytosine), (ii) modified nucleobases or nucleobase analogs (e.g., 5-methylcytosine, 5-bromouracil, or inosine) and (iii) nucleobase analogs.
  • a nucleobase analog is a compound whose molecular structure mimics that of a typical DNA or RNA base.
  • lipid is used broadly herein to encompass substances that are soluble in organic solvents, but sparingly soluble, if at all, in water.
  • the term lipid includes, but is not limited to, hydrocarbons, oils, fats (such as fatty acids and glycerides), sterols, steroids and derivative forms of these compounds.
  • Preferred lipids are fatty acids and their derivatives, hydrocarbons and their derivatives, and sterols, such as cholesterol.
  • Fatty acids usually contain even numbers of carbon atoms in a straight chain (commonly 12-24 carbons) and may be saturated or unsaturated, and can contain, or be modified to contain, a variety of substituent groups.
  • fatty acid also encompasses fatty acid derivatives, such as fatty or esters.
  • hydrocarbon encompasses compounds that consist only of hydrogen and carbon, joined by covalent bonds.
  • the term encompasses open chain (aliphatic) hydrocarbons, including straight chain and branched hydrocarbons, and saturated as well as mono-and poly-unsaturated hydrocarbons.
  • the term also encompasses hydrocarbons containing one or more aromatic rings.
  • lipid also includes amphipathic compounds containing both lipid and hydrophilic moieties.
  • substituted refers to a compound which has been modified by the exchange of one atom or moiety for another, typically substitution of hydrogen by a different atom or moiety.
  • the term is used in reference to halogenated hydrocarbons and fatty acids, particularly those in which one or more hydrogen atoms are substituted with fluorine.
  • HSP90 refers to one of three distinct members of the HSP90 molecular chaperone family in mammalian cells.
  • the cytosolic heat shock protein HSP90 has two isoforms, HSP90A and HSP90B, which are 76% identical and are thought to be the consequence of a gene duplication event.
  • a third family member, the 94kDa glucose-regulated protein (GRP94) is localized primarily to the endoplasmic reticulum and shows 50% homology with HSP90.
  • HSP90 inhibitor is a compound capable of interacting with HSP90 protein within a cell, to inhibit or block the activity of the protein.
  • an HSP90 inhibitor acts by binding to the ATP/ADP binding pocket of HSP90 with a relatively high affinity, e.g., higher than the affinity for the adenine nucleotides.
  • HSP90 inhibitors include the benzoquinone ansamycin antibiotics, such as galdanamycin, galdanamycin analogs, such as 17-AAG, and herbicycin A, B, and C.
  • an "hTR template inhibitor” is a compound that blocks the template region (the region spanning nucleotides 30-67 of SEQ ID NO: 1 herein) of the RNA component of human telomerase, thereby inhibiting the activity of the enzyme.
  • the inhibitor is typically an oligonucleotide that is able to hybridize to this region.
  • the oligonucleotide includes a sequence effective to hybridize to a more specific portion of this region, having sequence 5'-CUAACCCUAAC-3' (SEQ ID NO: 12), spanning nucleotides 46-56 of SEQ ID NO: 1 herein.
  • a compound is said to "inhibit the proliferation of cancer cells” if the proliferation of cells in the presence of the compound is less than that observed in the absence of the compound. That is, proliferation of the cells is either slowed or halted in the presence of the compound. Inhibition of cancer-cell proliferation may be evidenced, for example, by reduction in the number of cells or rate of expansion of cells, reduction in tumor mass or the rate of tumor growth, or increase in survival rate of a subject being treated.
  • telomerase inhibitor administered to a subject is effective to "enhance the anti-cancer treatment efficacy of an HSP90 inhibitor" if the subject shows a reduced rate of tumor growth and/or an enhanced survival rate with combined therapy over therapy with the HSP90 inhibitor alone.
  • oligonucleotide having "nuclease-resistant linkages” refers to one whose backbone has subunit linkages that are substantially resistant to nuclease cleavage, in non-hybridized or hybridized form, by common extracellular and intracellular nucleases in the body; that is, the oligonucleotide shows little or no nuclease cleavage under normal nuclease conditions in the body to which the oligonucleotide is exposed.
  • the N3'->P5' phosphoramidate (NP) or N3'->P5' thiophosphoramidate (NPS) linkages described below are nuclease resistant.
  • HSP90 referring to any and all of the three distinct members of the HSP90 molecular chaperone family in mammalian cells, HSP90A and HSP90B and GRP94, is a highly abundant and essential cytosolic protein and the expression levels of HSP90 increases by environmental stress.
  • HSP90 functions as a molecular chaperone by binding to various cellular proteins and supporting the proper folding, stability, and function of target proteins.
  • the HSP90 client proteins include a wide variety of signal-transducing proteins that regulate cell growth and differentiation, such as protein kinases and steroid hormone receptors.
  • HSP90 functions in an ATP-dependent manner in cooperation with other molecular chaperones such as Cdc37 and FKBP52.
  • HSP90 inhibitors act by inhibiting the essential ATPase activity of HSP90.
  • an HSP90 inhibitor such as geldanamycin
  • HSP90 client proteins play important roles in the regulation of the cell cycle, cell growth, cell survival, apoptosis, and oncogenesis
  • an HSP90 inhibitor obstructs the proliferation of cancer cells and shows anti-cancer activity in experimental animals.
  • HSP90 inhibitor such as 17-AAG, both alone and in combination with other drugs, for a variety of cancers.
  • the compound is aimed at treating subjects diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain and myelomas, among other cancers.
  • HSP90 inhibitors include, but are not limited to, benzoquinone ansamycin antibiotics, such as geldanamycin, and its analogs, including 17-AAG (17-N-allylamino-17-demethoxygaldanamycin), 17-(2-dimethylaminoethyl)amino- 17-desmethoxy-geldanamycin (" 17-DMAG"), 11-0-methyl-17-(2-(1- azetidinyl)ethyl)amino-17-demethoxygeldanamycin, 11 -0-methyl-17-(2- dimethylaminoethyl)amino-17-demethoxygeldanamycin, and 11-0-methyl-17-(2- (1-pyrrolidinyl)ethyl)amino-17-demethoxygeldanamycin, and other galdanamysin analogs disclosed in one or more of U.S. Patent Nos. 4,261 ,989, 5,932,566, 6,890,917, 6,
  • HSP90 inhibitors include pyrrolopyrimidine HSP90 inhibitors, as described in U.S. Patent Publication 2005/0107343 A1 , and pyrazolopyrimidine HSP90 inhibitors, as described in U.S. Patent Publication 2005/0119282 A1 , both of which are incorporated herein by reference.
  • the HSP90 inhibitor is administered at a dose effective to produce a serum concentration between 0.01 ⁇ M and 10 ⁇ M, typically between 0.025 ⁇ M and 2 ⁇ M, e.g., at a concentration between 0.04 ⁇ M and 1 ⁇ M.
  • This concentration may be achieved at a dose between about 0.01 mg/kg and 20 mg/kg, typically between 0.1 mg/kg and 2 mg/kg, e.g., 0.5 mg/kg and 2 mg/kg.
  • a patient with a cancer every day for five days of each week of a treatment cycle, or an equivalent dose administered less often, e.g., once per week, over a treatment period with several cycles, e.g., five cycles.
  • about 5 mg/kg of 17- AAG could be administered once a week for each week of a treatment cycle, where the patient is treated 4 weeks per treatment cycle over a 5-cycle treatment regimen.
  • One advantage of the present invention is that the dose of the compound may be reduced when administered in combination with a telomerase inhibitor.
  • Telomerase is a ribonucleoprotein that catalyzes the addition of telomeric repeat sequences (having the sequence 5'-TTAGGG-3' in humans) to chromosome ends. See e.g. Blackburn, 1992, Ann. Rev. Biochem. ⁇ 1:113-129. The enzyme is expressed in most cancer cells but not in mature somatic cells. Loss of telomeric DNA may play a role in triggering cellular senescence; see Harley, 1991 , Mutation Research 256:271 -282.
  • telomere-positive A variety of cancer cells have been shown to be telomerase-positive, including cells from cancer of the skin, connective tissue, adipose, breast, lung, stomach, pancreas, ovary, cervix, uterus, kidney, bladder, colon, prostate, central nervous system (CNS), retina and hematologic tumors (such as myeloma, leukemia and lymphoma).
  • CNS central nervous system
  • hematologic tumors such as myeloma, leukemia and lymphoma.
  • Inhibitors of telomerase identified to date include oligonucleotides, preferably oligonucleotides having nuclease resistant linkages, as well as small molecule compounds.
  • telomere inhibitors of telomerase include, for example, BRACO19 ((9-(4-(N,N-dimethylamino)phenylamino)-3,6-bis(3-pyrrolodino propionamido)acridine (see MoI. Pharmacol. 61_(5):1154-62, 2002); DODC (diethyloxadicarbocyanine), and telomestatin. These compounds may act as G- quad stabilizers, which promote the formation of an inactive G-quad configuration in the RNA component of telomerase.
  • telomerase inhibitors of telomerase include BIBR1532 (2-[(E)-3-naphthen-2-yl but-2- enoylaminojbenzoic acid) (see Ward & Autexier, MoI. Pharmacol. 68:779-786, 2005; also J. Biol. Chem. 277(18): 15566-72, 2002); AZT and other nucleoside analogs, such as ddG and ara-G (see, for example, U.S. Patent Nos. 5,695,932 and 6,368,789), and certain thiopyridine, benzo[b]thiophene, and pyrido[b]thiophene derivatives, described by Gaeta et al.
  • telomere protein component the human form of which is known as human telomerase reverse transcriptase, or hTERT
  • RNA component of the telomerase holoenzyme the human form of which is known as human telomerase RNA, or hTR
  • RNA component of human telomerase hTR
  • SEQ ID NO: 1 The nucleotide sequence of the RNA component of human telomerase (hTR) is shown in the Sequence Listing below (SEQ ID NO: 1 ), in the 5'-»3' direction.
  • the sequence is shown using the standard abbreviations for ribonucleotides; those of skill in the art will recognize that the sequence also represents the sequence of the cDNA, in which the ribonucleotides are replaced by deoxyribonucleotides, with uridine (U) being replaced by thymidine (T).
  • the template sequence of the RNA component is located within the region defined by nucleotides 46-56 ( ⁇ '-CUAACCCUAAC-S 1 ), which is complementary to a telomeric sequence composed of about one-and-two-thirds telomeric repeat units.
  • the template region functions to specify the sequence of the telomeric repeats that telomerase adds to the chromosome ends and is essential to the activity of the telomerase enzyme (see e.g. Chen et al., Ce// 100:503-514, 2000; Kim et al., Proc. Natl. Acad. Sci. USA 98(14):7982-7987, 2001 ).
  • RNA small interfering RNA
  • Such agents may be designed to target the hTERT mRNA and thereby inhibit production of hTERT protein in a target cell, such as a cancer cell (see, for example, U.S. Pat. Nos. 6,444,650 and 6,331 ,399).
  • Oligonucleotides targeting hTR act as inhibitors of telomerase enzyme activity by blocking or otherwise interfering with the interaction of hTR with the hTERT protein, which interaction is necessary for telomerase function. See, for example, Villeponteau et al., U.S. Patent No. 6,548,298.
  • a preferred target region of hTR is the template region, spanning nucleotides 30-67 of SEQ ID NO:1.
  • Oligonucleotides targeting this region are referred to herein as "hTR template inhibitors" (see e.g. Herbert et al., Oncogene 21(4):638-42 (2002).)
  • hTR template inhibitors see e.g. Herbert et al., Oncogene 21(4):638-42 (2002).
  • such an oligonucleotide includes a sequence which is complementary or near-complementary to some portion of the 11- nucleotide region having sequence 5'-CUAACCCUAAC-S 1 , spanning nucleotides 46-56 Of SEQ ID NO: 12.
  • Another preferred target region is the region spanning nucleotides 137- 179 of hTR (see Pruzan et al., Nucl. Acids Research, 30:559-568, 2002). Within this region, the sequence spanning 141-153 is a preferred target.
  • PCT publication WO 98/28442 describes the use of oligonucleotides of at least 7 nucleotides in length to inhibit telomerase, where the oligonucleotides are designed to be complementary to accessible portions of the hTR sequence outside of the template region, including nucleotides 137-196, 290-319, and 350- 380 of hTR.
  • Preferred hTR targeting sequence are given below, and identified by SEQ ID NOS: 2-22.
  • the region of the therapeutic oligonucleotide that is targeted to the hTR sequence is preferably exactly complementary to the corresponding hTR sequence. While mismatches may be tolerated in certain instances, they are expected to decrease the specificity and activity of the resultant oligonucleotide conjugate.
  • the base sequence of the oligonucleotide is thus selected to include a sequence of at least 5 nucleotides exactly complementary to the hTR target, and enhanced telomerase inhibition may be obtained if increasing lengths of complementary sequence are employed, such as at least 8, at least 10, at least 12, at least 13 or at least 15 nucleotides exactly complementary to the hTR target.
  • the sequence of the oligonucleotide includes a sequence of from at least 5 to 20, from at least 8 to 20, from at least 10 to 20 or from at least 10 to 15 nucleotides exactly complementary to the hTR target sequence.
  • Optimal telomerase inhibitory activity may be obtained when the full length of the oligonucleotide is selected to be complementary to the hTR target sequence.
  • the oligonucleotide sequence may include regions that are not complementary to the target sequence. Such regions may be added, for example, to confer other properties on the compound, such as sequences that facilitate purification.
  • an oligonucleotide may include multiple repeats of a sequence complementary to an hTR target sequence.
  • telomerase RNA RNA
  • the intemucleoside linkages in the oligonucleotide may include any of the available oligonucleotide chemistries, e.g. phosphodiester, phosphotri ester, methylphosphonate, P3'->N5' phosphoramidate, N3'->P5' phosphoramidate, N3'->P5' thiophosphoramidate, and phosphorothioate.
  • all of the internucleoside linkages within the oligonucleotide will be of the same type, although the oligonucleotide component may be synthesized using a mixture of different linkages.
  • a particularly preferred sequence for an hTR template inhibitor oligonucleotide is the sequence complementary to nucleotides 42-54 of SEQ ID NO: 12 above.
  • the oligonucleotide having this sequence (TAGGGTTAGACA) and N3'->P5' thiophosphoramidate (NPS) linkages is designated herein as GRN163.
  • GRN163 N3'->P5' thiophosphoramidate linkages
  • this oligonucleotide inhibits telomerase at low concentrations in a biochemical assay (FlashPlateTM; see Experimental Section).
  • An alternative 13-mer having the sequence CAGTTAGGGTTAG, complementary to nucleotides 46-58 of SEQ ID NO: 1 (fifth row of table), showed near-equivalent activity in the FlashPlateTM assay.
  • the corresponding NP-linked oligonucleotide, and shorter (11- and 12-mer) oligonucleotides targeting the same region (complementary to nucleotides 42-53 and 42-42, respectively, of SEQ ID NO: 1), showed moderate activity. The effect is clearly sequence-specific, as shown by the mismatch and non-targeting sequences in the table.
  • the oligonucleotide GRN163 administered alone has shown inhibitory activity in vitro in cell culture, including epidermoid carcinoma, breast epithelium, renal carcinoma, renal adenocarcinoma, pancreatic, brain, colon, prostate, leukemia, lymphoma, myeloma, epidermal, cervical, ovarian and liver cancer cells.
  • the oligonucleotide GRN163 has also been tested and shown to be therapeutically effective in a variety of animal tumor models, including ovarian and lung, both small cell and non-small cell.
  • the oligonucleotide-based enzyme inhibitor includes at least one covalently linked lipid group (see US Pubn. No. 2005/0113325, which is incorporated herein by reference).
  • This modification provides superior cellular uptake properties, such that an equivalent biological effect may be obtained using smaller amounts of the conjugated oligonucleotide compared to the unmodified form. When applied to the human therapeutic setting, this may translate to reduced toxicity risks, and cost savings.
  • the lipid group L is typically an aliphatic hydrocarbon or fatty acid, including derivatives of hydrocarbons and fatty acids, with examples being saturated straight chain compounds having 14-20 carbons, such as myristic (tetradecanoic) acid, palmitic (hexadecanoic) acid, and stearic (octadeacanoic) acid, and their corresponding aliphatic hydrocarbon forms, tetradecane, hexadecane and octadecane.
  • suitable lipid groups that may be employed are sterols, such as cholesterol, and substituted fatty acids and hydrocarbons, particularly polyfluorinated forms of these groups.
  • the scope of the lipid group L includes derivatives such as amine, amide, ester and carbamate derivatives. The type of derivative is often determined by the mode of linkage to the oligonucleotide, as exemplified below.
  • the lipid moiety is palmitoyl amide (derived from palmitic acid), conjugated through an aminoglycerol linker to the 5' thiophosphate group of an NPS-linked oligonucleotide.
  • the NPS oligonucleotide having the sequence shown for GRN 163 and conjugated in this manner (as shown below) is designated GRN163L herein.
  • the lipid, as a palmitoyl amide is conjugated through the terminal 3' amino group of an NPS oligonucleotide.
  • Table 4 presents further data directed to telomerase inhibition in vitro by GRN163 (unconjugated) and GRN163L (lipidated) in various cancer cell lines.
  • the conjugated oligonucleotide GRN163L had significantly greater telomerase inhibiting activity in vivo than the unconjugated GRN163, as demonstrated in hepatoma cell xenografts (Fig. 1 ) and flank CAG myeloma tumor xenografts (Fig. 2) following i.v. administration.
  • GRN163L inhibited tumor growth in mice (A549-luc IV lung metastases model) for at least 4 weeks after i.v. injection of cancer cells.
  • the dosage was 1 ⁇ M biweekly for 5 weeks prior to injection of cancer cells, followed by 5 mg/kg twice weekly after injection. Controls showed substantial tumor growth, but none was apparent in the GRN163L-treated mouse.
  • telomerase inhibitor is an hTR template blocking agent, as described above.
  • chemotherapeutic treatment has a supraadditive effect; that is, the combined benefit is greater than would be expected simply from the additive effects of the two therapeutic approaches. This is demonstrated herein for the combination of a telomerase inhibitor and the HSP90 inhibitor 17-AAG.
  • a subject having a cancer type that is responsive to an HSP90 inhibitor, or a subject currently receiving cancer therapy with an HSP90 inhibitor is initially identified as a candidate for the combined therapy.
  • Preferred cancer indications include, for example, breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain and myelomas.
  • an aspect of the invention involves identifying cancer patients who are candidates for effective anti-cancer treatment with a telomerase inhibitor.
  • the candidate patients are those whose cancer is responding to treatment with an HSP90 inhibitor, e.g., 17-AAG, but for whom combined treatment with a telomerase inhibitor is desired to enhance the anti-tumor efficacy of the HSP90 inhibitor alone.
  • the cancer should also be one that is responsive to cancer-cell inhibition by telomerase inhibition.
  • oligonucleotide telomerase inhibitors as exemplified by GRN163 and GRN163L, have shown inhibitory activity in vitro against human kidney, lung, pancreatic, brain, colon, prostate, breast, leukemia, lymphoma, myeloma, epidermal, cervical, ovarian and liver cancer cells, and in vivo, via local and systemic delivery, against human brain, prostate, lymphoma, myeloma, cervical, lung, and liver cancer cells.
  • Other preferred targets include small cell lung, esophogeal, head and neck, and stomach cancers.
  • the subject is administered the HSP90 inhibitor, e.g., 17-AAG, in an amount that is effective inhibiting proliferation of cancer cells in the subject.
  • the dose administered and the dosing schedule will follow, for example, known or recommended doses for the HSP90 inhibitor employed, as indicated, for example, in the drug product insert or published clinical or animal-model data.
  • a patient will receive 0.1 mg/kg to 2 mg/kg
  • about 1 mg/kg of 17-AAG is administered to a patient with a cancer every day for five days of each week of a treatment cycle, where the patient is treated for 5 cycles.
  • about 10 mg/kg of 17-AAG is administered once a week for each week of a treatment cycle, where the patient is treated 4 weeks per treatment cycle over a 5-cycle treatment regimen.
  • kits containing a dose of the telomerase inhibitor could optionally contain a product insert having one set of directions for using the inhibitor in monotherapy, and another set of directions for using the inhibitor in a combination therapy with an HSP90 inhibitor, such as a 17-AAG.
  • the set of instructions for the combination therapy could recommend (i) a lower dose of the telomerase inhibitor, when used in combination with the HSP90 inhibitor, (ii) a lower dose of the HSP90 inhibitor, when used in combination with the telomerase inhibitor, and/or (iii) a different dosing regimen for one or both inhibitors, when used together, than would normally be recommended for the inhibitor(s) when used alone.
  • the telomerase inhibitor may be administered, before, during, or after administration of the HSP90 inhibitor.
  • the two inhibitors are administered in a common dosing regimen, as described below, and the two inhibitors themselves may be administered in a combined-drug composition, e.g., by IV administration, or separately.
  • a dosing regimen in which the telomerase inhibitor is administered before or after administering the HSP90 inhibitor is also contemplated.
  • a person under treatment with an HSP90 inhibitor may be subsequently placed on a combined therapy that includes telomerase inhibitor.
  • the patient may be initially administered the HSP90 inhibitor, followed one-to-several days later with the telomerase treatment.
  • the HSP90 inhibitor may function, in part, to sensitize the cancer cells to inhibition by a telomerase inhibition, e.g., by synchronizing the cell-division cycle
  • INA6 myeloma cells were treated with the HSP90 inhibitor 17-AAG in combination with a telomerase-inhibitor oligonucleotide targeted against hTR, with the results shown in Fig. 3. Briefly, INA6 myeloma cells were cultured for a seven-day period.
  • the cells were treated with one of (i) a mismatch oligonucleotide identical to GRN 163L except in its base sequence, at a concentration of 1 ⁇ M (solid diamonds), (ii) GRN163L, at final a concentration of 1 ⁇ M (solid triangles), (iii) 17-AGG, diamonds), at final a concentration of 0.05 ⁇ M (solid squares), and (iv) a combination of GRN 163L (1 ⁇ M) and 17-AAG 0.05 ⁇ M (X symbol).
  • aliquots from each culture were removed, and the percent of dead cells was measured.
  • the combined treatment was effective in killing more than 90% of the cells, compared with about 50% killing for each individual treatment over the same time period.
  • the therapeutic protocol for administering such combinations will depend on various factors including, but not limited to, the type of cancer, the age and general health of the patient, the aggressiveness of disease progression, the TRF length (terminal restriction fragment length; see Section V below) and telomerase activity of the diseased cells to be treated, and the ability of the patient to tolerate the agents that comprise the combination.
  • the target disease comprises a solid tumor; in other embodiments, the target disease comprises a hematological malignancy.
  • An exemplary course of treatment involves multiple doses. Sequence of combination treatments will be determined by clinical compliance criteria and/or preclinical or clinical data supporting dose optimization strategies to augment efficacy or reduce toxicity of the combination treatment. In general, various combinations of the telomerase inhibitor and HSP90 inhibitor may be employed, used either sequentially or simultaneously. For multiple dosages, the two agents may directly alternate, or two or more closes of one agent may be alternated with a single dose of the other agent, for example.
  • Simultaneous administration of both agents may also be alternated or otherwise interspersed with dosages of the individual agents.
  • the time between dosages may be for a period from about 1-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1 -2 days, to about 1 -2 wk or longer following the initiation of treatment.
  • the need to complete the planned dosings may be reevaluated.
  • the compounds may be administered by direct injection of a tumor or its vasculature.
  • the tumor may be infused or perfused with the therapeutic compounds using any suitable delivery vehicle.
  • the compounds may be administered locally to an affected organ.
  • Systemic administration may also be performed.
  • Continuous administration may be applied where appropriate; for example, where a tumor is excised and the tumor bed is treated to eliminate residual disease. Delivery via syringe or catheterization is preferred.
  • Such continuous perfusion may take place for a period from about 1-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 weeks or longer following the initiation of treatment.
  • the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs.
  • the therapeutic agents are administered to a subject, such as a human patient, in a formulation and in an amount effective to achieve a clinically desirable result.
  • desirable results include reduction in tumor mass (as determined by palpation or imaging; e.g., by radiography, radionucleotide scan, CAT scan, or MRI), reduction in the rate of tumor growth, reduction in the rate of metastasis formation (as determined e.g., by histochemical analysis of biopsy specimens), reduction in biochemical markers (including general markers such as ESR, and tumor specific markers such as serum PSA), and improvement in quality of life (as determined by clinical assessment, e.g., Karnofsky score), increased time to progression, disease-free survival and overall survival.
  • each agent per dose and the number of doses required to achieve such effects will vary depending on many factors including the disease indication, characteristics of the patient being treated and the mode of administration.
  • the formulation and route of administration will provide a local concentration at the disease site of between 1 nM and 100 ⁇ M of each agent, preferably between 0.01 ⁇ M to 10 ⁇ M, as noted above.
  • the physician will be able to vary the amount of the compounds, the carrier, the dosing frequency, and the like, taking into consideration such factors as the particular neoplastic disease state and its severity; the overall condition of the patient; the patient's age, sex, and weight; the mode of administration; the suitability of concurrently administering systemic anti-toxicity agents; monitoring of the patient's vital organ functions; and other factors typically monitored during cancer chemotherapy.
  • the compounds are administered at a concentration that affords effective results without causing excessive harmful or deleterious side effects.
  • the amount of the agent used in combination with a telomerase inhibitor may be less than would be required for the agent used in non-combination therapy.
  • the pharmaceutical carrier(s) employed may be solid or liquid.
  • Liquid carriers can be used in the preparation of solutions, emulsions, suspensions and pressurized compositions.
  • the compounds are dissolved or suspended in a pharmaceutically acceptable liquid excipient.
  • suitable examples of liquid carriers for parenteral administration include water (which may contain additives, e.g., cellulose derivatives, preferably sodium carboxymethyl cellulose solution), phosphate buffered saline solution (PBS), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) and their derivatives, and oils (e.g., fractionated coconut oil and arachis oil).
  • the liquid carrier can contain other suitable pharmaceutical additives including, but not limited to, the following: solubilizers, suspending agents, emulsifiers, buffers, thickening agents, colors, viscosity regulators, preservatives, stabilizers and osmolarity regulators.
  • the carrier can also be an oily ester such as ethyl oleate and isopropyl myristate.
  • Sterile carriers are useful in sterile liquid form compositions for parenteral administration. Sterile liquid pharmaceutical compositions, solutions or suspensions can be utilized by, for example, intraperitoneal injection, subcutaneous injection, intravenously, or topically. The compositions can also be administered intravascularly or via a vascular stent.
  • the liquid carrier for pressurized compositions can be a halogenated hydrocarbon or other pharmaceutically acceptable propellant.
  • Such pressurized compositions may also be lipid encapsulated for delivery via inhalation.
  • the compositions may be formulated into an aqueous or partially aqueous solution, which can then be utilized in the form of an aerosol.
  • compositions may be administered topically as a solution, cream, or lotion, by formulation with pharmaceutically acceptable vehicles containing the active compound.
  • the compositions of this invention may be orally administered in any acceptable dosage including, but not limited to, formulations in capsules, tablets, powders or granules, and as suspensions or solutions in water or nonaqueous media.
  • Pharmaceutical compositions and/or formulations comprising the oligonucleotides of the present invention may include carriers, lubricants, diluents, thickeners, flavoring agents, emulsifiers, dispersing aids or binders.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • Modes of administration and formulation may be dependent on the drug and its approved mode of administration.
  • the chemotherapeutic agent is 17-AAG
  • IV administration is indicated.
  • the telomerase inhibitor is GRN163L
  • formulation in 0.9% sodium chloride (normal saline) and administration by i.v. is a preferred route, preferably via infusion over 4-8 hours, e.g. a 6 hr infusion.
  • lipid-conjugated oligonucleotides described herein have superior characteristics for cellular and tissue penetration
  • these and other compounds may be formulated to provide further benefit in this area, e.g. in liposome carriers.
  • liposomes to facilitate cellular uptake is described, for example, in U.S. Pat. Nos. 4,897,355 and 4,394,448, and numerous publications describe the formulation and preparation of liposomes.
  • Liposomal formulations can also be engineered, by attachment of targeting ligands to the liposomal surface, to target sites of neovascularization, such as tumor angiogenic regions.
  • the compounds can also be formulated with additional penetration/transport enhancers, such as unconjugated forms of the lipid moieties described above, including fatty acids and their derivatives.
  • additional penetration/transport enhancers such as unconjugated forms of the lipid moieties described above, including fatty acids and their derivatives. Examples include oleic acid, lauric add, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, recinleate, . monoolein (a.k.a.
  • acylcarnitines acylcholines, mono- and di-glycerides and physiologically acceptable salts thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.).
  • Other useful adjuvants include substrates for transendothelial migration, such as glucose uptake systems for facilitated egress from the vascular space to the tumor microenvironment.
  • telomere length can be measured by a flow cytometry method using fluorescence in situ hybridization, referred to as flow FISH (see e.g. M. Hultdin et al., Nucleic Acids Res. 26(16):3651-6, 1998; N. Rufer et ai, Nature Biotechnology 16:743-7 , 1998).
  • TRF terminal restriction fragment
  • telomere inhibiting agent may be predicted for tumor cells having shorter telomeric DNA, although telomerase has been shown to have other inhibitory effects independent of telomere length.
  • telomerase has been shown to have other inhibitory effects independent of telomere length.
  • the TRAP assay (see Experimental, below) is a standard method for measuring telomerase activity in a cell extract system (Kim et al., Science 266:2011 , 1997; Weinrich et al., Nature Genetics 17:498, 1997). Briefly, this assay measures the amount of nucleotides incorporated into elongation products (polynucleotides) formed by nucleotide addition to a labeled telomerase substrate or primer.
  • the TRAP assay is described in detail in U.S. Pat. Nos. 5,629,154, 5,837,453 and 5,863,726, and its use in testing the activity of telomerase inhibitory compounds is described in various publications, including WO 01/18015.
  • kits are available commercially for research purposes for measuring telomerase activity: TRAPezeTM XK Telomerase Detection Kit (Intergen Co., Purchase N.Y.); and TeIoTAGGG Telomerase PCR ELISA plus (Roche Diagnostics, Indianapolis Ind.).
  • the anticancer activity of the therapeutic combinations can be evaluated using standard in vitro and in vivo assays.
  • the ability of a composition to specifically inhibit the growth of tumor cells can be assayed using tumor cell lines in vitro, or in xenograft animal models in vivo.
  • a preferred protocol for such growth curve assays is the short term cell viability assay described in Asai et al. (2003, cited above).
  • the test compound is administered either directly to the tumor site or systemically, and the growth of the tumor is followed by physical measurement.
  • a preferred example of a suitable in vivo tumor xenograft assay is also described in Asai et al. (2003, cited above).
  • a variety of synthetic approaches can be used to conjugate a lipid moiety L to the oligonucleotide, depending on the nature of the linkage selected; see, for example, Mishra ef al., Biochim. et Biophys. Acta 1264:229-237 (1995), Shea et al., Nucleic Acids Res. 18:3777-3783 (1995), or Rump et al., Bioconj. Chem. 9:341 -349 (1995).
  • conjugation is achieved through the use of a suitable functional groups at an oligonucleotide terminus.
  • the 3'- amino group present at the 3'-terminus of the NP and NPS oligonucleotides can be reacted with carboxylic acids, acid chlorides, anhydrides and active esters, using suitable coupling catalysts, to form an amide linkage.
  • Thiol groups are also suitable as functional groups (see Kupihar et al., Bioorg. Med. Chem. 9:1241-1247 (2001 )).
  • Various amino- and thiol-functionalized modifiers of different chain lengths are commercially available for oligonucleotide synthesis.
  • lipids may also be attached to the oligonucleotide chain using a phosphoramidite derivative of the lipid, to produce a phosphoramidate or thiophosphoramidate linkage connecting the lipid and the oligonucleotide.
  • the free 3'-amino group of the fully protected support-bound oligonucleotide may also be reacted with a suitable lipid aldehyde, followed by reduction with sodium cyanoborohydride, which produces an amine linkage.
  • the oligonucleotide can be synthesized using a modified, lipid-containing solid support. Reaction of 3-amino-1 ,2-propanediol with a fatty acyl chloride (RC(O)CI), followed by dimethoxytritylation of the primary alcohol and succinylation of the secondary alcohol, provides an intermediate which is then coupled, via the free succinyl carboxyl group, to the solid support.
  • RC(O)CI fatty acyl chloride
  • R represents a lipid.
  • GRN163L The structure above, when -R is -(CH 2 )i 3 CH3 (palmitoyl), is designated herein as GRN163L.
  • the assay detects and/or measures telomerase activity by measuring the addition of TTAGGG telomeric repeats to a biotinylated telomerase substrate primer.
  • the biotinylated products are captured on streptavidin-coated microtiter plates, and an oligonucleotide probe complementary to 3.5 telomere repeats, labeled with 33P, is used for measuring telomerase products. Unbound probe is removed by washing, and the amount of probe annealing to the captured telomerase products is determined by scintillation counting.
  • telomere activity in a cell can be determined using the TRAP (Telomeric Repeat Amplification Protocol) assay, which is described, for example, in Kim et a/., U.S. Patent No. 5,629,154; Harley et al., U.S. Patent No. 5,891 ,639; and Harley et al., PCT Pubn. No. WO 2005/000245. Briefly, telomerase-expressing tumor cell lines are incubated with test compositions, lysed, and treated with a labeled oligonucleotide telomerase substrate, appropriate primers, and internal standard for quantitation purposes.
  • TRAP Telomeric Repeat Amplification Protocol
  • telomere repeats will be added to the substrate, to form telomerase extended products.
  • the mixture is incubated at room temperature, followed by multiple cycles of PCR.
  • the mixture is separated on a gel, and labeled extension product is detected and quantitated via comparison with the internal standard.
  • SEQ ID NO: 1 the RNA component of human telomerase (hTR): GGGUUGCGGA GGGUGGGCCU GGGAGGGGUG GUGGCCAUUU UUUGUCUAAC CCUAACUGAG 60
  • GUGCGCAUCC GUCACCCCUC GCCGGCAGUG GGGGCUUGUG AACCCCCAAA CCUGACUGAC 540 UGGGCCAGUG UGCU
  • SEQ ID NOS: 2-26 the nucleotide sequences of targeting agents against SEQ ID NO: 1 :

Abstract

A method and kit for inhibiting the proliferation of cancer cells are disclosed, based on a combination of an HSP90 inhibitor and a telomerase inhibitor. When used in cancer therapy, the two compounds in combination enhance the anti-cancer treatment efficacy obtained with the HSP90 inhibitor alone or the telomerase inhibitor alone. Preferably, efficacy is supraadditive or synergistic in nature relative to the combined effects of the individual agents, with minimal exacerbation of side effects.

Description

CANCER TREATMENT BY COMBINED INHIBITION OF TELOMERASE AND HSP90 ACTIVITIES
Field of the Invention
The invention is directed to anticancer treatment, and in particular to inhibition of tumor growth or cancer-cell proliferation, by treatment with a telomerase inhibitor in combination with an HSP90 inhibitor.
Background
Although many cancers can be cured by surgical resection, chemotherapy is often used as an adjunct to surgical therapy, and it is widely used in the treatment of inoperable or metastatic malignancy. In view of the continuing high number of deaths each year resulting from cancer, a continuing need exists to identify effective and relatively nontoxic therapeutic regimens for use in anticancer treatment.
Many effective chemotherapeutic agents have been identified over the past few decades, and these are generally grouped into several categories on the basis of their mechanism of action. Combined-therapy treatments have become more common, in view of the perceived advantage of attacking the disease via multiple avenues. In practice, however, many such combinations do not provide even simple additivity of therapeutic effects.
Ideally, a combined-drug approach for cancer treatment should provide a significant boost in efficacy and/or a significant reduction in undesired side effects, due to a reduced dose of the more toxic component and/or a reduction in the development of drug-resistance in the cancer being treated. Particularly desirable are combination therapies which produce therapeutic results that are supraadditive or synergistic in nature relative to the effects of the individual agents, with minimal exacerbation of side effects.
Summary
The invention includes a method for inhibiting the proliferation of cancer cells, by (a) exposing the cells to an HSP90 inhibitor, and (b) either proceeding, following, or concomitantly with step (a), exposing the cells to a telomerase inhibitor. In one embodiment, the amount of HSP90 inhibitor to which the cells are exposed is effective, by itself, to inhibit proliferation of the cancer cells. In a further embodiment, the amount of each inhibitor is effective, by itself, to inhibit proliferation of the cancer cells. Preferably, the combination provides a enhanced inhibiting effect relative to either component alone; more preferably, the combination provides a supraadditive or synergistic effect relative to the combined or additive effects of the components.
The telomerase inhibitor may include an oligonucleotide having nuclease- resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR. The intern u cleoside linkages in the oligonucleotide may be selected from N3'->P5' phosphoramidate and N3'->P5' thiophosphoramidate linkages. The telomerase inhibitor may include a lipid moiety, such as a fatty acid, sterol, or derivative thereof, which is attached covalently at one end of the oligonucleotide. The oligonucleotide may be 10-20 bases in length, preferably 13-20 bases in length, and may have the sequence identified by SEQ ID NO:12 (δ'-TAGGGTTAGACAA-S1). One exemplary telomerase inhibitor is the compound designated herein as GRN163L.
The HSP90 inhibitor may include a benzoquinone ansamycin antibiotic, such as galdanamycin or an analogue thereof, as exemplified by 17-AAG (17-N- allylamino-17-demethoxygaldanamycin), or the closely related compounds herbimycin A, B, or C, and radicicol, or a pyrrolopyrimidine or pyrazolopyrimidine HSP90 inhibitor, such as disclosed in U.S. Patent Publications 2005/0107343A1 , and 2005/0119282 A1 , respectively.
The method may be used for use in treating a subject having a cancer, where, preferably, exposing step (a) includes administering the HSP90 inhibitor to the subject in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject. In a further embodiment, each exposing step (a) and (b) includes administering the respective inhibitor to the subject in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
The method may be used in treating a variety of cancers in a subject. In exemplary embodiments, the method is used in treating a subject diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain or myelomas.
Where the telomerase inhibitor is the compound GRN163L, it may be administered to the subject by intravenous infusion, under infusion conditions effective to produce a blood concentration of the inhibitor of between 1 nM and 10O uM.
In another aspect, the invention is directed to a method for enhancing the anti-cancer treatment efficacy of an HSP90 inhibitor in a subject. The method includes administering to the subject, before, during, or after administering the HSP90 inhibitor, an oligonucleotide telomerase inhibitor of the type composed of an oligonucleotide having nuclease-resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR. In one embodiment, the amount of the telomerase inhibitor is effective to inhibit the proliferation of cancer cells in the subject when the telomerase inhibitor is administered alone.
The enhancement of treatment efficacy may be evidenced, for example, by an increased survival time of the subject, or by an inhibition of tumor growth in the subject, relative to treatment with the HSP90 inhibitor alone.
The method may be used to treat a subject diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non- small-cell lung cancer, head, neck, and gastrointestinal cancers, prostate and testicular cancers, and melanomas.
The oligonucleotide may be 10-20 bases in length. Preferably, the oligonucleotide is 13-20 bases in length and includes the sequence identified by SEQ ID NO: 12 (δ'-TAGGGTTAGACAA-S1). An exemplary telomerase inhibitor is the compound identified as GRN163L, or an analog thereof. This compound has (i) N3'->P5' thiophosphoramidate intemucleoside linkages in the oligonucleotide; (ii) the sequence identified as SEQ ID NO:12; and (iii) a palmitoyl (C16) moiety linked to the 5' end of the oligonucleotide through a glycerol or aminoglycerol linker. One preferred HSP90 inhibitor is galdanamycin or an analog thereof, such as 17-AAG. The HSP90 and telomerase inhibitors may be administered as a composition containing both inhibitors.
The invention may also be practiced to identify cancer patients who are candidates for effective anti-cancer treatment with a telomerase inhibitor. The candidate patients are those whose cancer is responding to treatment with an HSP90 inhibitor, such as 17-AAG, but for whom combined treatment with a telomerase inhibitor is desired to enhance the anti-tumor efficacy of the HSP90 inhibitor alone.
Also disclosed in a kit for use in cancer therapy, comprising (a) a dose of an HSP90 inhibitor, in an amount of the inhibitor effective, when administered alone, to inhibit the proliferation of cancer cells in the subject, and (b) a dose of an oligonucleotide telomerase inhibitor having nuclease-resistant intersubunit linkages, and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR. In one embodiment, the telomerase inhibitor is also provided in an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
In an exemplary embodiment, HSP90 inhibitor is 17-AAG, and the telomerase inhibitor is the compound identified as GRN163L, or an analog thereof. The latter compound has (i) N3'->P5' thiophosphoramidate internucleoside linkages in the oligonucleotide;(ii) the sequence identified as SEQ ID NO: 12; and (iii) a palmitoyl (C16) moiety linked to the 5' end of the oligonucleotide through a glycerol or aminoglycerol linker.
Also provided is a kit comprising a telomerase inhibitor and an HSP90 inhibitor, such as galdanamycin or an analog thereof, for use in therapy, such as the treatment of cancer. Such therapy preferably comprises administering the HSP90 inhibitor to a subject, either preceding, following, or concomitantly with administration of said telomerase inhibitor. The telomerase inhibitor is preferably a nuclease-resistant oligonucleotide which binds in a sequence-specific manner to the template region of hTR. Preferred and/or exemplary inhibitors and cancer indications are as set out above.
In a related aspect, the invention provides a kit containing a dose of an oligonucleotide telomerase inhibitor having nuclease-resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR, preferably in an amount effective to inhibit proliferation of cancer cells in a subject. The kit may optionally include an insert that provides a user with one set of instructions for using the inhibitor in monotherapy and a separate set of instructions for using the inhibitor in combination with 17-AAG or other HSP90 inhibitor. The set of instructions for the combination therapy may recommend (i) a lower dose of the telomerase inhibitor, when used in combination with the HSP90 inhibitor, (ii) a lower dose of the HSP90 inhibitor, when used in combination with the HSP90 inhibitor, and/or (iii) a different dosing regimen for one or both inhibitors, when used together, than would normally be recommended for the inhibitor(s) when used alone.
Also provided is the use of a telomerase inhibitor for preparation of a medicament for use in treatment of cancer, wherein said treatment comprises administering said telomerase inhibitor to a subject in combination with an HSP90 inhibitor, such as galdanamycin or an analogue thereof. Said treatment may comprise administering the HSP90 inhibitor to the subject either preceding, following, or concomitantly with the telomerase inhibitor, which is preferably a nuclease-resistant oligonucleotide which binds in a sequence-specific manner to the template region of hTR.
In a related aspect, the invention provides the use of an HSP90 inhibitor and a telomerase inhibitor in the manufacture of a medicament for treating cancer in a subject. Preferred and/or exemplary inhibitors and cancer indications are as set out above.
Further disclosed is the use of a telomerase inhibitor, in the manufacture of a medicament for treating cancer in a subject who is being treated with an HSP90 inhibitor, for the purpose of enhancing the anti-cancer efficacy of an HSP90 inhibitor in the subject. Preferred and/or exemplary inhibitors and cancer indications are as set out above.
These and other objects and features of the invention will become more fully apparent when the following detailed description of the invention is read in conjunction with the accompanying drawings.
Brief Description of the Drawings
Figs. 1 -2 show enhancement of telomerase inhibiting activity of an NPS oligonucleotide hTR template inhibitor (GRN163) by conjugation to a lipid (to produce GRN163L), in human myeloma tumor xenografts (Fig. 1) and liver tissue (Fig. 2), respectively, in mice; and
Fig. 3 illustrates the enhanced effect on inhibition of cancer cells achieved by co-administration of an HSP90 inhibitor, exemplified by 17-AAG and the telomerase inhibitor GRN163L, in INA6 myeloma cells.
Detailed Description of the Invention I. Definitions
The terms below have the following meanings unless indicated otherwise.
A "polynucleotide" or "oligonucleotide" refers to a ribose and/or deoxyribose nucleoside subunit polymer or oligomer having between about 2 and about 200 contiguous subunits. The nucleoside subunits can be joined by a variety of intersubunit linkages, including, but not limited to, phosphodiester, phosphodiester, methylphosphonate, P3'->N5' phosphoramidate, N3'->P5' phosphoramidate, N3'->'P5' thiophosphoramidate, and phosphorothioate linkages. The term also includes such polymers or oligomers having modifications, known to one skilled in the art, to the sugar (e.g., 2' substitutions), the base (see the definition of "nucleoside" below), and the 3' and 5' termini. In embodiments where the oligonucleotide moiety includes a plurality of intersubunit linkages, each linkage may be formed using the same chemistry, or a mixture of linkage chemistries may be used. When an oligonucleotide is represented by a sequence of letters, such as "ATGUCCTG," it will be understood that the nucleotides are in 5'->3' order from left to right. Representation of the base sequence of the oligonucleotide in this manner does not imply the use of any particular type of internucleoside subunit in the oligonucleotide.
The term "nucleoside" includes the natural nucleosides, including 2'-deoxy and 2'-hydroxyl forms, e.g., as described in Komberg and Baker, DNA Replication, 2nd Ed. (Freeman, San Francisco, 1992), and analogs. "Analogs", in reference to nucleosides, includes synthetic nucleosides having modified nucleobase moieties (see definition of "nucleobase" below) and/or modified sugar moieties, e.g., described generally by Scheit, Nucleotide Analogs (John Wiley, New York, 1980). Such analogs include synthetic nucleosides designed to enhance binding properties, e.g., stability, specificity, or the like, such as disclosed by Uhlmann and Peyman (Chemical Reviews 90:543-584, 1990). An oligonucleotide containing such nucleosides, and which typically contains synthetic nuclease-resistant internucleoside linkages, may itself be referred to as an "analog".
A "nucleobase" includes (i) native DNA and RNA nucleobases (uracil, thymine, adenine, guanine, and cytosine), (ii) modified nucleobases or nucleobase analogs (e.g., 5-methylcytosine, 5-bromouracil, or inosine) and (iii) nucleobase analogs. A nucleobase analog is a compound whose molecular structure mimics that of a typical DNA or RNA base.
The term "lipid" is used broadly herein to encompass substances that are soluble in organic solvents, but sparingly soluble, if at all, in water. The term lipid includes, but is not limited to, hydrocarbons, oils, fats (such as fatty acids and glycerides), sterols, steroids and derivative forms of these compounds. Preferred lipids are fatty acids and their derivatives, hydrocarbons and their derivatives, and sterols, such as cholesterol.
Fatty acids usually contain even numbers of carbon atoms in a straight chain (commonly 12-24 carbons) and may be saturated or unsaturated, and can contain, or be modified to contain, a variety of substituent groups. For simplicity, the term "fatty acid" also encompasses fatty acid derivatives, such as fatty or esters.
The term "hydrocarbon" encompasses compounds that consist only of hydrogen and carbon, joined by covalent bonds. The term encompasses open chain (aliphatic) hydrocarbons, including straight chain and branched hydrocarbons, and saturated as well as mono-and poly-unsaturated hydrocarbons. The term also encompasses hydrocarbons containing one or more aromatic rings.
As used herein, the term "lipid" also includes amphipathic compounds containing both lipid and hydrophilic moieties.
The term "substituted" refers to a compound which has been modified by the exchange of one atom or moiety for another, typically substitution of hydrogen by a different atom or moiety. In particular, the term is used in reference to halogenated hydrocarbons and fatty acids, particularly those in which one or more hydrogen atoms are substituted with fluorine.
HSP90 refers to one of three distinct members of the HSP90 molecular chaperone family in mammalian cells. The cytosolic heat shock protein HSP90 has two isoforms, HSP90A and HSP90B, which are 76% identical and are thought to be the consequence of a gene duplication event. A third family member, the 94kDa glucose-regulated protein (GRP94), is localized primarily to the endoplasmic reticulum and shows 50% homology with HSP90.
A "HSP90 inhibitor" is a compound capable of interacting with HSP90 protein within a cell, to inhibit or block the activity of the protein. Typically, an HSP90 inhibitor acts by binding to the ATP/ADP binding pocket of HSP90 with a relatively high affinity, e.g., higher than the affinity for the adenine nucleotides. One class of HSP90 inhibitors include the benzoquinone ansamycin antibiotics, such as galdanamycin, galdanamycin analogs, such as 17-AAG, and herbicycin A, B, and C.
An "hTR template inhibitor" is a compound that blocks the template region (the region spanning nucleotides 30-67 of SEQ ID NO: 1 herein) of the RNA component of human telomerase, thereby inhibiting the activity of the enzyme. The inhibitor is typically an oligonucleotide that is able to hybridize to this region. Preferably, the oligonucleotide includes a sequence effective to hybridize to a more specific portion of this region, having sequence 5'-CUAACCCUAAC-3' (SEQ ID NO: 12), spanning nucleotides 46-56 of SEQ ID NO: 1 herein.
A compound is said to "inhibit the proliferation of cancer cells" if the proliferation of cells in the presence of the compound is less than that observed in the absence of the compound. That is, proliferation of the cells is either slowed or halted in the presence of the compound. Inhibition of cancer-cell proliferation may be evidenced, for example, by reduction in the number of cells or rate of expansion of cells, reduction in tumor mass or the rate of tumor growth, or increase in survival rate of a subject being treated.
Administration of a telomerase inhibitor to a subject is effective to "enhance the anti-cancer treatment efficacy of an HSP90 inhibitor" if the subject shows a reduced rate of tumor growth and/or an enhanced survival rate with combined therapy over therapy with the HSP90 inhibitor alone.
An oligonucleotide having "nuclease-resistant linkages" refers to one whose backbone has subunit linkages that are substantially resistant to nuclease cleavage, in non-hybridized or hybridized form, by common extracellular and intracellular nucleases in the body; that is, the oligonucleotide shows little or no nuclease cleavage under normal nuclease conditions in the body to which the oligonucleotide is exposed. The N3'->P5' phosphoramidate (NP) or N3'->P5' thiophosphoramidate (NPS) linkages described below are nuclease resistant.
II. Treatment of cancer with an HSP90 inhibitor
HSP90, referring to any and all of the three distinct members of the HSP90 molecular chaperone family in mammalian cells, HSP90A and HSP90B and GRP94, is a highly abundant and essential cytosolic protein and the expression levels of HSP90 increases by environmental stress. HSP90 functions as a molecular chaperone by binding to various cellular proteins and supporting the proper folding, stability, and function of target proteins. The HSP90 client proteins include a wide variety of signal-transducing proteins that regulate cell growth and differentiation, such as protein kinases and steroid hormone receptors. HSP90 functions in an ATP-dependent manner in cooperation with other molecular chaperones such as Cdc37 and FKBP52. HSP90 inhibitors act by inhibiting the essential ATPase activity of HSP90. Thus, treatment of cells with an HSP90 inhibitor, such as geldanamycin, results in inactivation, destabilization, and degradation of HSP90 client proteins. Because HSP90 client proteins play important roles in the regulation of the cell cycle, cell growth, cell survival, apoptosis, and oncogenesis, an HSP90 inhibitor obstructs the proliferation of cancer cells and shows anti-cancer activity in experimental animals.
A number of clinical trials are underway evaluating HSP90 inhibitor such as 17-AAG, both alone and in combination with other drugs, for a variety of cancers. The compound is aimed at treating subjects diagnosed with breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain and myelomas, among other cancers.
Exemplary HSP90 inhibitors include, but are not limited to, benzoquinone ansamycin antibiotics, such as geldanamycin, and its analogs, including 17-AAG (17-N-allylamino-17-demethoxygaldanamycin), 17-(2-dimethylaminoethyl)amino- 17-desmethoxy-geldanamycin (" 17-DMAG"), 11-0-methyl-17-(2-(1- azetidinyl)ethyl)amino-17-demethoxygeldanamycin, 11 -0-methyl-17-(2- dimethylaminoethyl)amino-17-demethoxygeldanamycin, and 11-0-methyl-17-(2- (1-pyrrolidinyl)ethyl)amino-17-demethoxygeldanamycin, and other galdanamysin analogs disclosed in one or more of U.S. Patent Nos. 4,261 ,989, 5,932,566, 6,890,917, 6,855,705, and 6,872,715, all of which are all incorporated herein by reference.
Other HSP90 inhibitors include pyrrolopyrimidine HSP90 inhibitors, as described in U.S. Patent Publication 2005/0107343 A1 , and pyrazolopyrimidine HSP90 inhibitors, as described in U.S. Patent Publication 2005/0119282 A1 , both of which are incorporated herein by reference.
In treating a subject with cancer, the HSP90 inhibitor is administered at a dose effective to produce a serum concentration between 0.01 μM and 10 μM, typically between 0.025 μM and 2 μM, e.g., at a concentration between 0.04 μM and 1 μM. This concentration may be achieved at a dose between about 0.01 mg/kg and 20 mg/kg, typically between 0.1 mg/kg and 2 mg/kg, e.g., 0.5 mg/kg and 2 mg/kg. For example, about 1 mg/kg of 17-AAG is administered to a patient with a cancer every day for five days of each week of a treatment cycle, or an equivalent dose administered less often, e.g., once per week, over a treatment period with several cycles, e.g., five cycles. Thus, about 5 mg/kg of 17- AAG could be administered once a week for each week of a treatment cycle, where the patient is treated 4 weeks per treatment cycle over a 5-cycle treatment regimen. One advantage of the present invention is that the dose of the compound may be reduced when administered in combination with a telomerase inhibitor.
III. Treatment of cancer with a telomerase inhibitor
Telomerase is a ribonucleoprotein that catalyzes the addition of telomeric repeat sequences (having the sequence 5'-TTAGGG-3' in humans) to chromosome ends. See e.g. Blackburn, 1992, Ann. Rev. Biochem. §1:113-129. The enzyme is expressed in most cancer cells but not in mature somatic cells. Loss of telomeric DNA may play a role in triggering cellular senescence; see Harley, 1991 , Mutation Research 256:271 -282. A variety of cancer cells have been shown to be telomerase-positive, including cells from cancer of the skin, connective tissue, adipose, breast, lung, stomach, pancreas, ovary, cervix, uterus, kidney, bladder, colon, prostate, central nervous system (CNS), retina and hematologic tumors (such as myeloma, leukemia and lymphoma). Targeting of telomerase can be effective in providing treatments that discriminate between malignant and normal cells to a high degree, avoiding many of the deleterious side effects that can accompany chemotherapeutic regimens which target dividing cells indiscriminately.
Inhibitors of telomerase identified to date include oligonucleotides, preferably oligonucleotides having nuclease resistant linkages, as well as small molecule compounds.
A. Small Molecule Compounds
Small molecule inhibitors of telomerase include, for example, BRACO19 ((9-(4-(N,N-dimethylamino)phenylamino)-3,6-bis(3-pyrrolodino propionamido)acridine (see MoI. Pharmacol. 61_(5):1154-62, 2002); DODC (diethyloxadicarbocyanine), and telomestatin. These compounds may act as G- quad stabilizers, which promote the formation of an inactive G-quad configuration in the RNA component of telomerase. Other small molecule inhibitors of telomerase include BIBR1532 (2-[(E)-3-naphthen-2-yl but-2- enoylaminojbenzoic acid) (see Ward & Autexier, MoI. Pharmacol. 68:779-786, 2005; also J. Biol. Chem. 277(18): 15566-72, 2002); AZT and other nucleoside analogs, such as ddG and ara-G (see, for example, U.S. Patent Nos. 5,695,932 and 6,368,789), and certain thiopyridine, benzo[b]thiophene, and pyrido[b]thiophene derivatives, described by Gaeta et al. in U.S. Patent Nos. 5,767,278, 5,770,613, 5,863,936, 5,656,638 and 5,760,062. One example is 3- chlorobenzo[b]thiophene-2-carboxy-2'-[(2,5-dichlorophenyl amino)thia]hydrazine, described in U.S. Patent No. 5,760,062. B. Oliqonucleotide-Basecl Telomerase Inhibitors: Sequence and Composition
The genes encoding both the protein and RNA components of human telomerase have been cloned and sequenced (see U.S. Pat. Nos. 6,261 ,836 and 5,583,016, respectively, both of which are incorporated herein by reference). Oligonucleotides can be targeted against the mRNA encoding the telomerase protein component (the human form of which is known as human telomerase reverse transcriptase, or hTERT) or the RNA component of the telomerase holoenzyme (the human form of which is known as human telomerase RNA, or hTR).
The nucleotide sequence of the RNA component of human telomerase (hTR) is shown in the Sequence Listing below (SEQ ID NO: 1 ), in the 5'-»3' direction. The sequence is shown using the standard abbreviations for ribonucleotides; those of skill in the art will recognize that the sequence also represents the sequence of the cDNA, in which the ribonucleotides are replaced by deoxyribonucleotides, with uridine (U) being replaced by thymidine (T). The template sequence of the RNA component is located within the region defined by nucleotides 46-56 (δ'-CUAACCCUAAC-S1), which is complementary to a telomeric sequence composed of about one-and-two-thirds telomeric repeat units. The template region functions to specify the sequence of the telomeric repeats that telomerase adds to the chromosome ends and is essential to the activity of the telomerase enzyme (see e.g. Chen et al., Ce// 100:503-514, 2000; Kim et al., Proc. Natl. Acad. Sci. USA 98(14):7982-7987, 2001 ). The design of aπtisense, ribozyme or small interfering RNA (siRNA) agents to inhibit or cause the destruction of mRNAs is well known (see, for example, Lebedeva et al. Annual Review of Pharmacology and Toxicology, Vol. 41.: 403-419, April 2001 ; Macejak, D. et al., Journal of Virology, Vol. 73 (9): p. 7745-7751 , September 1999, and Zeng, Y. et al., PNAS Vol. 100(17) pp. 9779-9784, August 19, 2003), and such agents may be designed to target the hTERT mRNA and thereby inhibit production of hTERT protein in a target cell, such as a cancer cell (see, for example, U.S. Pat. Nos. 6,444,650 and 6,331 ,399).
Oligonucleotides targeting hTR (that is, the RNA component of the enzyme) act as inhibitors of telomerase enzyme activity by blocking or otherwise interfering with the interaction of hTR with the hTERT protein, which interaction is necessary for telomerase function. See, for example, Villeponteau et al., U.S. Patent No. 6,548,298.
A preferred target region of hTR is the template region, spanning nucleotides 30-67 of SEQ ID NO:1. Oligonucleotides targeting this region are referred to herein as "hTR template inhibitors" (see e.g. Herbert et al., Oncogene 21(4):638-42 (2002).) Preferably, such an oligonucleotide includes a sequence which is complementary or near-complementary to some portion of the 11- nucleotide region having sequence 5'-CUAACCCUAAC-S1, spanning nucleotides 46-56 Of SEQ ID NO: 12.
Another preferred target region is the region spanning nucleotides 137- 179 of hTR (see Pruzan et al., Nucl. Acids Research, 30:559-568, 2002). Within this region, the sequence spanning 141-153 is a preferred target. PCT publication WO 98/28442 describes the use of oligonucleotides of at least 7 nucleotides in length to inhibit telomerase, where the oligonucleotides are designed to be complementary to accessible portions of the hTR sequence outside of the template region, including nucleotides 137-196, 290-319, and 350- 380 of hTR. Preferred hTR targeting sequence are given below, and identified by SEQ ID NOS: 2-22.
The region of the therapeutic oligonucleotide that is targeted to the hTR sequence is preferably exactly complementary to the corresponding hTR sequence. While mismatches may be tolerated in certain instances, they are expected to decrease the specificity and activity of the resultant oligonucleotide conjugate. In particular embodiments, the base sequence of the oligonucleotide is thus selected to include a sequence of at least 5 nucleotides exactly complementary to the hTR target, and enhanced telomerase inhibition may be obtained if increasing lengths of complementary sequence are employed, such as at least 8, at least 10, at least 12, at least 13 or at least 15 nucleotides exactly complementary to the hTR target. In other embodiments, the sequence of the oligonucleotide includes a sequence of from at least 5 to 20, from at least 8 to 20, from at least 10 to 20 or from at least 10 to 15 nucleotides exactly complementary to the hTR target sequence.
Optimal telomerase inhibitory activity may be obtained when the full length of the oligonucleotide is selected to be complementary to the hTR target sequence. However, it is not necessary that the full length of the oligonucleotide is exactly complementary to the target sequence, and the oligonucleotide sequence may include regions that are not complementary to the target sequence. Such regions may be added, for example, to confer other properties on the compound, such as sequences that facilitate purification. Alternatively, an oligonucleotide may include multiple repeats of a sequence complementary to an hTR target sequence.
If the oligonucleotide is to include regions that are not complementary to the target sequence, such regions are typically positioned at one or both of the 5' or 31 termini. Exemplary sequences targeting human telomerase RNA (hTR) include the following:
Figure imgf000015_0001
The intemucleoside linkages in the oligonucleotide may include any of the available oligonucleotide chemistries, e.g. phosphodiester, phosphotri ester, methylphosphonate, P3'->N5' phosphoramidate, N3'->P5' phosphoramidate, N3'->P5' thiophosphoramidate, and phosphorothioate. Typically, but not necessarily, all of the internucleoside linkages within the oligonucleotide will be of the same type, although the oligonucleotide component may be synthesized using a mixture of different linkages.
In preferred embodiments, the oligonucleotide has at least one N3'->P5' phosphoramidate (NP) or N3'->P5' thiophosphoramidate (NPS) linkage, which linkage may be represented by the structure: 3'-(-NH-P(=O)(-XR)-O-)-5', wherein c X is O or S and R is selected from the group consisting of hydrogen, alkyl, and aryl; and pharmaceutically acceptable salts thereof, when XR is OH or SH. More preferably, the oligonucleotide includes all NP or, most preferably, all NPS linkages.
A particularly preferred sequence for an hTR template inhibitor oligonucleotide is the sequence complementary to nucleotides 42-54 of SEQ ID NO: 12 above. The oligonucleotide having this sequence (TAGGGTTAGACA) and N3'->P5' thiophosphoramidate (NPS) linkages is designated herein as GRN163. See, for example, Asai et al., Cancer Research 63:3931-3939 (2003); Gryaznov et al., Nucleosides Nucleotides Nucleic Acids 22(5-8):577-81 (2003).
As shown in Table 1 below, this oligonucleotide (first row of table) inhibits telomerase at low concentrations in a biochemical assay (FlashPlate™; see Experimental Section). An alternative 13-mer, having the sequence CAGTTAGGGTTAG, complementary to nucleotides 46-58 of SEQ ID NO: 1 (fifth row of table), showed near-equivalent activity in the FlashPlate™ assay. The corresponding NP-linked oligonucleotide, and shorter (11- and 12-mer) oligonucleotides targeting the same region (complementary to nucleotides 42-53 and 42-42, respectively, of SEQ ID NO: 1), showed moderate activity. The effect is clearly sequence-specific, as shown by the mismatch and non-targeting sequences in the table.
The oligonucleotide GRN163 administered alone has shown inhibitory activity in vitro in cell culture, including epidermoid carcinoma, breast epithelium, renal carcinoma, renal adenocarcinoma, pancreatic, brain, colon, prostate, leukemia, lymphoma, myeloma, epidermal, cervical, ovarian and liver cancer cells.
The oligonucleotide GRN163 has also been tested and shown to be therapeutically effective in a variety of animal tumor models, including ovarian and lung, both small cell and non-small cell.
Table 1. Inhibition of Telomerase by NPS Oligonucleotides: Biochemical FlashPlate Assa
Figure imgf000017_0001
C. Lipid-Oligonucleotide Conjugates
Preferably, the oligonucleotide-based enzyme inhibitor includes at least one covalently linked lipid group (see US Pubn. No. 2005/0113325, which is incorporated herein by reference). This modification provides superior cellular uptake properties, such that an equivalent biological effect may be obtained using smaller amounts of the conjugated oligonucleotide compared to the unmodified form. When applied to the human therapeutic setting, this may translate to reduced toxicity risks, and cost savings.
The lipid group L is typically an aliphatic hydrocarbon or fatty acid, including derivatives of hydrocarbons and fatty acids, with examples being saturated straight chain compounds having 14-20 carbons, such as myristic (tetradecanoic) acid, palmitic (hexadecanoic) acid, and stearic (octadeacanoic) acid, and their corresponding aliphatic hydrocarbon forms, tetradecane, hexadecane and octadecane. Examples of other suitable lipid groups that may be employed are sterols, such as cholesterol, and substituted fatty acids and hydrocarbons, particularly polyfluorinated forms of these groups. The scope of the lipid group L includes derivatives such as amine, amide, ester and carbamate derivatives. The type of derivative is often determined by the mode of linkage to the oligonucleotide, as exemplified below.
In one exemplary structure, the lipid moiety is palmitoyl amide (derived from palmitic acid), conjugated through an aminoglycerol linker to the 5' thiophosphate group of an NPS-linked oligonucleotide. The NPS oligonucleotide having the sequence shown for GRN 163 and conjugated in this manner (as shown below) is designated GRN163L herein. In a second exemplary structure, the lipid, as a palmitoyl amide, is conjugated through the terminal 3' amino group of an NPS oligonucleotide.
Figure imgf000018_0001
-(CH2)I3CH3 (palmitoyl)
GRN163L
As shown in Table 2, conjugation of a single fatty acid-type lipid significantly increased telomerase inhibitory activity in cell systems relative to the unconjugated oligonucleotide. Table 2. Inhibition of Telomerase by Lipid-Coniugated NPS Oligonucleotides (based on GRN163)
Figure imgf000019_0001
The effect of lipid conjugation on pharmacokinetics is illustrated by the data shown in Table 3, below, for a 4 mg/kg dose administered in rats. Target organ concentrations 6 hours after administration were also more favorable for GRN163L, with approx. 4-5 μM found in liver, kidney, and fat tissue, 2-3 μM in bone marrow and spleen, and about 0.5 μM in lymph node. Distribution of the urilipidated oligonucleotide, GRN163, was primarily to the kidney (about 18 μM), with only 1 μM or less in the remaining organ tissues noted above.
Table 4 presents further data directed to telomerase inhibition in vitro by GRN163 (unconjugated) and GRN163L (lipidated) in various cancer cell lines.
Table 3.
Comparative Pharmacokinetics of Lipidated (GRN163L) and Unlipidated (GRN 163) NPS Oligonucleotide (Rat, 4 mg/kg dose)
Figure imgf000019_0002
Table 4.
Comparative Telomerase Inhibitory Activity of ϋpidated (GRN163L) and Unlipidated (GRN163) NPS Oligonucleotide in vitro
Figure imgf000020_0001
The conjugated oligonucleotide GRN163L had significantly greater telomerase inhibiting activity in vivo than the unconjugated GRN163, as demonstrated in hepatoma cell xenografts (Fig. 1 ) and flank CAG myeloma tumor xenografts (Fig. 2) following i.v. administration.
Administration of GRN163L inhibited tumor growth in mice (A549-luc IV lung metastases model) for at least 4 weeks after i.v. injection of cancer cells. The dosage was 1 μM biweekly for 5 weeks prior to injection of cancer cells, followed by 5 mg/kg twice weekly after injection. Controls showed substantial tumor growth, but none was apparent in the GRN163L-treated mouse.
IV. Combination therapy with HSP90 and telomerase Inhibitors
Therapeutic benefits for treatment of cancer can be realized by combining a telomerase inhibitor with other anti-cancer agents. In one embodiment, the telomerase inhibitor is an hTR template blocking agent, as described above. Preferably, the combination of telomerase inhibition with chemotherapeutic treatment has a supraadditive effect; that is, the combined benefit is greater than would be expected simply from the additive effects of the two therapeutic approaches. This is demonstrated herein for the combination of a telomerase inhibitor and the HSP90 inhibitor 17-AAG.
In accordance with the present invention, it has been discovered that combined exposure of cancer cells to both an HSP90 inhibitor and a telomerase activity enhances the extent to which cell proliferation is inhibited relative to the HSP90 inhibitor alone, or the telomerase inhibitor alone. The effect is seen for inhibition of cancer cell growth in vitro, where the inhibition is evidenced by an increased rate of cell death in INA6 myeloma cells over a 8 day treatment period, as detailed below with respect to Fig. 3.
In practicing the method of the invention, a subject having a cancer type that is responsive to an HSP90 inhibitor, or a subject currently receiving cancer therapy with an HSP90 inhibitor, is initially identified as a candidate for the combined therapy. Preferred cancer indications include, for example, breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain and myelomas.
Thus, an aspect of the invention involves identifying cancer patients who are candidates for effective anti-cancer treatment with a telomerase inhibitor. The candidate patients are those whose cancer is responding to treatment with an HSP90 inhibitor, e.g., 17-AAG, but for whom combined treatment with a telomerase inhibitor is desired to enhance the anti-tumor efficacy of the HSP90 inhibitor alone.
The cancer should also be one that is responsive to cancer-cell inhibition by telomerase inhibition. As noted above, oligonucleotide telomerase inhibitors, as exemplified by GRN163 and GRN163L, have shown inhibitory activity in vitro against human kidney, lung, pancreatic, brain, colon, prostate, breast, leukemia, lymphoma, myeloma, epidermal, cervical, ovarian and liver cancer cells, and in vivo, via local and systemic delivery, against human brain, prostate, lymphoma, myeloma, cervical, lung, and liver cancer cells. Other preferred targets include small cell lung, esophogeal, head and neck, and stomach cancers.
In the preferred treatment method, the subject is administered the HSP90 inhibitor, e.g., 17-AAG, in an amount that is effective inhibiting proliferation of cancer cells in the subject. The dose administered and the dosing schedule will follow, for example, known or recommended doses for the HSP90 inhibitor employed, as indicated, for example, in the drug product insert or published clinical or animal-model data. In a typical treatment method for 17-AAG, a patient will receive 0.1 mg/kg to 2 mg/kg For example, about 1 mg/kg of 17-AAG is administered to a patient with a cancer every day for five days of each week of a treatment cycle, where the patient is treated for 5 cycles. In another example, about 10 mg/kg of 17-AAG is administered once a week for each week of a treatment cycle, where the patient is treated 4 weeks per treatment cycle over a 5-cycle treatment regimen.
One advantage of the present invention is that lower-than-normal doses of the HSP90 inhibitor may be administered, if necessary, due to the compensating enhancement effect of the telomerase inhibitor. Such a protocol allows for a reduced dosage of the HSP90 inhibitor, which can have significant toxic effects at higher dosages. Thus, a kit containing a dose of the telomerase inhibitor could optionally contain a product insert having one set of directions for using the inhibitor in monotherapy, and another set of directions for using the inhibitor in a combination therapy with an HSP90 inhibitor, such as a 17-AAG. The set of instructions for the combination therapy could recommend (i) a lower dose of the telomerase inhibitor, when used in combination with the HSP90 inhibitor, (ii) a lower dose of the HSP90 inhibitor, when used in combination with the telomerase inhibitor, and/or (iii) a different dosing regimen for one or both inhibitors, when used together, than would normally be recommended for the inhibitor(s) when used alone.
The telomerase inhibitor may be administered, before, during, or after administration of the HSP90 inhibitor. Typically, the two inhibitors are administered in a common dosing regimen, as described below, and the two inhibitors themselves may be administered in a combined-drug composition, e.g., by IV administration, or separately. However, a dosing regimen in which the telomerase inhibitor is administered before or after administering the HSP90 inhibitor is also contemplated. For example, a person under treatment with an HSP90 inhibitor may be subsequently placed on a combined therapy that includes telomerase inhibitor.
Alternatively, the patient may be initially administered the HSP90 inhibitor, followed one-to-several days later with the telomerase treatment. In this regimen, the HSP90 inhibitor may function, in part, to sensitize the cancer cells to inhibition by a telomerase inhibition, e.g., by synchronizing the cell-division cycle
!1 and/or promoting apoptosis in the cells. Preferred dose levels and dosing schedules are considered further below.
In one exemplary method, INA6 myeloma cells were treated with the HSP90 inhibitor 17-AAG in combination with a telomerase-inhibitor oligonucleotide targeted against hTR, with the results shown in Fig. 3. Briefly, INA6 myeloma cells were cultured for a seven-day period. At day seven, the cells were treated with one of (i) a mismatch oligonucleotide identical to GRN 163L except in its base sequence, at a concentration of 1 μM (solid diamonds), (ii) GRN163L, at final a concentration of 1 μM (solid triangles), (iii) 17-AGG, diamonds), at final a concentration of 0.05 μM (solid squares), and (iv) a combination of GRN 163L (1 μM) and 17-AAG 0.05 μM (X symbol). At intervals of 11 and 15 days (4 and 8 days post treatment, respectively), aliquots from each culture were removed, and the percent of dead cells was measured. As seen in the figure, the combined treatment was effective in killing more than 90% of the cells, compared with about 50% killing for each individual treatment over the same time period.
A. Administration
The therapeutic protocol for administering such combinations will depend on various factors including, but not limited to, the type of cancer, the age and general health of the patient, the aggressiveness of disease progression, the TRF length (terminal restriction fragment length; see Section V below) and telomerase activity of the diseased cells to be treated, and the ability of the patient to tolerate the agents that comprise the combination.
In general, treatment of all carcinoma and hematological malignancy types is contemplated. In selected embodiments, the target disease comprises a solid tumor; in other embodiments, the target disease comprises a hematological malignancy. An exemplary course of treatment involves multiple doses. Sequence of combination treatments will be determined by clinical compliance criteria and/or preclinical or clinical data supporting dose optimization strategies to augment efficacy or reduce toxicity of the combination treatment. In general, various combinations of the telomerase inhibitor and HSP90 inhibitor may be employed, used either sequentially or simultaneously. For multiple dosages, the two agents may directly alternate, or two or more closes of one agent may be alternated with a single dose of the other agent, for example. Simultaneous administration of both agents may also be alternated or otherwise interspersed with dosages of the individual agents. The time between dosages may be for a period from about 1-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1 -2 days, to about 1 -2 wk or longer following the initiation of treatment. During a course of treatment, the need to complete the planned dosings may be reevaluated.
The compounds may be administered by direct injection of a tumor or its vasculature. Alternatively, the tumor may be infused or perfused with the therapeutic compounds using any suitable delivery vehicle. The compounds may be administered locally to an affected organ. Systemic administration may also be performed. Continuous administration may be applied where appropriate; for example, where a tumor is excised and the tumor bed is treated to eliminate residual disease. Delivery via syringe or catheterization is preferred. Such continuous perfusion may take place for a period from about 1-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 weeks or longer following the initiation of treatment. Generally, the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs.
The therapeutic agents are administered to a subject, such as a human patient, in a formulation and in an amount effective to achieve a clinically desirable result. For the treatment of cancer, desirable results include reduction in tumor mass (as determined by palpation or imaging; e.g., by radiography, radionucleotide scan, CAT scan, or MRI), reduction in the rate of tumor growth, reduction in the rate of metastasis formation (as determined e.g., by histochemical analysis of biopsy specimens), reduction in biochemical markers (including general markers such as ESR, and tumor specific markers such as serum PSA), and improvement in quality of life (as determined by clinical assessment, e.g., Karnofsky score), increased time to progression, disease-free survival and overall survival. The amount of each agent per dose and the number of doses required to achieve such effects will vary depending on many factors including the disease indication, characteristics of the patient being treated and the mode of administration. Typically, the formulation and route of administration will provide a local concentration at the disease site of between 1 nM and 100 μM of each agent, preferably between 0.01 μM to 10 μM, as noted above. The physician will be able to vary the amount of the compounds, the carrier, the dosing frequency, and the like, taking into consideration such factors as the particular neoplastic disease state and its severity; the overall condition of the patient; the patient's age, sex, and weight; the mode of administration; the suitability of concurrently administering systemic anti-toxicity agents; monitoring of the patient's vital organ functions; and other factors typically monitored during cancer chemotherapy. In general, the compounds are administered at a concentration that affords effective results without causing excessive harmful or deleterious side effects.
In accordance with the invention, the amount of the agent used in combination with a telomerase inhibitor, may be less than would be required for the agent used in non-combination therapy.
B. Formulations
The pharmaceutical carrier(s) employed may be solid or liquid. Liquid carriers can be used in the preparation of solutions, emulsions, suspensions and pressurized compositions. The compounds are dissolved or suspended in a pharmaceutically acceptable liquid excipient. Suitable examples of liquid carriers for parenteral administration include water (which may contain additives, e.g., cellulose derivatives, preferably sodium carboxymethyl cellulose solution), phosphate buffered saline solution (PBS), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) and their derivatives, and oils (e.g., fractionated coconut oil and arachis oil). The liquid carrier can contain other suitable pharmaceutical additives including, but not limited to, the following: solubilizers, suspending agents, emulsifiers, buffers, thickening agents, colors, viscosity regulators, preservatives, stabilizers and osmolarity regulators.
For parenteral administration, the carrier can also be an oily ester such as ethyl oleate and isopropyl myristate. Sterile carriers are useful in sterile liquid form compositions for parenteral administration. Sterile liquid pharmaceutical compositions, solutions or suspensions can be utilized by, for example, intraperitoneal injection, subcutaneous injection, intravenously, or topically. The compositions can also be administered intravascularly or via a vascular stent.
The liquid carrier for pressurized compositions can be a halogenated hydrocarbon or other pharmaceutically acceptable propellant. Such pressurized compositions may also be lipid encapsulated for delivery via inhalation. For administration by intranasal or intrabronchial inhalation or insufflation, the compositions may be formulated into an aqueous or partially aqueous solution, which can then be utilized in the form of an aerosol.
The compositions may be administered topically as a solution, cream, or lotion, by formulation with pharmaceutically acceptable vehicles containing the active compound. The compositions of this invention may be orally administered in any acceptable dosage including, but not limited to, formulations in capsules, tablets, powders or granules, and as suspensions or solutions in water or nonaqueous media. Pharmaceutical compositions and/or formulations comprising the oligonucleotides of the present invention may include carriers, lubricants, diluents, thickeners, flavoring agents, emulsifiers, dispersing aids or binders. In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
Modes of administration and formulation may be dependent on the drug and its approved mode of administration. For example, when the chemotherapeutic agent is 17-AAG , IV administration is indicated. When the telomerase inhibitor is GRN163L, formulation in 0.9% sodium chloride (normal saline) and administration by i.v. is a preferred route, preferably via infusion over 4-8 hours, e.g. a 6 hr infusion.
While the lipid-conjugated oligonucleotides described herein, such as GRN163L, have superior characteristics for cellular and tissue penetration, these and other compounds may be formulated to provide further benefit in this area, e.g. in liposome carriers. The use of liposomes to facilitate cellular uptake is described, for example, in U.S. Pat. Nos. 4,897,355 and 4,394,448, and numerous publications describe the formulation and preparation of liposomes. Liposomal formulations can also be engineered, by attachment of targeting ligands to the liposomal surface, to target sites of neovascularization, such as tumor angiogenic regions. The compounds can also be formulated with additional penetration/transport enhancers, such as unconjugated forms of the lipid moieties described above, including fatty acids and their derivatives. Examples include oleic acid, lauric add, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, recinleate, . monoolein (a.k.a. 1 -monooleoyl-rac-glycerol), dilaurin, caprylic acid, arichidonic acid, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, acylcarnitines, acylcholines, mono- and di-glycerides and physiologically acceptable salts thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.). Other useful adjuvants include substrates for transendothelial migration, such as glucose uptake systems for facilitated egress from the vascular space to the tumor microenvironment.
V. Measurement of Telomere Length, Telomerase Activity, and/or Cell Proliferation
When employing a therapeutic regimen that involves administration of a telomerase inhibitor, it may be useful to determine telomere length and/or telomerase activity in a cell or tissue sample. These parameters can be measured by assays known in the art. Telomere length can be measured by a flow cytometry method using fluorescence in situ hybridization, referred to as flow FISH (see e.g. M. Hultdin et al., Nucleic Acids Res. 26(16):3651-6, 1998; N. Rufer et ai, Nature Biotechnology 16:743-7 , 1998). Other methods include terminal restriction fragment (TRF) analysis, in which genomic DNA is digested with a restriction enzyme having a four-base recognition sequence not present in telomere repeat sequences, and the restriction fragments are separated according to size, e.g. by gel electrophoresis. See, for example, U.S. Patent No. 5,489,508 (West et ai) and Harley et ai, Nature 345:458, 1990. The West et ai patent also describes methods of measuring telomere length by a "anchored terminal primer" method and by a modified Maxam-Gilbert reaction.
In addition, a more rapid response to a telomerase inhibiting agent may be predicted for tumor cells having shorter telomeric DNA, although telomerase has been shown to have other inhibitory effects independent of telomere length. (e.g. Stewart et ai, PNAS 99:12606, 2002; Zhu et al., PNAS 93:6091 , 1996; Rubaiyat et al., Oncogene 24(8):1320, 2005); and Folini et al., Curr. Pharm. Design Vl(Q)-A 105, 2005).
The TRAP assay (see Experimental, below) is a standard method for measuring telomerase activity in a cell extract system (Kim et al., Science 266:2011 , 1997; Weinrich et al., Nature Genetics 17:498, 1997). Briefly, this assay measures the amount of nucleotides incorporated into elongation products (polynucleotides) formed by nucleotide addition to a labeled telomerase substrate or primer. The TRAP assay is described in detail in U.S. Pat. Nos. 5,629,154, 5,837,453 and 5,863,726, and its use in testing the activity of telomerase inhibitory compounds is described in various publications, including WO 01/18015. In addition, the following kits are available commercially for research purposes for measuring telomerase activity: TRAPeze™ XK Telomerase Detection Kit (Intergen Co., Purchase N.Y.); and TeIoTAGGG Telomerase PCR ELISA plus (Roche Diagnostics, Indianapolis Ind.).
The anticancer activity of the therapeutic combinations can be evaluated using standard in vitro and in vivo assays. The ability of a composition to specifically inhibit the growth of tumor cells can be assayed using tumor cell lines in vitro, or in xenograft animal models in vivo. A preferred protocol for such growth curve assays is the short term cell viability assay described in Asai et al. (2003, cited above). In established xenograft models of human tumors, the test compound is administered either directly to the tumor site or systemically, and the growth of the tumor is followed by physical measurement. A preferred example of a suitable in vivo tumor xenograft assay is also described in Asai et al. (2003, cited above). Other examples are described in Scorski et al., Proc. Natl. Acad. ScL USA, 94: 3966-3971 (1997) and Damm ef al., EMBO J., 20:6958-6968 (2001 ).
Il Experimental
A. Preparation and Lipid Conjugation of Oligonucleotide N3'->P5' Phosphoramidates or N3'->P5' Thiophosphoramidates
These compounds may be prepared as described, for example, in McCurdy ef al., Tetrahedron Letters 38:207-210 (1997) or Pongracz & Gryaznov, Tetrahedron Letters 49:7661-7664 (1999). The starting 3'-amino nucleoside monomers may be prepared as described in Nelson et al., J. Org. Chem. 62:7278-7287 (1997) or by the methods described in Gryaznov et al., US Appn. Pubn. No. 2006/0009636.
A variety of synthetic approaches can be used to conjugate a lipid moiety L to the oligonucleotide, depending on the nature of the linkage selected; see, for example, Mishra ef al., Biochim. et Biophys. Acta 1264:229-237 (1995), Shea et al., Nucleic Acids Res. 18:3777-3783 (1995), or Rump et al., Bioconj. Chem. 9:341 -349 (1995). Typically, conjugation is achieved through the use of a suitable functional groups at an oligonucleotide terminus. For example, the 3'- amino group present at the 3'-terminus of the NP and NPS oligonucleotides can be reacted with carboxylic acids, acid chlorides, anhydrides and active esters, using suitable coupling catalysts, to form an amide linkage. Thiol groups are also suitable as functional groups (see Kupihar et al., Bioorg. Med. Chem. 9:1241-1247 (2001 )). Various amino- and thiol-functionalized modifiers of different chain lengths are commercially available for oligonucleotide synthesis.
Specific approaches for attaching lipid groups to a terminus of an NP or NPS oligonucleotide include those described in US Appn. Pubn. No. 2005/0113325, which is incorporated herein by reference. In addition to the amide linkages noted above, for example, lipids may also be attached to the oligonucleotide chain using a phosphoramidite derivative of the lipid, to produce a phosphoramidate or thiophosphoramidate linkage connecting the lipid and the oligonucleotide. The free 3'-amino group of the fully protected support-bound oligonucleotide may also be reacted with a suitable lipid aldehyde, followed by reduction with sodium cyanoborohydride, which produces an amine linkage.
For attachment of a lipid to the 5' terminus, as also described in US Appn. Pubn. No. 2005/0113325, the oligonucleotide can be synthesized using a modified, lipid-containing solid support. Reaction of 3-amino-1 ,2-propanediol with a fatty acyl chloride (RC(O)CI), followed by dimethoxytritylation of the primary alcohol and succinylation of the secondary alcohol, provides an intermediate which is then coupled, via the free succinyl carboxyl group, to the solid support. An example of a modified support is shown below, where S-- represents a long chain alkyl amine CPG support, and R represents a lipid.
Figure imgf000030_0001
This procedure is followed by synthesis of the oligonucleotide in the 5' to 3' direction, as described, for example, in Pongracz & Gryaznov (1999), starting with deprotection and phosphitylation of the -ODMT group. This is effective to produce, for example, the following structure, after cleavage from the solid support:
Figure imgf000030_0002
The structure above, when -R is -(CH2)i3CH3 (palmitoyl), is designated herein as GRN163L.
B. Flash PlateTM Assay
This assay was carried out essentially as described in Asai et ai, Cancer Research, 63:3931 3939 (2003). Briefly, the assay detects and/or measures telomerase activity by measuring the addition of TTAGGG telomeric repeats to a biotinylated telomerase substrate primer. The biotinylated products are captured on streptavidin-coated microtiter plates, and an oligonucleotide probe complementary to 3.5 telomere repeats, labeled with 33P, is used for measuring telomerase products. Unbound probe is removed by washing, and the amount of probe annealing to the captured telomerase products is determined by scintillation counting.
C. TRAP Assay
The ability of a compound to increase or inhibit telomerase activity in a cell can be determined using the TRAP (Telomeric Repeat Amplification Protocol) assay, which is described, for example, in Kim et a/., U.S. Patent No. 5,629,154; Harley et al., U.S. Patent No. 5,891 ,639; and Harley et al., PCT Pubn. No. WO 2005/000245. Briefly, telomerase-expressing tumor cell lines are incubated with test compositions, lysed, and treated with a labeled oligonucleotide telomerase substrate, appropriate primers, and internal standard for quantitation purposes. Depending on the telomerase activity of the medium, telomere repeats will be added to the substrate, to form telomerase extended products. The mixture is incubated at room temperature, followed by multiple cycles of PCR. The mixture is separated on a gel, and labeled extension product is detected and quantitated via comparison with the internal standard.
Although the invention has been described with respect to specific agents, formulations, and applications, it will be appreciated that various modifications may be made without departing from the invention as claimed.
3 u Sequence Listing
SEQ ID NO: 1 : the RNA component of human telomerase (hTR): GGGUUGCGGA GGGUGGGCCU GGGAGGGGUG GUGGCCAUUU UUUGUCUAAC CCUAACUGAG 60
AAGGGCGUAG GCGCCGUGCU UUUGCUCCCC GCGCGCUGUU UUUCUCGCUG ACUUUCAGCG 120
GGCGGAAAAG CCUCGGCCUG CCGCCUUCCA CCGUUCAUUC UAGAGCAAAC AAAAAAUGUC 180
AGCUGCUGGC CCGUUCGCCU CCCGGGGACC UGCGGCGGGU CGCCUGCCCA GCCCCCGAAC 240
CCCGCCUGGA GCCGCGGUCG GCCCGGGGCU UCUCCGGAGG CACCCACUGC CACCGCGAAG 300
AGUUGGGCUC UGUCAGCCGC GGGUCUCUCG GGGGCGAGGG CGAGGUUCAC CGUUUCAGGC 360
CGCAGGAAGA GGAACGGAGC GAGUCCCGCC GCGGCGCGAU UCCCUGAGCU GUGGGACGUG 420
CACCCAGGAC UCGGCUCACA CAUGCAGUUC GCUUUCCUGU UGGUGGGGGG AACGCCGAUC 480
GUGCGCAUCC GUCACCCCUC GCCGGCAGUG GGGGCUUGUG AACCCCCAAA CCUGACUGAC 540 UGGGCCAGUG UGCU
SEQ ID NOS: 2-26, the nucleotide sequences of targeting agents against SEQ ID NO: 1 :
Figure imgf000032_0001
Figure imgf000033_0001

Claims

IT IS CLAIMED:
1. A method for inhibiting the proliferation of cancer cells, comprising
(a) exposing the cells to an HSP90 inhibitor, and
(b) either proceeding, following, or concomitantly with step (a), exposing the cells to a telomerase inhibitor, where the amount of each inhibitor to which the cells are exposed is effective, by itself, to inhibit proliferation of the cancer cells.
2. The method of claim 1 , wherein the telomerase inhibitor includes an oligonucleotide having nuclease-resistant intersubunit linkages, and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
3. The method of claim 2, wherein the internucleoside linkages in the oligonucleotide are selected from N3'->P5' phosphoramidate and N3'->P5' thiophosphoramidate linkages.
4. The method of claim 2, wherein the telomerase inhibitor includes a lipid moiety (i) selected from the group consisting of and fatty acids, sterols, and derivatives thereof, and (ii) attached covalently at one end of the oligonucleotide.
5. The method of claim 2, wherein the oligonucleotide is 10-20 bases in length, and includes the sequence identified by SEQ ID NO: 12.
6. The method of claim 5, wherein the telomerase inhibitor is characterized by:
(i) N3'->P5' thiophosphoramidate internucleoside linkages in the oligonucleotide;
(ii) having the sequence identified as SEQ ID NO: 12; and
(iii) linked , at its 5' end, to a palmitoyl (C16) moiety via a glycerol or aminoglycerol linker.
7. The method of claim 1 , wherein the telomerase inhibitor is GRN163L.
8. The method of claim 1 , wherein the HSP90 inhibitor is selected from the group consisting of galdamycin or an analogue thereof, herbimycin A, and radicicol.
9. The method of claim 8, wherein the HSP90 inhibitor is 17-AAG.
10. The method of claim 1 , for use in treating a subject having a cancer, wherein said HSP90 inhibitor is administered to the subject in an amount which is effective, by itself, to inhibit proliferation of the cancer cells in the subject.
11. The method of claim 10, wherein each exposing step (a) and (b) includes administering the respective inhibitor to the subject in an amount effective, when each inhibitor is administered alone, to inhibit proliferation of cancer cells in the subject.
12. The method of claim 10, wherein the inhibitors are administered to a subject diagnosed with a cancer selected from the group consisting of breast cancer, including metastatic breast cancer, ovarian cancer, lung cancer, including non-small-cell lung cancer, and gastrointestinal cancers, prostate, brain and myelomas.
13. The method of claim 10, wherein the telomerase inhibitor is the compound designated herein as GRN163L, and step (b) includes infusing the telomerase inhibitor intravenously to the subject under infusion conditions effective to produce a blood concentration of the inhibitor of between 1 nM and 100 μM.
14. The method of claim 10, wherein said method provides a supraadditive inhibiting effect relative to the effects of the individual inhibitors.
15. A method for enhancing the anti-cancer treatment efficacy of an HSP90 inhibitor administered to a subject, comprising administering to the subject, before, during, or after administering the HSP90 inhibitor, an oligonucleotide telomerase inhibitor of the type composed of an oligonucleotide having nuclease-resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
16. The method of claim 15, wherein the telomerase inhibitor is administered in an amount effective to inhibit the proliferation of cancer cells in the subject, when the telomerase inhibitor is administered alone.
17. The method of claim 15, wherein enhanced treatment efficacy is evidenced by an increased survival time of the subject, an inhibition of tumor growth in the subject, or a combination thereof.
18. The method of claim 15, wherein the oligonucleotide telomerase inhibitor is characterized by:
(i) N3'->P5' thiophosphoramidate internucleoside linkages; (ii) having the sequence identified as SEQ ID NO: 12; and (iii) a palmitoyl (C16) moiety linked to the 5' terminus of the oligonucleotide via a glycerol or aminoglycerol linker.
19. The method of claim 18, wherein the telomerase inhibitor is GRN163L.
20. The method of claim 15, wherein the HSP90 inhibitor is selected from the group consisting of galdamycin or an analogue thereof, herbimycin A, and radicicol.
21. The method of claim 20, wherein the HSP90 inhibitor is 17-AAG.
22. The method of claim 15, wherein the two inhibitors are administered to the subject as a composition containing both inhibitors.
23. A kit for use in cancer therapy, comprising
(a) a dose of an HSP90 inhibitor in an amount effective, when administered alone, to inhibit the proliferation of cancer cells in a subject, and
(b) a dose of an oligonucleotide telomerase inhibitor having nuclease- resistant intersubunit linkages and an oligonucleotide sequence effective to bind by sequence-specific hybridization to a template region of hTR.
24. The kit of claim 23, wherein the dose of oligonucleotide telomerase inhibitor is an amount effective, when administered alone, to inhibit proliferation of cancer cells in the subject.
25. The kit of claim 23, wherein the oligonucleotide telomerase inhibitor is characterized by:
(i) N3'->P5' thiophosphoramidate intemucleoside linkages; (ii) having the sequence identified as SEQ ID NO: 12; and (iii) a palmitoyl (C16) moiety linked to the 5' terminus of the oligonucleotide via a glycerol or aminoglycerol linker.
26. The kit of claim 23, wherein the HSP90 inhibitor is galdanamycin or an analogue thereof.
27. The use of an HSP90 inhibitor and a telomerase inhibitor in the manufacture of a medicament for treating cancer in a subject.
28. The use of a telomerase inhibitor in the manufacture of a medicament for treating cancer in a subject who is being treated with an HSP90 inhibitor, for the purpose of enhancing the anti-cancer efficacy of the HSP90 inhibitor in the subject.
PCT/US2006/018927 2005-05-16 2006-05-16 Cancer treatment by combined inhibition of telomerase and hsp90 activities WO2006124904A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68202905P 2005-05-16 2005-05-16
US60/682,029 2005-05-16

Publications (2)

Publication Number Publication Date
WO2006124904A2 true WO2006124904A2 (en) 2006-11-23
WO2006124904A3 WO2006124904A3 (en) 2007-02-08

Family

ID=37432039

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/018927 WO2006124904A2 (en) 2005-05-16 2006-05-16 Cancer treatment by combined inhibition of telomerase and hsp90 activities

Country Status (1)

Country Link
WO (1) WO2006124904A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2134364A2 (en) * 2007-03-09 2009-12-23 Geron Corporation Treatment of carcinomas with a combination of egf-pathway and telomerase inhibitors
US7989428B2 (en) 2006-10-30 2011-08-02 Geron Corporation Combined telomerase inhibitor and gemcitabine for the treatment of cancer
US8071766B2 (en) 2008-02-01 2011-12-06 Takeda Pharmaceutical Company Limited HSP90 inhibitors
EP2685990A1 (en) * 2011-03-15 2014-01-22 The University Of British Columbia Combination of anti-clusterin oligonucleotide with hsp90 inhibitor for the treatment of prostate cancer
US9200327B2 (en) 2012-11-30 2015-12-01 Geron Corporation Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
US11278561B2 (en) 2016-08-02 2022-03-22 Geron Corporation Combination treatment for hematological cancers

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
AKIYAMA ET AL.: 'Effects of oligonucleotide N3-P5' thio-phosphoramidate (GRN163) targeting telomerase RNA in human multiple myeloma cells' CANCER RESEARCH vol. 63, 01 October 2003, pages 6187 - 6194, XP003005976 *
ASAI ET AL.: 'A novel telomerase template antagonist (GRN163) as a potential anticancer agent' CANCER RESEARCH vol. 63, 15 July 2003, pages 3931 - 3939, XP003005975 *
ENMON ET AL.: 'Combination Treatment with 17-N-Allylamino-17-Demethoxy Geldanamycin and acute irradiation produces supra-additive growth suppression in human prostate carcinoma spheroids' CANCER RESEARCH vol. 63, 01 December 2003, pages 8393 - 8399, XP003005977 *
MIYATA Y.: 'Hsp90 inhibitor geldanamycin and its derivatives as novel cancer chemotherapeutic agents' CURRENT PHARMACEUTICAL DESIGN vol. 11, 2005, pages 1131 - 1138, XP008075940 *
VILLA ET AL.: 'Inhibition of telomerase activity by geldanamycin and 17-allylamino, 17-demethoxygeldanamycin in human melanoma cells' CARCINOGENESIS vol. 24, no. 5, pages 851 - 859, XP003005978 *
XIANG ET AL.: 'The antiproliferative effect of inhibitor of telomerase cultured retinal pigment epithelial cells' J. TONGJI MED. UNIV. vol. 21, no. 2, 2001, pages 174 - 176, XP008018763 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7989428B2 (en) 2006-10-30 2011-08-02 Geron Corporation Combined telomerase inhibitor and gemcitabine for the treatment of cancer
EP2134364A2 (en) * 2007-03-09 2009-12-23 Geron Corporation Treatment of carcinomas with a combination of egf-pathway and telomerase inhibitors
EP2134364A4 (en) * 2007-03-09 2010-11-03 Geron Corp Treatment of carcinomas with a combination of egf-pathway and telomerase inhibitors
US9155753B2 (en) 2007-03-09 2015-10-13 Geron Corporation Treatment of carcinomas with a combination of EGF-pathway and telomerase inhibitors
US8071766B2 (en) 2008-02-01 2011-12-06 Takeda Pharmaceutical Company Limited HSP90 inhibitors
US8618290B2 (en) 2008-02-01 2013-12-31 Takeda Pharmaceutical Company Limited HSP90 inhibitors
EP2685990A1 (en) * 2011-03-15 2014-01-22 The University Of British Columbia Combination of anti-clusterin oligonucleotide with hsp90 inhibitor for the treatment of prostate cancer
EP2685990A4 (en) * 2011-03-15 2014-09-17 Univ British Columbia Combination of anti-clusterin oligonucleotide with hsp90 inhibitor for the treatment of prostate cancer
US9457045B2 (en) 2011-03-15 2016-10-04 The University Of British Columbia Combination of anti-clusterin oligonucleotide with Hsp90 inhibitor for the treatment of prostate cancer
US9200327B2 (en) 2012-11-30 2015-12-01 Geron Corporation Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
US9951389B2 (en) 2012-11-30 2018-04-24 Geron Corporation Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
US11278561B2 (en) 2016-08-02 2022-03-22 Geron Corporation Combination treatment for hematological cancers

Also Published As

Publication number Publication date
WO2006124904A3 (en) 2007-02-08

Similar Documents

Publication Publication Date Title
US7998938B2 (en) Cancer treatment by combined inhibition of proteasome and telomerase activities
JP6158966B2 (en) Modified oligonucleotides for telomerase inhibition
US9155753B2 (en) Treatment of carcinomas with a combination of EGF-pathway and telomerase inhibitors
DK2101789T3 (en) Combined telomerase inhibitor and gemcitabine for the treatment of cancer
US8153604B2 (en) CNS-tumor treatment method and composition
WO2006124904A2 (en) Cancer treatment by combined inhibition of telomerase and hsp90 activities
WO2006113426A2 (en) Cancer treatment by combined inhibition of telomerase and tubulin activities

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06770442

Country of ref document: EP

Kind code of ref document: A2