WO2004106518A1 - Oligonucleotides antisens diriges contre la ribonucleotide reductase r2 et leurs utilisations dans le traitement du cancer - Google Patents

Oligonucleotides antisens diriges contre la ribonucleotide reductase r2 et leurs utilisations dans le traitement du cancer Download PDF

Info

Publication number
WO2004106518A1
WO2004106518A1 PCT/CA2004/000806 CA2004000806W WO2004106518A1 WO 2004106518 A1 WO2004106518 A1 WO 2004106518A1 CA 2004000806 W CA2004000806 W CA 2004000806W WO 2004106518 A1 WO2004106518 A1 WO 2004106518A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumour
antisense oligonucleotide
seq
cancer
oligonucleotide according
Prior art date
Application number
PCT/CA2004/000806
Other languages
English (en)
Inventor
Aiping H. Young
Jim A. Wright
Original Assignee
Genesense Technologies Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genesense Technologies Inc. filed Critical Genesense Technologies Inc.
Publication of WO2004106518A1 publication Critical patent/WO2004106518A1/fr

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
    • A61K38/00Medicinal preparations containing peptides
    • 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

Definitions

  • the present invention pertains to the field of cancer therapeutics and in particular to the use of antisense oligonucleotides alone or in combination with one or more chemotherapeutic drugs for the treatment of cancer.
  • ribonucleotide reductase and particularly the R2 component, is altered in malignant cells exposed to some tumour promoters and to the growth factor TGF- ⁇ [A ara, et al., 1994; Chen et al., 1993; Amara et al., 1995b; Hurta and Wright, 1995; Hurta et al., 1991].
  • Antisense oligonucleotides directed to the Rl or R2 component of ribonucleotide reductase have been shown to be effective in reducing the growth of cancer cells [see, for example, U.S. Patent Nos. 5,998,383 and 6,121,000].
  • U.S. Patent Nos. 5,998,383 and 6,121,000 In view of the high incidence of various types of cancer throughout the world, there remains a need for improved therapies for the treatment of cancer.
  • An object of the present invention is to provide antisense oligonucleotides directed to ribonucleotide reductase R2 and uses thereof in the treatment of cancer.
  • an antisense oligonucleotide of between 7 and 100 nucleotides in length comprising at least 7 consecutive nucleotides from SEQ ID NO:l for use in combination with one or more chemotherapeutic agents in the treatment of cancer in a mammal in need of such therapy.
  • an antisense oligonucleotide of between 20 and 100 nucleotides in length comprising the sequence as set forth in SEQ ID NO: 1 for use in combination with one or more chemotherapeutic agents in the treatment of a human having a cancer selected from the group of: a solid tumour, renal cancer, breast cancer, lung cancer, prostate cancer, colon cancer and leukaemia.
  • an antisense oligonucleotide of between 7 and 100 nucleotides in length comprising at least 7 consecutive nucleotides from SEQ ID NO:l for use in the treatment of cancer in a mammal in need of such therapy.
  • an antisense oligonucleotide of between 7 and 100 nucleotides in length comprising at least 7 consecutive nucleotides from SEQ ID NO: 1 in the manufacture of a medicament for the treatment of cancer.
  • Figure 1 depicts effects of combination therapy on HT-29 colon tumour growth in nude mice
  • Figure 2 depicts effects of combination therapy on HT-29 colon tumour growth in nude mice
  • Figure 3 depicts effects of combination therapy on HT-29 colon tumour growth in nude mice
  • Figure 4 depicts effects of combination therapy on HT-29 colon tumour growth in nude mice
  • FIG. 5 depicts effects of combination therapy on Caki-1 renal tumour growth in SCID mice
  • Figure 6 depicts effects of combination therapy on prostatic tumour growth in SCID mice
  • Figure 7 depicts effects of combination therapy on prostatic tumour growth in SCID mice.
  • Figure 8 depicts effects of combination therapy on A2058 melanoma growth in nude mice
  • Figure 9 depicts effects of combination therapy on breast tumour growth in CD-I nude mice
  • Figure 10 depicts effects of combination therapy on ovary tumour growth in CD-I nude mice
  • Figure 11 depicts effects of SEQ ID NO: 1 in the treatment of human pancreatic carcinoma in CD-I nude mice
  • Figure 12 depicts effects of SEQ ID NO: 1 in the treatment of human cervix epitheloid carcinoma resistant to hydroxyurea (HU) in SCID mice
  • Figure 13 depicts effects of SEQ ID NO: 1 in the treatment of human breast adenocarcinoma resistant to cisplatin in SCID mice;
  • Figure 14 depicts effects of SEQ ID NO: 1 in the treatment of human breast adenocarcinoma resistant to cisplatin in SCID mice;
  • Figure 15 depicts effects of SEQ ID NO: 1 in the treatment of human breast adenocarcinoma resistant to taxol in SCID mice;
  • Figure 16 depicts effects of SEQ ID NO: 1 in the treatment of human breast adenocarcinoma resistant to taxol in SCID mice;
  • Figure 17 depicts effects of SEQ ID NO: 1 in the treatment of human promyelocytic leukaemia resistant to taxol in SCID mice;
  • Figure 18 depicts effects of SEQ ID NO: 1 in the treatment of LS513, human multi- drug resistant colon adenocarcinoma in SCID micev
  • Figure 19 depicts effects of SEQ ID NO: 1 on HT-29 colon tumour growth in CD-I nude mice
  • Figure 20 depicts effects of SEQ ID NO: 1 on (A) A2058 melanoma growth in CD-I nude mice, (B) MDA-MB-231 breast tumour growth in CD-I nude mice, (C) SK-ON- 3 ovary tumour growth in Balb/c nude mice, and (D) ⁇ CI-H460 lung tumour growth in CD-I nude mice;
  • Figure 21 depicts effects of SEQ ID NO: 1 on SU.86.86 pancreatic tumour growth in CD-I nude mice;
  • Figure 22 depicts effects of SEQ ID NO: 1 on HepG2 liver tumour growth in CD-I nude mice
  • Figure 23 depicts effects of SEQ ID NO: 1 on (A) Caki-1 kidney tumour growth in CD-I nude mice, and (B) A498 kidney tumour growth in SCID mice;
  • Figure 24 depicts effects of SEQ ID NO: 1 on SIHA cervical tumour growth in SCID mice
  • Figure 25 depicts effects of SEQ ID NO: 1 on HeLa S3 cervical tumour growth in SCID mice
  • Figure 26 depicts the reduction of lung nodule formation by SEQ ID NO: 1 in a mouse experimental model of metastasis using (A) mouse fibrosarcoma (R3) cells, and (B) C8161 human melanoma cells;
  • Figure 27 depicts survival time of SCID mice bearing human lymphoma (Raji);
  • Figure 28 depicts survival time of CB-17 SCID mice bearing erythroleukaemia (CB7)
  • Figure 29 depicts the effect of SEQ ID NO:l on the growth of R3 mouse fibrosarcoma cells in C3H mice;
  • Figure 30 depicts the effect of SEQ ID NO:l on the growth of Caki-1 human kidney cancer cells and A498 human kidney cells in SCID mice;
  • Figure 31 depicts the effect of SEQ ID NO: 1 in a variety of human tumour cell lines in xenograft experiments
  • Figure 32 depicts the effect of SEQ ID NO:l on the survival of mice injected with human Burkitt's lymphoma (Raji) cells;
  • Figure 33 depicts the effect of SEQ ID NO:l on metastasis of C8161 melanoma cells in SCID mice;
  • Figure 34 depicts effects of SEQ ID NO:l on Caki renal tumour xenografts in SCID/Beige mice;
  • Figure 35 depicts effects of SEQ ID NOs:l, 4 and 5 on HT29 tumour xenografts in CD-I nude mice.
  • Figure 36 presents the results from Phase I clinical trials for SEQ ID NO:l, (A) depicts the AUC vs. Actual Dose plot for SEQ ID NO:l, and (B) depicts the plasma concentration of SEQ ID NO: 1 vs. time. DETAILED DESCRIPTION OF THE INVENTION
  • the present invention relates to antisense oligonucleotides against the gene encoding a mammalian ribonucleotide reductase R2 protein and combinations of such antisense oligonucleotides and one or more chemotherapeutic agents in the treatment of various cancers.
  • the antisense oligonucleotides and combinations of antisense oligonucleotides with one or more chemotherapeutic agents are effective in decreasing the growth and/or metastasis of cancers, including refractory, advanced and drug resistant cancer cells, than treatment with the antisense oligonucleotide or the chemotherapeutic agent(s) alone.
  • antisense oligonucleotide as used herein means a nucleotide sequence that is complementary to the mRNA for a desired gene.
  • the desired gene is the gene encoding a mammalian ribonucleotide multiplitase R2 protein.
  • selective hybridise refers to the ability of a nucleic acid to bind detectably and specifically to a second nucleic acid. Oligonucleotides selectively hybridise to target nucleic acid strands under hybridisation and wash conditions that minimise appreciable amounts of detectable binding to non-specific nucleic acids. High stringency conditions can be used to achieve selective hybridisation conditions as known in the art and discussed herein.
  • hybridisation and washing conditions are performed at high stringency according to conventional hybridisation procedures. Washing conditions are typically 1-3 x SSC, 0.1-1% SDS, 50-70°C with a change of wash solution after about 5-30 minutes.
  • the term "corresponds to” as used herein with reference to nucleic acid sequences means a polynucleotide sequence that is identical to all or a portion of a reference polynucleotide sequence.
  • the term “complementary to” is used herein to mean that the polynucleotide sequence is identical to all or a portion of the complement of a reference polynucleotide sequence.
  • the nucleotide sequence "TAT AC” corresponds to a reference sequence "TAT AC” and is complementary to a reference sequence "GTATA”.
  • sequence identity is a defined sequence used as a basis for a sequence comparison; a reference sequence may be a subset of a larger sequence, for example, as a segment of a full-length cDNA or gene sequence, or may comprise a complete cDNA or gene sequence. Generally, a reference polynucleotide sequence is at least 20 nucleotides in length, and often at least 50 nucleotides in length.
  • a “window of comparison”, as used herein, refers to a conceptual segment of the reference sequence of at least 15 contiguous nucleotide positions over which a candidate sequence may be compared to the reference sequence and wherein the portion of the candidate sequence in the window of comparison may comprise additions or deletions (t.e. gaps) of 20 percent or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the present invention contemplates various lengths for the window of comparison, up to and including the full length of either the reference or candidate sequence.
  • Optimal alignment of sequences for aligning a comparison window may be conducted using the local homology algorithm of Smith and Waterman (Adv. Appl Math.
  • sequence identity means that two polynucleotide sequences are identical (i.e. on a nucleotide-by-nucleotide basis) over the window of comparison.
  • percent (%) sequence identity as used herein with respect to a reference sequence is defined as the percentage of nucleotide residues in a candidate sequence that are identical with the residues in the reference polynucleotide sequence over the window of comparison after optimal alignment of the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, without considering any conservative substitutions as part of the sequence identity.
  • substantially identical denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 50% sequence identity as compared to a reference sequence over the window of comparison.
  • Polynucleotide sequences at least 60% sequence identity, at least 70% sequence identity, at least 80% sequence identity, and at least 90% sequence identity as compared to a reference sequence over the window of comparison are also considered to have substantial identity with the reference sequence.
  • therapy refers to an intervention performed with the intention of improving a recipient's status.
  • the improvement can be subjective or objective and is related to the amelioration of the symptoms associated with, preventing the development of, or altering the pathology of a disease, disorder or condition being treated.
  • therapy and treatment are used in the broadest sense, and include the prevention (prophylaxis), moderation, reduction, and curing of a disease, disorder or condition at various stages. Prevention of deterioration of a recipient's status is also encompassed by the term.
  • Those in need of therapy/treatment include those already having the disease, disorder or condition as well as those prone to, or at risk of developing, the disease, disorder or condition and those in whom the disease, disorder or condition is to be prevented.
  • the term “ameliorate” or “amelioration” includes the arrest, prevention, decrease, or improvement in one or more the symptoms, signs, and features of the disease being treated, both temporary and long-term.
  • subject or "patient” as used herein refers to a mammal in need of treatment.
  • Administration of the compounds of the invention "in combination with" one or more further therapeutic agents is intended to include simultaneous (concurrent) administration and consecutive administration. Consecutive administration is intended to encompass administration of the therapeutic agent(s) and the compound(s) of the invention to the subject in various orders.
  • the term "about” refers to a +/-10% variation from the nominal value. It is to be understood that such a variation is always included in any given value provided herein, whether or not it is specifically referred to.
  • the antisense oligonucleotides of the present invention are targeted to the gene encoding a mammalian ribonucleotide reductase R2 protein.
  • the sequences of various mammalian ribonucleotide reductase genes are known in the art, for example, the sequence for the human ribonucleotide reductase R2 gene is provided in Pavloff et al. [J. DNA sequencing and Mapping, 2;227-234 (1992)]. This and other mammalian R2 sequences are also available from the GenBank database maintained by the NCBI.
  • the antisense oligonucleotides of the present invention comprise at least 7 contiguous nucleotides, or nucleotide analogues, that correspond to a part of the coding region of a mammalian ribonucleotide reductase R2 gene.
  • antisense oligonucleotide examples include those disclosed in U.S. Patent Nos. 5,998,383 and 6,121,000 (herein incorporated by reference) which are targeted to the ribonucleotide reductase R2 gene.
  • the antisense oligonucleotide comprises at least 7 consecutive nucleotides of the antisense oligonucleotide represented by the sequence:
  • the antisense oligonucleotides in accordance with the present invention are selected such that the sequence exhibits the least likelihood of forming duplexes, hair-pins, dimers, or of containing homooligomer / sequence repeats.
  • the oligonucleotide may further contain a GC clamp.
  • a GC clamp One skilled in the art will appreciate that these properties can be determined qualitatively using various computer modelling programs, for example, the program OLIGO ® Primer Analysis Software, Version 5.0 (distributed by National Biosciences, Inc., Plymouth, MN).
  • antisense oligonucleotides are typically between 7 and 100 nucleotides in length. In one embodiment of the present invention, the antisense oligonucleotides are between about 7 to about 50 nucleotides in length. In other embodiments, the antisense oligonucleotides are between about 7 to about 35 nucleotides in length, between about 15 to about 25 nucleotides in length, and about 20 nucleotides in length.
  • an antisense oligonucleotide need not have 100% identity with the complement of its target sequence.
  • the antisense oligonucleotides in accordance with the present invention have a sequence that is at least about 75% identical to the complement of target sequence.
  • the antisense oligonucleotides have a sequence that is at least about 90% identical to the complement of the target sequence.
  • they have a sequence that is at least about 95% identical to the complement of target sequence, allowing for gaps or mismatches of several bases.
  • Identity can be determined, for example, by using the BLASTN program of the University of Wisconsin Computer Group (GCG) software or provided on the NCBI website.
  • antisense oligonucleotides as used herein includes other oligomeric antisense compounds, including oligonucleotide mimetics, modified oligonucleotides, and chimeric antisense compounds.
  • Chimeric antisense compounds are antisense compounds that contain two or more chemically distinct regions, each made up of at least one monomer unit.
  • oligonucleotide refers to an oligomer or polymer of ribonucleic acid (RNA), deoxyribonucleic acid (DNA), or RNA or DNA mimetics.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • RNA or DNA mimetics oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions, which function similarly.
  • backbone internucleoside
  • modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
  • a nucleoside is a base-sugar combination and a nucleotide is a nucleoside that further includes a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound, with the normal linkage or backbone of RNA and DNA being a 3' to 5' phosphodiester linkage.
  • antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages.
  • oligonucleotides having modified backbones include both those that retain a phosphorus atom in the backbone and those that lack a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleotides.
  • Exemplary antisense oligonucleotides having modified oligonucleotide backbones include, for example, those with one or more modified internucleotide linkages that are phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'.
  • the antisense oligonucleotide comprises one or more phosphorothioate internucleotide linkage. In another embodiment, the antisense oligonucleotide comprises phosphorothioate internucleotide linkages that link the four, five or six 3 '-terminal nucleotides of the oligonucleotide. In a further embodiment, the antisense oligonucleotide comprises phosphorothioate internucleotide linkages that link all the nucleotides of the oligonucleotide.
  • Exemplary modified oligonucleotide backbones that do not include a phosphorus atom are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • Such backbones include morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulphone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulphamate backbones; methyleneimino and methylenehydrazino backbones; sulphonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • the present invention also contemplates oligonucleotide mimetics in which both the sugar and the internucleoside linkage of the nucleotide units are replaced with novel groups.
  • the base units are maintained for hybridisation with an appropriate nucleic acid target compound.
  • An example of such an oligonucleotide mimetic which has been shown to have excellent hybridisation properties, is a peptide nucleic acid (PNA) [Nielsen et al, Science, 254:1497-1500 (1991)].
  • PNA peptide nucleic acid
  • the sugar- backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • the nucleobases are retained and are bound directly or indirectly to aza-nitrogen atoms of the amide portion of the backbone.
  • the present invention also contemplates oligonucleotides comprising "locked nucleic acids” (LNAs), which are confbrmationally restricted oligonucleotide analogues containing a methylene bridge that connects the 2'-O of ribose with the 4'-C (see, Singh et al, Chem. Commun., 1998, 4:455-456).
  • LNA and LNA analogues display very high duplex thermal stabilities with complementary DNA and RNA, stability towards 3'-exonuclease degradation, and good solubility properties.
  • Antisense oligonucleotides containing LNAs have been described (Wahlestedt et al, Proc. Natl. Acad. Set U. S. A., 2000, 97:5633-5638), which were efficacious and non- toxic.
  • the LNA DNA copolymers were not degraded readily in blood serum and cell extracts.
  • LNAs form duplexes with complementary DNA or RNA or with complementary LNA, with high thermal affinities.
  • the universality of LNA-mediated hybridization has been emphasized by the formation of exceedingly stable LNA:LNA duplexes (Koshkin et al, J. Am. Chem. Soc, 1998, 120:13252-13253).
  • LNA:LNA hybridization was shown to be the most thermally stable nucleic acid type duplex system, and the RNA-mimicking character of LNA was established at the duplex level.
  • Introduction of three LNA monomers (T or A) resulted in significantly increased melting points toward DNA complements.
  • oligonucleotides may comprise sugars with one of the following substituents at the 2' position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci to Cio alkyl or C 2 to Cio alkenyl and alkynyl.
  • Examples of such groups are: O[(CH 2 ) n O] m CH 3 , O(CH 2 ) n OCH 3 , O(CH 2 ) n NH 2 , O(CH 2 ) n CH 3 , O(CH 2 ) supervise ONH 2 , and O(CH 2 ) n ON[(CH 2 ) n CH 3 )] 2 , where n and m are from 1 to about 10.
  • the oligonucleotides may comprise one of the following substituents at the 2' position: Ci to C 10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O- alkaryl or O-aralkyl, SH, SCH 3 , OCN, CI, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • 2'-methoxyethoxy (2'-O ⁇ CH 2 CH 2 OCH 3 , also known as 2*-O-(2- methoxyethyl) or 2'-MOE) [Martin et al, Helv. Chim. Acta, 78:486-504(1995)], 2'- dimethylaminooxyethoxy (O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE), 2'- methoxy (2'-O-CH 3 ), 2'-aminopropoxy (2'-OCH 2 CH 2 CH 2 NH 2 ) and 2'-fluoro (2'-F).
  • the antisense oligonucleotide comprises at least one nucleotide comprising a substituted sugar moiety. In another embodiment, the antisense oligonucleotide comprises at least one 2'-O-(2-methoxyethyl) or 2'-MOE modified nucleotide.
  • Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
  • Oligonucleotides may also include modifications or substitutions to the nucleobase.
  • "unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5- hydroxymethyl cytosine, xanthine, hypoxanthine, 2- aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substitute
  • nucleobases include those disclosed in U.S. Pat. No. 3,687,808; The Concise Encyclopedia Of Polymer Science And Engineering, (1990) pp 858-859, Kroschwitz, J. I., ed. John Wiley & Sons; Englisch et al, Angewandte Chemie, Int. Ed., 30:613 (1991); and Sanghvi, Y. S., (1993) Antisense Research and Applications, pp 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention.
  • 5-substituted pyrimidines include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N- 6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C [Sanghvi, Y. S., (1993) Antisense Research and Applications, pp 276-278, Crooke, S. T. and Lebleu, B., ed., CRC Press, Boca Raton].
  • oligonucleotide modification included in the present invention is the chemical linkage to the oligonucleotide of one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties include, but are not limited to, lipid moieties such as a cholesterol moiety [Letsinger et al, Proc. Natl. Acad. Sci. USA, 86:6553-6556 (1989)], cholic acid [Manoharan et al, Bioorg. Med. Chem. Let, 4:1053-1060 (1994)], a thioether, e.g.
  • a phospholipid e.g. di-hexadecyl-rac-glycerol or triethylammonium l,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate [Manoharan et al, Zetrahedron Lett, 36:3651-3654 (1995); Shea etal, Nucl.
  • the present invention contemplates the incorporation of more than one of the aforementioned modifications into a single oligonucleotide or even at a single nucleoside within the oligonucleotide.
  • the present invention further includes antisense compounds that are chimeric compounds. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonuclease that cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridising to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridisation techniques known in the art.
  • an antisense oligonucleotide is "nuclease resistant" when it has either been modified such that it is not susceptible to degradation by DNA and RNA nucleases or alternatively has been placed in a delivery vehicle which in itself protects the oligonucleotide from DNA or RNA nucleases.
  • Nuclease resistant oligonucleotides include, for example, methyl phosphonates, phosphorothioates, phosphorodithioates, phosphotriesters, and morpholino oligomers.
  • Suitable delivery vehicles for conferring nuclease resistance include, for example, liposomes.
  • the antisense oligonucleotides are nuclease resistant.
  • the present invention further contemplates antisense oligonucleotides that contain groups for improving the pharmacokinetic properties of the oligonucleotide, or groups for improving the pharmacodynamic properties of the oligonucleotide.
  • the antisense oligonucleotides of the present invention can be prepared by conventional techniques well-known to those skilled in the art.
  • the oligonucleotides can be prepared using solid-phase synthesis using commercially available equipment, such as the equipment available from Applied Biosystems Canada Inc., Mississauga, Canada.
  • modified oligonucleotides such as phosphorothioates and alkylated derivatives, can also be readily prepared by similar methods.
  • the antisense oligonucleotides of the present invention can be prepared by enzymatic digestion of the naturally occurring ribonucelotide reductase R2 gene by methods known in the art.
  • Antisense oligonucleotides can also be prepared through the use of recombinant methods in which expression vectors comprising nucleic acid sequences that encode the antisense oligonucleotides are expressed in a suitable host cell.
  • expression vectors can be readily constructed using procedures known in the art. Examples of suitable vectors include, but are not limited to, plasmids, phagemids, cosmids, bacteriophages, baculoviruses and retroviruses, and DNA viruses.
  • suitable vectors include, but are not limited to, plasmids, phagemids, cosmids, bacteriophages, baculoviruses and retroviruses, and DNA viruses.
  • host cells include, but are not limited to, bacterial, yeast, insect, plant and mammalian cells.
  • the expression vector may further include regulatory elements, such as transcriptional elements, required for efficient transcription of the antisense oligonucleotide sequences.
  • regulatory elements such as transcriptional elements
  • Examples of regulatory elements that can be incorporated into the vector include, but are not limited to, promoters, enhancers, terminators, and polyadenylation signals.
  • selection of suitable regulatory elements is dependent on the host cell chosen for expression of the antisense oligonucleotide and that such regulatory elements may be derived from a variety of sources, including bacterial, fungal, viral, mammalian or insect genes.
  • the expression vectors can be introduced into a suitable host cell or tissue by one of a variety of methods known in the art. Such methods can be found generally described in Sambrook et al, 1992; Ausubel et al, 1989; Chang et al, 1995; Nega et al, 1995; and Vectors: A Survey of Molecular Cloning Vectors and Their Uses (1988) and include, for example, stable or transient transfection, lipofection, electroporation, and infection with recombinant viral vectors.
  • the chemotherapeutic agent can be selected from a wide range of cancer chemotherapeutic agents known in the art.
  • chemotherapeutic agents include those that are specific for the treatment of a particular type of cancer as well as those that are applicable to a range of cancers, such as doxorubicin, capecitabine, mitoxantrone, irinotecan (CPT-11) and gemcitabine.
  • Etoposide is generally applicable in the treatment of leukaemias
  • Cytarabine is also applicable in the treatment of various leukaemias, including acute myeloid leukaemia, meningeal leukaemia, acute lymphocytic leukaemia, chronic myeloid leukaemia, erythroleukaemia , as well as non-Hodgkin's lymphoma.
  • the present invention contemplates the use of both types of chemotherapeutic agent in conjunction with the antisense oligonucleotides.
  • chemotherapeutic agent in conjunction with the antisense oligonucleotides.
  • Exemplary chemotherapeutics that can be used alone or in various combinations for the treatment specific cancers are provided in Table 1.
  • Table 1 One skilled in the art will appreciate that many other chemotherapeutics are available and that the following list is representative only.
  • Acute lymphocytic Pegaspargase e.g. Oncaspar®
  • L-asparaginase leukaemia ALL
  • Interleukin-2 e.g. Proleukin®
  • Mitoxantrone e.g. Novantrone®
  • AML Acute myeloid Cytarabine Idarubicin leukaemia
  • Brain cancer Procarbazine e.g. Matulane®
  • Paclitaxel e.g. Taxol®
  • Cisplatin Cisplatin
  • Docetaxel e.g. Taxotere®
  • Ifosfamide Ifosfamide
  • IFN Chronic myeloid Low-dose Interferon
  • CML leukaemia
  • Cytarabine Fludarabine e.g. Fludara®
  • Gemcitabine e.g. Gemzar®
  • Colorectal cancer Irinotecan (CPT-l l, e.g. Camptosar®)
  • Loperamide e.g. Imodium®
  • CPT-ll Small cell lung Irinotecan (CPT-ll, e.g. Camptosar®) cancer
  • Paclitaxel e.g. Taxol®
  • Cisplatin Cisplatin
  • Docetaxel e.g. Taxotere®
  • Carboplatin e.g. Taxotere®
  • Combination therapies using standard cancer chemotherapeutics are well known in the art and such combinations also can be used in conjunction with the antisense oligonucleotides of the invention.
  • Exemplary combination therapies include for the treatment of breast cancers the combination of epirubicin with paclitaxel or docetaxel, or the combination of doxorubicin or epirubicin with cyclophosphamide.
  • Polychemotherapeutic regimens are also useful and may consist, for example, of doxorubicin/cyclophosphamide/5- fluorouracil or cyclophosphamide/epirubicin/5-fluorouracil. Many of the above combinations are useful in the treatment of a variety of other solid tumours.
  • Combinations of etoposide with either cisplatin or carboplatin are used in the treatment of small cell lung cancer.
  • combinations of doxorubicin or epirubicin with cisplatin and 5-fluorouracil are useful.
  • CPT-11 in combination with 5-fluorouracil-based drugs, or oxaliplatin in combination with 5-fluorouracil-based drugs can be used.
  • Oxaliplatin may also be used in combination with capecitabine.
  • cyclophosphamide doxorubicin, vincristine and prednisone
  • doxorubicin bleomycin, vinblastine and dacarbazine
  • DTIC dacarbazine
  • gemcitabine paclitaxel
  • docetaxel docetaxel
  • vinorelbine etoposide
  • sarcomas are treated by combination therapy, for example, for osteosarcoma combinations of doxorubicin and cisplatin or methotrexate with leucovorin are used; for advanced sarcomas etoposide can be used in combination with ifosfamide; for soft tissue sarcoma doxorubicin or dacarbazine can be used alone or, for advanced sarcomas doxorubicin can be used in combination with ifosfamide or dacarbazine, or etoposide in combination with ifosfamide.
  • Ewing's sarcoma/peripheral neuroectodermal tumour (PNET) or rhabdomyosarcoma can be treated using etoposide and ifosfamide, or a combination of vincristine, doxorubicin and cyclophosphamide.
  • alkylating agents cyclophosphamide, cisplatin and melphalan are also often used in combination therapies with other chemotherapeutics in the treatment of various cancers.
  • Suitable combinations of the antisense oligonucleotide and one or more chemotherapeutic agent include, but are not limited to, a combination of the antisense oligonucleotide
  • capecitabine alone or in combination with other chemotherapeutics, for the treatment of solid tumours including, but not limited to, breast cancer, renal cancer, colon cancer, colorectal cancer and pancreatic cancer, for example, a combination of capecitabine and oxaliplatin for the treatment of colorectal cancer, colon cancer and pancreatic cancer or a combination of capecitabine and gemcitabine for the treatment of colon cancer;
  • SCLC small-cell lung carcinoma
  • cytarabine alone or in combination with other chemotherapeutics, for the treatment of acute myeloid leukaemia (AML) and chronic myeloid leukaemia (CML), for example, a combination of cytarabine, fludarabine and filgrastim for, the treatment of CML, or a combination of cytarabine, mitoxantrone and etoposide for the treatment of AML;
  • NSCLC non-small cell lung carcinoma
  • breast cancer breast cancer
  • prostate cancer cancer of the genitourinary tract
  • gemcitabine alone or in combination with other chemotherapeutics, for the treatment of solid tumours, including, but not limited to, NSCLC, breast cancer and renal cancer, for example, a combination of gemcitabine and oxaliplatin for the treatment of breast cancer;
  • - with irinotecan alone or in combination with other chemotherapeutics, for the treatment of pancreatic cancer and colon cancer
  • - with mitoxantrone alone or in combination with other chemotherapeutics, for the treatment of prostate cancer and colon cancer, for example, a combination of mitoxantrone and prednisone for the treatment of prostate cancer
  • the antisense oligonucleotides of the present invention can be initially tested, alone or in combination with other chemotherapeutic(s), for their ability to attenuate the growth and/or metastasis of cancer cells in vitro and/or in vivo.
  • Methods of testing potential anti-cancer compounds are known in the art. Exemplary, non-limiting tests are provided below and in the Examples included herein.
  • the antisense oligonucleotides or combinations of the antisense oligonucleotides with one or more chemotherapeutic agents can be tested in vitro by determining their ability to inhibit anchorage-independent growth of tumour cells.
  • Anchorage-independent growth is known in the art to be a good indicator of tumourigenicity. In general, anchorage-independent growth is assessed by plating cells from an appropriate cancer cell-line onto soft agar and determining the number of colonies formed after an appropriate incubation period.
  • Growth of cells treated with the antisense oligonucleotides alone or combinations can then be compared with that of cells treated with an appropriate control (such as cells treated with a scrambled control oligonucleotide or a known chemotherapeutic, or untreated cells) and with that of untreated cells.
  • an appropriate control such as cells treated with a scrambled control oligonucleotide or a known chemotherapeutic, or untreated cells
  • in vitro testing of the antisense oligonucleotides and combinations is conducted in a human cancer cell-line.
  • suitable cancer cell-lines for in vitro testing of the antisense oligonucleotides or combinations of the present invention are known in the art and include those described in the Examples provided herein.
  • the toxicity of the antisense oligonucleotides and combinations can also be initially assessed in vitro using standard techniques.
  • human primary fibroblasts can be treated in vitro with the oligonucleotide in the presence of a commercial lipid carrier such as lipofectamine.
  • a commercial lipid carrier such as lipofectamine.
  • Cells are then tested at different time points following treatment for their viability using a standard viability assay, such as the trypan-blue exclusion assay.
  • a standard viability assay such as the trypan-blue exclusion assay.
  • Cells are also assayed for their ability to synthesize DNA, for example, using a thymidine incorporation assay, and for changes in cell cycle dynamics, for example, using a standard cell sorting assay in conjunction with a fluorocytometer cell sorter (FACS).
  • FACS fluorocytometer cell sorter
  • antisense oligonucleotides and combinations to inhibit tumour growth or proliferation in vivo can be determined in an appropriate animal model using standard techniques known in the art (see, for example, Enna, et al, Current Protocols in Pharmacology, J. Wiley & Sons, Inc., New York, NY).
  • xenograft models in which a human tumour has been implanted into an animal.
  • xenograft models of human cancer include, but are not limited to, human solid tumour xenografts in mice, implanted by sub-cutaneous injection and used in tumour growth assays; human solid tumour isografts in mice, implanted by fat pad injection and used in tumour growth assays; experimental models of lymphoma and leukaemia in mice, used in survival assays, and experimental models of lung metastasis in mice. Representative, non-limiting examples are provided in Table 2 and in the Examples provided herein.
  • the antisense oligonucleotides and combinations can be tested in vivo on solid tumours using mice that are subcutaneously grafted bilaterally with a predetermined amount of a tumour fragment on day 0.
  • the animals bearing tumours are mixed before being subjected to the various treatments and controls.
  • tumours are allowed to develop to the desired size, animals having insufficiently developed tumours being eliminated.
  • the selected animals are distributed at random into groups that will undergo the treatments or act as controls. Suitable groupings would be, for example, those receiving the combination of the invention, those receiving the antisense alone, those receiving the chemotherapeutic agent(s) alone and those receiving no treatment.
  • tumour-bearing animals Animals not bearing tumours may also be subjected to the same treatments as the tumour-bearing animals in order to be able to dissociate the toxic effect from the specific effect on the tumour.
  • Chemotherapy generally begins from 3 to 22 days after grafting, depending on the type of tumour, and the animals are observed every day.
  • the antisense oligonucleotides or combinations of the present invention can be administered to the animals, for example, by bolus infusion.
  • the different animal groups are weighed about 3 or 4 times a week until the maximum weight loss is attained, after which the groups are weighed at least once a week until the end of the trial.
  • tumours are measured about 2 or 3 times a week until the tumour reaches a pre- determined size and / or weight, or until the animal dies if this occurs before the tumour reaches the pre-determined size / weight.
  • the animals are then sacrificed and the tissue histology, size and / or proliferation of the tumour assessed.
  • the animals are grafted with a particular number of cells, and the anti- tumour activity is determined by the increase in the survival time of the treated mice relative to the controls.
  • tumour cells are typically treated with the composition ex vivo and then injected into a suitable test animal. The spread of the tumour cells from the site of injection is then monitored over a suitable period of time by standard techniques.
  • In vivo toxic effects of the oligonucleotides can be evaluated by measuring their effect on animal body weight during treatment and by performing haematological profiles and liver enzyme analysis after the animal has been sacrificed.
  • the antisense oligonucleotide may be administered as a pharmaceutical composition comprising the antisense oligonucleotide in admixture with an appropriate pharmaceutically physiologically acceptable carrier, diluent, excipient or vehicle.
  • the pharmaceutical compositions may also be formulated to contain the antisense oligonucleotide and one or more other chemotherapeutic agents for concurrent administration to a patient, where appropriate.
  • compositions of the present invention may be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intrasternal, intrathecal injection or infusion techniques.
  • compositions may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions and may contain one or more agents selected from the group of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with suitable non- toxic pharmaceutically acceptable excipients including, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch, or alginic acid; binding agents, such as starch, gelatine or acacia, and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • suitable non- toxic pharmaceutically acceptable excipients including, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch, or alginic acid; binding agents, such as starch, gelatine or acacia, and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated,
  • compositions for oral use may also be presented as hard gelatine capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatine capsules wherein the active ingredient is mixed with water or an oil medium such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • an oil medium such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active compound in admixture with suitable excipients including, for example, suspending agents, such as sodium carboxymethylcellulose, methyl cellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethyene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, hepta-decaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol for example, polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene sorbitan monooleate.
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or H-propyl -hydroxy- benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or H-propyl -hydroxy- benzoate
  • colouring agents for example ethyl, or H-propyl -hydroxy- benzoate
  • flavouring agents for example sucrose or saccharin.
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and/or flavouring agents may be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti- oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active compound in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent, suspending agent and one or more preservatives are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present.
  • compositions of the invention may also be in the form of oil-in- water emulsions.
  • the oil phase may be a vegetable oil, for example, olive oil or arachis oil, or a mineral oil, for example, liquid paraffin, or it may be a mixtures of these oils.
  • Suitable emulsifying agents may be naturally-occurring gums, for example, gum acacia or gum tragacanth; naturally-occurring phosphatides, for example, soy bean, lecithin; or esters or partial esters derived from fatty acids and hexitol, anhydrides, for example, sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monoleate.
  • the emulsions may also contain sweetening and flavouring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, and/or flavouring and colouring agents.
  • sweetening agents for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, and/or flavouring and colouring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to known art using suitable dispersing or wetting agents and suspending agents such as those mentioned above.
  • the sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol, water, Ringer's solution, lactated Ringer's solution or isotonic sodium chloride solution.
  • acceptable vehicles and solvents include, but are not limited to, sterile, fixed oils which are conventionally employed as a solvent or suspending medium, and a variety of bland fixed oils including, for example, synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions are also suitable for administration by continuous infusion.
  • the antisense oligonucleotide is formulated as an injectable composition.
  • Other pharmaceutical compositions and methods of preparing pharmaceutical compositions are known in the art and are described, for example, in "Remington: Zhe Science and Practice of Pharmacy,” Gennaro, A., Lippincott, Williams & Wilkins, Philidelphia, PA (2000) (formerly “Remingtons Pharmaceutical Sciences”).
  • the antisense oligonucleotides of the present invention and combinations comprising an antisense oligonucleotide and one or more chemotherapeutic agents can be used in the treatment of a variety of cancers.
  • the combination is more effective in reducing the growth and/or metastasis of cancer cells than the chemotherapeutic agent(s) alone.
  • the antisense oligonucleotides and combinations can also be used to effectively treat drug resistant tumours.
  • Carcinomas, adenocarcinomas and sarcomas are also frequently referred to as "solid tumours," examples of commonly occurring solid tumours include, but are not limited to, cancer of the brain, breast, cervix, colon, ' head and neck, kidney, lung, ovary, pancreas, prostate, lung, stomach and uterus, and colorectal cancer. Lymphomas are also considered to be solid tumours.
  • leukaemia refers broadly to progressive, malignant diseases of the blood- forming organs. Leukaemia is typically characterised by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow but can also refer to malignant diseases of other blood cells such as erythroleukaemia, which affects immature red blood cells. Leukaemia is generally clinically classified on the basis of (1) the duration and character of the disease - acute or chronic; (2) the type of cell involved - myeloid (myelogenous), lymphoid (lymphogenous) or monocytic, and (3) the increase or non-increase in the number of abnormal cells in the blood - leukaemic or aleukaemic (subleukaemic).
  • Leukaemia includes, for example, acute nonlymphocytic leukaemia, chronic lymphocytic leukaemia, acute granulocytic leukaemia, chronic granulocytic leukaemia, acute promyelocytic leukaemia, acute myeloid leukaemia (AML), chronic myeloid leukaemia (CML), adult T-cell leukaemia, aleukaemic leukaemia, aleukocythemic leukaemia, basophylic leukaemia, blast cell leukaemia, bovine leukaemia, chronic myelocytic leukaemia, leukaemia cutis, embryonal leukaemia, eosinophilic leukaemia, Gross' leukaemia, hairy-cell leukaemia, hemoblastic leukaemia, hemocytoblastic leukaemia, histiocytic leukaemia, stem cell leukaemia, acute monocytic leukaemia, leukopenic leukaemia
  • carcinoma refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases.
  • exemplary carcinomas include, for example, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colorectal carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid carcinoma, epiermoid carcinoma, carcinoma epitheliale adenoides, exophytic carcinoma, carcinoma ex
  • carcinomas that may be treated with the antisense oligonucleotides of the present invention, include, for example, pancreatic, ovarian, lung, liver, renal and cervical carcinomas.
  • carcinomas that originate in cells that make organs which have glandular (secretory) properties or that originate in cells that line hollow viscera, such as the gastrointestinal tract or bronchial epithelia. Examples include, but are not limited to, adenocarcinomas of the breast, lung, pancreas, colon and prostate.
  • tumour generally refers to a tumour which originates in connective tissue, such as muscle, bone, cartilage or fat, and is made up of a substance like embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance.
  • Sarcomas include soft tissue sarcomas, chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abemethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumour sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcoma, fibroblastic sarcoma, giant cell sarcoma, granulocytic sarcoma, Hodgkin's sarcoma, idiopathic multiple pigmented haemorrhagic
  • melanoma is taken to mean a tumour arising from the melanocytic system of the skin and other organs.
  • Melanomas include, for example, acral-lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma, Harding-Passey melanoma, juvenile melanoma, lentigo maligna melanoma, malignant melanoma, nodular melanoma, subungal melanoma, and superficial spreading melanoma.
  • the antisense oligonucleotides of the present invention can also be used in the treatment of lymphomas including Hodgkin's and non-Hodgkin's lymphomas and brain cancers including primary brain tumours, gliomas, glioblastoma multiforme; malignant astrocytomas; oligdendroglioma; ependymoma; low-grade astrocytomas; meningioma; mesenchymal tumours; pituitary tumours; nerve sheath tumours such as schwannomas; central nervous system lymphoma; medulloblastoma; primitive neuroectodermal tumours; neuron and neuron/glial tumours; craniopharyngioma; germ cell tumours and choroid plexus tumours.
  • lymphomas including Hodgkin's and non-Hodgkin's lymphomas and brain cancers including primary brain tumours, gliomas, glioblastoma multiforme
  • Additional cancers include multiple myeloma, neuroblastoma, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, small-cell lung tumours, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, thyroid cancer, oesophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer and mesothelioma.
  • the cancer may be indolent or it may be aggressive.
  • the antisense oligonucleotides are useful in the treatment of refractory cancers, advanced cancers, recurrent cancers, relapsed and metastatic cancers.
  • refractory cancers advanced cancers
  • recurrent cancers recurrent cancers
  • metastatic cancers One skilled in the art will appreciate that many of these categories may overlap, for example, aggressive cancers are typically also advanced and/or metastatic.
  • Aggressive cancer refers to a rapidly growing cancer.
  • aggressive cancer will refer to an advanced cancer that has relapsed within approximately the earlier two-thirds of the spectrum of relapse times for a given cancer, whereas for other types of cancer, such as small cell lung carcinoma (SCLC) nearly all cases present rapidly growing cancers which are considered to be aggressive.
  • SCLC small cell lung carcinoma
  • the term can thus cover a subsection of a certain cancer type or it may encompass all of other cancer types.
  • a “refractory” cancer or tumour refers to a cancer or tumour that has not responded to treatment.
  • Advanced cancer refers to overt disease in a patient, wherein such overt disease is not amenable to cure by local modalities of treatment, such as surgery or radiotherapy.
  • Advanced disease may refer to a locally advanced cancer or it may refer to metastatic cancer.
  • metastatic cancer refers to cancer that has spread from one part of the body to another.
  • Advanced cancers may also be unresectable, that is, they have spread to surrounding tissue and cannot be surgically removed.
  • the antisense oligonucleotides may also be used to treat drug resistant cancers, including multidrug resistant tumours.
  • drug resistant cancers including multidrug resistant tumours.
  • the resistance of cancer cells to chemotherapy is one of the central problems in the management of cancer.
  • Certain cancers such as prostate and breast cancer, can be treated by hormone therapy, i.e. with hormones or anti-hormone drugs that slow or stop the growth of certain cancers by blocking the body's natural hormones. Such cancers may develop resistance, or be intrinsically resistant, to hormone therapy.
  • the present invention further contemplates the use of the antisense oligonucleotide in the treatment of these "hormone-resistant " or "hormone-refractory” cancers.
  • the antisense oligonucleotide alone, or in combination with one or more chemotherapeutic is used in the treatment of solid tumours including metastatic, advanced, drug- or hormone-resistant versions of solid tumours.
  • the solid tumour is a renal tumour, breast tumour, lung tumour, prostate tumour, colon tumour, melanoma, ovarian tumour, cervical tumour, brain tumour, liver tumour, colorectal tumour, pancreatic tumour, genitourinary tumour, gall bladder tumour, head and neck tumour, oesophageal tumour biliary duct tumour, a lymphoma, or a sarcoma, including a metastatic, advanced, drug- or hormone-resistant version thereof.
  • the solid tumour is an ovarian tumour, a renal tumour, a brain tumour, or a sarcoma, including a metastatic, advanced, or drug-resistant version thereof.
  • the antisense oligonucleotide alone, or in combination with one or more chemotherapeutic is used in the treatment of a leukaemia, including a metastatic, advanced or drug-resistant version thereof.
  • the dose of the antisense oligonucleotide of the present invention to be administered to a patient should be a sufficient amount to effect a beneficial therapeutic response in the patient over time, i.e. an "effective amount.”
  • a beneficial therapeutic response may be, for example, stabilisation of the disease, tumour shrinkage, decreased time to progression or prolonged survival.
  • the dose will be determined by the efficacy of the particular oligonucleotide employed, the type of cancer to be treated and the condition of the patient to be treated, as well as the body weight or surface area of the patient. Appropriate doses can be readily determined by a skilled practitioner.
  • antisense oligonucleotides are administered systemically to patients. Administration can be accomplished by bolus injection as a single dose or as divided doses, or by continuous infusion over an appropriate period of time.
  • the antisense oligonucleotides are administered by continuous infusion. In another embodiment, the antisense oligonucleotides are administered by continuous intravenous infusion.
  • the dosage of the antisense oligonucleotide to be administered will be dependent upon the type of cancer to be treated and the size of the patient and can be readily determined by a skilled practitioner.
  • appropriate doses determined by Phase I clinical trials are between about 18.5 mg/m 2 /day and about 222 mg/m 2 /day.
  • the dose of antisense oligonucleotide is between about 37 mg/m 2 /day and about 222 mg/m 2 /day.
  • the dose of antisense oligonucleotide is between about 74 mg/m 2 /day and about 185 mg/m 2 /day. In further embodiments, the dose of antisense oligonucleotide is between about 100 mg/m 2 /day and about 185 mg/m 2 /day and between about 148 mg/m 2 /day and about 185 mg/m 2 /day.
  • Other exemplary doses for SEQ ID NO:l include doses between about 2 mg/kg/day and about 10 mg/kg/day, between about 3 mg/kg/day and about 8 mg/kg/day and between about 3 mg/kg/day and about 5 mg/kg/day.
  • Treatment regimens can be designed such that the antisense oligonucleotide is administered to the patient in cycles.
  • Treatment with antisense oligonucleotide in accordance with the present invention may be part of a treatment regimen that involves one cycle of administration or more than one cycle.
  • a cycle is between about 1 and about 4 weeks.
  • Exemplary dosing schedules comprise one or more cycle of 21 days continuous infusion followed by 7 days of rest or one or more cycles of 14 days continuous infusion followed by 7 days of rest. Further examples are provided in the Examples section herein.
  • Other treatment regimens can be readily determined by the skilled practitioner. Between one and sixteen cycles of treatment are contemplated, however, additional cycles may be incorporated into the treatment regimen as necessary.
  • the present invention contemplates the use of the antisense oligonucleotides, alone or in combination with one or more other chemotherapeutic agents, to treat patients who have undergone prior chemotherapy.
  • the antisense oligonucleotides are used as a second or subsequent (for example, third or fourth) line of therapy.
  • the antisense oligonucleotides are used to treat patients who have already undergone more than one course of prior chemotherapy.
  • the antisense oligonucleotides, alone or in combination with one or more other chemotherapeutic agents may also be used as a first line of therapy in the treatment of patients for whom standard chemotherapy is not suitable.
  • the antisense oligonucleotide can be administered to the patient in conjunction with one or more chemotherapeutic agents.
  • the antisense oligonucleotide can be administered prior to, or after, administration of the one or more other chemotherapeutic agents, or it can be administered concurrently.
  • the one or more chemotherapeutic may be administered systemically, for example, by bolus injection or continuous infusion, or it may be administered orally.
  • the one or more other chemotherapeutic may also be administered in cycles, which may or may not overlap with the cycles of admimstration for the antisense oligonucleotide.
  • the length of time between the initiation of administration of the antisense oligonucleotide and the other agent(s) will depend on the mode of administration, the size of the patient and the nature of the other agent(s) being administered.
  • administering may be initiated at the same time, or administration of the other chemotherapeutic(s) may be initiated at a suitable time prior to or after administration of the antisense oligonucleotide is initiated.
  • Appropriate treatment regimens can be readily determined by the skilled practitioner.
  • Capecitabine can be administered at a dose of between about 500 and about 2000 mg/m 2 /day.
  • Capecitabine is typically administered orally. Administration of the daily amount may be via a single dose or divided doses. Exemplary doses would be between about 500 - 1500 mg/m /day, between about 600 - 1000 mg/m 2 /day, and between about 1100 -2000 mg/m 2 /day depending on the type of cancer being treated.
  • capecitabine at a dose of between 850 and 1700 mg/m 2 /day is used in conjunction with the antisense oligonucleotide.
  • doses of 850, 1250 and 1660 mg/m 2 /day are used.
  • Cytarabine can be administered at various doses between about 5 and about 3000 mg/m 2 /day depending on the type of cancer being treated and the dosing schedule employed. Administration of the daily amount of cytarabine may be via a single dose, divided dose or continuous infusion. Exemplary doses would be between about 500 - 1000 mg/m 2 /day, between about 1000 - 2000 mg/m 2 /day and between about 4000 - 6000 mg/m 2 /day. In one embodiment, cytarabine at a dose of between about between about 4000 - 6000 mg/m 2 /day is used in conjunction with the antisense oligonucleotide. ⁇
  • cytarabine can be administered intrathecally at a dose of between about 5 - 75 mg/m 2 /day and between about 100 - 200 mg/m 2 /day, depending on the type of cancer being treated and the dosing schedule employed.
  • cytarabine is used at a dose of between about 5 - 75 mg/m 2 /day in conjunction with the antisense oligonucleotide.
  • Docetaxel can be administered at a dose of between about 20 and about 100 mg/m 2 per one dose. Exemplary doses would be between about 30 - 35 mg/m 2 , between
  • docetaxel at a dose of between about 60 mg/m 2 and about 75 mg/m 2 is used in conjunction with the antisense oligonucleotide.
  • Single dose units of gemcitabine are typically between about 100 and about 2500 mg/m 2 .
  • Exemplary dose units suitable for use with the antisense oligonucleotides would be between about 400 - 1000 mg/m 2 , between about 600 - 1000 mg/m 2 , between about 800 - 1000 mg/m 2 , between about 500 - 1250 mg/m 2 , between about 750 - 1200 mg/m 2 , between about 800 - 1250 mg/m 2 , between about 1000 - 1200 mg/m 2 , between about 1250 - 2500 mg/m 2 , depending on the type of cancer being treated and the dosing schedule employed.
  • the dose maybe administered, for example, weekly or biweekly. In one embodiment, a weekly unit dose of between about 400 - 1000 mg/m 2 gemcitabine is used in conjunction with the antisense oligonucleotide.
  • gemcitabine can also be administered at lower doses, for example, between about 100 to about 400 mg/m 2 /day depending on the type of cancer being treated.
  • Oxaliplatin can be administered at a dose of between about 30 and about 135 mg/m 2 /day. Administration of the daily amount of oxaliplatin may be via a single dose or divided doses, or by continuous infusion. Exemplary doses would be between about 80 - 100 mg/m 2 /day and between about 85 - 135 mg/m 2 /day depending on the type of cancer being treated and the dosing schedule employed. In one embodiment, oxaliplatin at a dose of about 130 mg/m 2 /day is used in conjunction with the antisense oligonucleotide.
  • Phase I trials are used to determine the best mode of administration (for example, by pill or by injection), the frequency of administration, and the toxicity for the compounds.
  • Phase I studies frequently include laboratory tests, such as blood tests and biopsies, to evaluate the effects of a compound in the body of the patient.
  • a Phase I trial a small group of cancer patients are treated with a specific dose of the antisense oligonucleotide and the one or more chemotherapeutic agent(s).
  • the dose is typically increased group by group in order to determine the maximum tolerated dose (MTD) and the dose-limiting toxicities (DLT) associated with the compound. This process determines an appropriate dose to use in a subsequent Phase II trial.
  • MTD maximum tolerated dose
  • DLT dose-limiting toxicities
  • a Phase II trial can be conducted to evaluate further the effectiveness and safety of the antisense oligonucleotides alone or combinations.
  • the antisense oligonucleotides alone or the combination is administered to groups of patients with either one specific type of cancer or with related cancers, using the dosage found to be effective in Phase I trials.
  • Phase III trials focus on determining how a compound compares to the standard, or most widely accepted, treatment.
  • patients are randomly assigned to one of two or more "arms".
  • one arm will receive the standard treatment (control group) and the other arm will receive treatment with the antisense oligonucleotide or combination of the present invention (investigational group).
  • Phase IV trials are used to further evaluate the long-term safety and effectiveness of a compound. Phase IV trials are less common than Phase I, II and III trials and will take place after the antisense oligonucleotide or combination has been approved for standard use.
  • Participant eligibility criteria can range from general (for example, age, sex, type of cancer) to specific (for example, type and number of prior treatments, tumour characteristics, blood cell counts, organ function). Eligibility criteria may also vary with trial phase. For example, in Phase I and II trials, the criteria often exclude patients who may be at risk from the investigational treatment because of abnormal organ function or other factors. In Phase II and III trials additional criteria are often included regarding disease type and stage, and number and type of prior treatments. Phase I cancer trials usually comprise 15 to 30 participants for whom other treatment options have not been effective. Phase II trials typically comprise up to 100 participants who have already received chemotherapy, surgery, or radiation treatment, but for whom the treatment has not been effective. Participation in Phase II trials is often restricted based on the previous treatment received.
  • Phase III trials usually comprise hundreds to thousands of participants. This large number of participants is necessary in order to determine whether there are true differences between the effectiveness of the antisense oligonucleotides or combination of the present invention and the standard treatment. Phase III may comprise patients ranging from those newly diagnosed with cancer to those with extensive disease in order to cover the disease continuum.
  • clinical trials should be designed to be as inclusive as possible without making the study population too diverse to determine whether the treatment might be as effective on a more narrowly defined population.
  • the more diverse the population included in the trial the more applicable the results could be to the general population, particularly in Phase III trials. Selection of appropriate participants in each phase of clinical trial is considered to be within the ordinary skills of a worker in the art.
  • Patients Prior to commencement of the study, several measures known in the art can be used to first classify the patients. Patients can first be assessed, for example, using the Eastern Cooperative Oncology Group (ECOG) Performance Status (PS) scale or the Karnofsky Performance Status (KPS) scale, both of which are widely accepted standards for the assessment of the progression of a patient's disease as measured by functional impairment in the patient.
  • ECOG Eastern Cooperative Oncology Group
  • PS Performance Status
  • KPS Karnofsky Performance Status
  • MQOL McGill Quality of Life Questionnaire
  • SDS Symptom Distress Scale
  • Patients can also be classified according to the type and/or stage of their disease and/or by tumour size.
  • the antisense oligonucleotide and the one or more chemotherapeutuc agent(s) are typically administered to the trial participants parenterally.
  • the antisense oligonucleotide or combination is administered by intravenous infusion.
  • Methods of administering drugs by intravenous infusion are known in the art. Usually intravenous infusion takes place over a certain time period, for example, over the course of 60 minutes.
  • the endpoint of a clinical trial is a measurable outcome that indicates the effectiveness of a treatment under evaluation.
  • the endpoint is established prior to the commencement of the trial and will vary depending on the type and phase of the clinical trial.
  • Examples of endpoints include, for example, tumour response rate - the proportion of trial participants whose tumour was reduced in size by a specific amount, usually described as a percentage; disease-free survival - the amount of time a participant survives without cancer occurring or recurring, usually measured in months; overall survival - the amount of time a participant lives, typically measured from the beginning of the clinical trial until the time of death.
  • disease stabilisation the proportion of trial participants whose disease has stabilised, for example, whose tumour(s) has ceased to grow and/or ' metastasise, can be used as an endpoint.
  • Other endpoints include toxicity and quality of life.
  • Tumour response rate is a typical endpoint in Phase II trials. However, even if a treatment reduces the size of a participant's tumour and lengthens the period of disease-free survival, it may not lengthen overall survival. In such a case, side effects and failure to extend overall survival might outweigh the benefit of longer disease- * free survival. Alternatively, the participant's improved quality of life during the tumour-free interval might outweigh other factors. Thus, because tumour response rates are often temporary and may not translate into long-term survival benefits for the participant, response rate is a reasonable measure of a treatment's effectiveness in a Phase II trial, whereas participant survival and quality of life are typically used as endpoints in a Phase III trial.
  • kits containing the antisense oligonucleotide alone or in combination with one or more chemotherapeutic agents.
  • the kits can include one or more chemotherapeutic agents in pharmaceutical compositions for use in the treatment of cancer.
  • Individual components of the kit would be packaged in separate containers and, associated with such containers, can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the liquid solution can be an aqueous solution, for example a sterile aqueous solution.
  • the container means may itself be an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the composition may be administered to a patient.
  • kits of the invention may also be provided in dried or lyophilised form and the kit can additionally contain a suitable solvent for reconstitution of the lyophilised components.
  • the kits of the invention also may comprise an instrument for assisting with the administration of the composition to a patient.
  • Such an instrument may be an inhalant, syringe, pipette, forceps, measured spoon, eye dropper or any such medically approved delivery vehicle.
  • SEQ ID NO:l as referred to throughout the Examples is a fully phosphorothibated oligonucleotide with the sequence: 5'-GGCTAAATCGCTCCACCAAG-3' [SEQ ID NO:l] SEQ ID NO : 1 hybridizes to the coding region of R2 mRNA.
  • SEQ ID NO:2 is a mismatched control analogue of SEQ ID NO: 1, having four base changes in the middle of the sequence: 5'-GGCTAAACTCGTCCACCAAG-3' [SEQ ID NO:2]
  • SEQ ID NO:3 is a scrambled control analogue of SEQ ID NO: 1 that is not complementary to R2, but retains the same base composition ratio as SEQ ID NO: 1. 5'-ACGCACTCAGCTAGTGACAC-3' [SEQ ID NO:3]
  • the phosphorothioates were synthesized on an automated DNA synthesizer (Perkin- Elmer, USA) by Boston BioSystem Inc. (Boston, MA) and were purified by reversed- phase high performance liquid chromatography.
  • EXAMPLE 1 In vivo Testing of SEQ ID NO: 1 in Combination with Various Chemotherapeutics in Mouse Xenograft Models 1.1 HT-29 human colon cancer cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female CD-I nude mice. After the size of tumour reached an approximate volume of 50 mm 3 , 4 days post tumour cell injection, mitomycin C was administered by bolus infusion into the tail vein at days 4, 11 and 18 with a dose of 3.5 mg/kg/week. Antitumour effect of mitomycin C was further compared to that of SEQ ID NO:l in combination with mitomycin C.
  • SEQ ID NO: 1 was administered by bolus infusion into the tail vein every day at 6 mg/kg and mitomycin C was administered intravenously at days 4, 11 and 18 with a dose of 3.5 mg/kg/week, one hour after the treatments with SEQ ID NO: 1.
  • Control animals received saline alone for the same period as SEQ ID NO:l. All treatments were stopped at day 22. A day after the last treatment, tumours were excised from the animals and their weights were measured. A standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean'tumour weight calculated from 5 animals.
  • mitomycin C treatments resulted in significant delay of tumour growth compared to saline control.
  • the antitumour effects elicited by the combination of SEQ ID NO:l and mitomycin C were more potent than those obtained using mitomycin C alone (see Figure 1).
  • 1.2 HT-29 human colon cancer cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 5-6 week old female CD-I nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 7 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO:l was further compared to that of CPT- 11 alone or that of SEQ ID NO: 1 in combination with
  • CPT-11 was administered intraperitoneally for 5 days in a row from day 7-12 with a dose of 20mg/kg in 100 ⁇ l saline. All treatments were stopped at day 32. A day after the last treatment, tumours were excised from the animals and their weights were measured. A standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean tumour weight calculated from 9 animals.
  • SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control. The delay in tumour growth achieved with SEQ ID NO:l was superior to the inhibitory effects observed with CPT-11 alone.
  • the combination treatments of SEQ ID NO:l and CPT-11 showed excellent cooperative effects that were more potent than either agent alone (see Figure 2).
  • HT-29 human colon cancer cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old female CD-I nude mice. Treatments started at day 5 post tumour cell injection, SEQ ID NO:l and 5- FU were administered as outlined below: Groups and treatment:
  • Saline treated group saline: 0.1 ml/2 days, i. v.; Group 2.
  • SEQ ID NO:l treated group 10 mg/kg/2 days in 100 ⁇ L saline, i. v.; Group 3.
  • 5-FU treated group 13 mg/kg/5 days/week (one week on and one week off), i. v.; Group 4.
  • HT-29 human colon cancer cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 5-6 week old female CD-I nude mice. Treatments started 4 days post tumour cell injection, SEQ ID NO: 1 and
  • Group 1 treated with 0.2 ml vehicle solution for capecitabine, o.p.;
  • Group 2 treated with 0.1 ml saline, i.v.;
  • Group 3 treated with 359 mg/kg/day x 5 /w Capecitabine in 0.2 ml vehicle solution, o.p.;
  • Group 4 treated with 10 mg/kg/2days SEQ ID NO:l in 0.1 ml saline i.v.;
  • Group 5 treated with 10 mg/kg/2days SEQ ID NO:l in 0.1 ml saline, i.v. plus
  • the antitumour effect of SEQ ID NO:l was further compared to that of two chemotherapeutic agents: 5-FU and vinblastin.
  • 5-FU was administered intraperitoneally at days 7-13, 21-27 and 35-36 with a dose of 13 mg/kg/day
  • vinblastin was administered intraperitoneally at days 7, 14, 21, 28 and 35 at a dose of 0.6mg/kg/week.
  • Antitumour effects of each of these compounds were further compared to those of SEQ ID NO: 1 in combination with 5-FU or with vinblastin.
  • the two chemotherapeutic agents were applied as described above, one hour after the treatments with SEQ ID NO:l when combination treatments occurred on the same day. All treatments were stopped at day 36.
  • tumours were excised from the animals and their weights were measured.
  • a standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean tumour weight calculated from 5 animals.
  • SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control.
  • the delay in tumour growth achieved with SEQ ID NO: 1 was superior to the inhibitory effects observed with each of two chemotherapeutic compounds.
  • the combination of SEQ ID NO:l with 5-FU or vinblastine was more effective than either agent alone (see Figure 5 A).
  • mice After 52 days the mice were sacrificed and the tumours weighed. The results are shown in Figure 5B. Each bar represents the mean tumour weight and standard error calculated for each treatment group.
  • FIG. 6 shows results from two independent experiments.
  • PC-3 human prostatic cancer cells (1X10 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old male SCID mice. After the size of tumour reached an approximate volume of 50 mm , 14 days post tumour cell injection, SEQ ID NO: 1 was administered by bolus infusion into the tail vein every other day at 10 mg/kg 18 times ( Figure 6 A) or 17 times ( Figure 6B), respectively. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was further compared to that of mitoxantrone (novantrone ® ) alone or in combination.
  • mitoxantrone novantrone ®
  • Mitoxantrone was administered intravenously once at the beginning of the treatments at a dose of 2 mg/kg ( Figure 6A) or once a week for four weeks at a reduced dose of 0.8 mg/kg (Figure 6B). All treatments were stopped at day 50 ( Figure 6A) or 48 ( Figure 6B), respectively. A day after the last treatment, tumours were excised from the animals and their weights were measured. A standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean tumour weight calculated from 5 ( Figure 6 A) or 10 ( Figure 6B) animals. As illustrated in Figure 6 A, SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control.
  • FIG. 7 shows results from two independent experiments.
  • DU145 human prostatic cancer cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old male SCID mice.
  • SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg 15 times ( Figure 7 A) or 14 times ( Figure 7B), respectively.
  • Control animals received saline alone for the same period.
  • Antitumour effect of SEQ ID NO: 1 was further compared to that of mitoxantrone (novantrone ® ) alone or in combination.
  • Mitoxantrone was administered intravenously once at the beginning of the treatments at a dose of 2 mg/kg ( Figure 7A) or once a week for four weeks at a reduced dose of 0.8 mg/kg ( Figure 7B). All treatments were stopped at day 42
  • FIG. 7 A Figure 7 A or 38 ( Figure 7B), respectively.
  • a day after the last treatment tumours were excised from the animals and their weights were measured.
  • a standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean tumour weight calculated from 5 ( Figure 7A) or 10 ( Figure 7B) animals.
  • SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control.
  • the delay in tumour growth achieved with SEQ ID NO: 1 was similar to the inhibitory effects observed with mitoxantrone alone.
  • the combination of SEQ ID NO:l with mitoxantrone SEQ ID NO: 1 +
  • mitoxantrone alone resulted in significant delay of tumour growth and the combination therapy was more potent than mitoxantrone monotherapy.
  • A2058 human melanoma cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old female CD-I nude mice.
  • A2058 is a metastatic melanoma cell line. After the size of tumour reached an approximate volume of 100 mm , 6 days post tumour cell injection, SEQ ID NO: 1
  • SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was further compared to that of dacarbazine (DTIC) alone or that of SEQ ID NO:l in combination with DTIC. DTIC was administered intravenously for 5 days in a row from day 6-10 at a dose of 80mg/kg in 100 ⁇ l saline. All treatments were stopped at day 24. A day after the last treatment, tumours were excised from the animals and their weights were measured. A standard bar graph was used to demonstrate the differences in tumour weights with each bar representing mean tumour weight calculated from 10 animals. As illustrated, SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control. The delay in tumour growth achieved with
  • SEQ ID NO:l was superior to the inhibitory effects observed with DTIC alone.
  • the combination of SEQ ID NO: 1 and DTIC was more potent than either agent alone ( Figure 8).
  • FIG. 9 shows results from three independent experiments.
  • MDA-MB-231 human breast cancer cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female CD-I nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 5 days post tumour cell injection, SEQ ID NO:l, or the scrambled control oligonucleotide (SEQ ID NO: 3) were administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO:l was further compared to that of taxol or doxorubicin alone or in combination.
  • Taxol was administered intravenously once a week at a dose of 10 mg/kg for three ( Figure 9 A) or four weeks (Figure 9C).
  • Doxorubicin was administered intravenously once a week at a dose of 5 mg/kg for first three weeks ( Figure 9A) or for two weeks (Figure 9C). All treatments were stopped at day 33
  • FIG. 9B demonstrates that a control oligonucleotide that has the same base composition as SEQ ID NO:l, but is not complementary to R2 mRNA has no significant antitumour activity as a monotherapy and does not cooperate with doxorubicin, suggesting that the effects of SEQ ID NO: 1 are sequence specific.
  • 1.10SK.-OV-3 human ovary adenocarcinoma cells (1X10 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female CD-I nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 6 days post tumour cell injection, SEQ ID NO: 1 was administered by bolus infusion into the tail vein every other day at 10 mg/kg 17 times. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was further compared to that of taxol or cisplatin alone or in combination.
  • Taxol was administered intravenously once a week for first three weeks and intraperitoneally once a week for next two weeks at a dose of 10 mg/kg.
  • Cisplatin was administered intravenously once a week for first three weeks and intraperitoneally once a week for next two weeks at a dose of 4 mg/kg. All treatments were stopped at day 40. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper. Each point represents mean tumour volume calculated from 9 animals per experimental group.
  • SEQ ID NO: 1 treatments resulted in significant delay of tumour growth compared to saline control.
  • the delay in tumour growth achieved with SEQ ID NO:l was similar or superior to the inhibitory effects observed with taxol or cisplatin alone, repectively.
  • the combination therapy of SEQ ID NO: 1 with taxol or cisplatin was more potent than either monotherapy (Figure 10).
  • Results shown are mean tumour weights presented as a percentage of saline treated controls. ** is tumour volume data as percentage of saline control.
  • EXAMPLE 2 In vivo Testing of SEQ ID NO:l Alone or in Combination with Various Chemotherapeutics in Drug-Resistant Tumours
  • BxPC-3 human pancreatic carcinoma cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female CD-I nude mice.
  • BxPC-3 is a gemcitabine resistant call line.
  • SEQ ID NO: 1 was administered by bolus infusion into the tail vein every other day at 10 mg/kg 17 times.
  • Control animals received saline alone for the same period.
  • Antitumour effect of SEQ ID NO: 1 was further compared to that of Gemcitabine.
  • Gemcitabine was administered intravenously every three days at a dose of 100 mg/kg.
  • SEQ ID NO: 1 was administered by bolus infusion into the tail vein every other day at 10 mg/kg 6 times. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO:l was further compared to that of Hydroxyurea or Cisplatin alone or in combination. Hydroxyurea was administered intraperitoneally every day at a dose of 250 mg/kg for 10 days. Cisplatin was administered intravenously once a week for three weeks at a dose of 4 mg/kg. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper. Each point represents mean tumour volume calculated from 10 animals per experimental group.
  • SEQ ID NO:l treatments resulted in significant delay of tumour growth compared to saline control.
  • treatment with Hydroxyurea during the same period was ineffective against Hydroxyurea-resistant tumour.
  • the delay in tumour growth achieved with SEQ ID NO:l was superior to the inhibitory effects observed with Cisplatin alone, which was used as a positive control.
  • the combination therapy of SEQ ID NO:l with Hydroxyurea was only as effective as SEQ ID NO: 1 monotherapy, as expected.
  • Cisplatin-resistant breast adenocarcinoma cells 4X10 6 cells in 100 ⁇ l of PBS) were injected into the fat pad (inside of right leg) of 6-7 weeks old female SCID mice. After the size of tumour reached an approximate volume of 100 mm 3 , 7 days post tumour cell injection, SEQ ID NO: 1 was administered by bolus infusion into the tail vein every other day at 10 mg/kg 9 times. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was further compared to that of Cisplatin or Taxol alone.
  • Cisplatin was administered intravenously once a week for three weeks at a dose of 4 mg/kg.
  • Taxol was administered intravenously once a week for three weeks at a dose of 10 mg/kg.
  • Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper. Each point represents mean tumour volume calculated from 10 animals per experimental group.
  • SEQ ID NO: 1 treatments caused significant reduction of tumour weight compared to saline control.
  • treatment with Cisplatin during the same period was ineffective against Cisplatin-resistant tumour.
  • the delay in tumour growth achieved with SEQ ID NO: 1 was similar to the inhibitory effects observed with Taxol, which was used as a positive control ( Figure 13).
  • Cisplatin-resistant breast adenocarcinoma cells (4X10 6 cells in 100 ⁇ l of PBS) were injected into the fat pad (inside of right leg) of 6-7 weeks old female CB-17 SCID mice. After the size of tumour reached an approximate volume of 100 mm , 9 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was further compared to that of Taxol alone and in combination. Taxol was administered i.p. once a week at a dose of 10 mg/kg.
  • Taxol was administered intravenously once a week for four weeks at a dose of 20 mg/kg. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper. Each point represents mean tumour volume calculated from 9-10 animals per experimental group. As illustrated, SEQ ID NO:l treatments caused significant reduction of tumour weight compared to saline control. As expected, treatment with Taxol during the same period was ineffective against Taxol-resistant tumour. The delay in tumour growth achieved with SEQ ID NO:l was superior to the inhibitory effects observed with Cisplatin, which was used as a positive control (see Figure 15A & B).
  • MDA-MB435-To.l human Taxol-resistant breast adenocarcinoma cells (4X10 6 cells in 100 ⁇ l of PBS) were injected into the fat pad (inside of right leg) of 6-7 weeks old female CB-17 SCID mice. After the size of tumour reached an approximate volume of 100 mm , 17 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO: 1 was compared to that of Cisplatin alone and in combination. Cisplatin was administered intravenously once a week for four weeks at a dose of 4 mg/kg.
  • Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper. Each point represents mean tumour volume calculated from 10 animals per experimental group. At the end of the study the animals were sacrificed and tumours weighed. As illustrated, SEQ ID NO:l treatment caused significant reduction of tumour weight compared to saline control. The delay in tumour growth achieved with SEQ ID NO:l was superior to the inhibitory effects observed with Cisplatin, which was used as a positive control. The combination of the two compounds produced anti-tumour efficacy that was superior to either one alone (see Figure 16A & B).
  • Each point represents mean tumour volume calculated from 10 animals per experimental group. In addition animals were sacrificed and tumour weights taken at the end of the study. SEQ ID NO:l treatments caused significant reduction of tumour weight compared to saline control. As expected, treatment with taxol had no effect on tumour growth or weight (see Figure 17A & B).
  • 2.8 LS513 multi-drug resistant colon carcinoma cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female SCID mice. After the size of tumour reached an approximate volume of 100 mm 3 , 8 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Antitumour effect of SEQ ID NO:l was further compared to that of CPT-11 alone or in combination. CPT-11 was administered i.p. for 5 days at a dose of 20 mg/kg/day. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with caliper.
  • Each point represents mean tumour volume calculated from 10 animals per experimental group. Tumour weights were measured after animals were sacrificed at the end of the treatment. These cells are not resistant to CPT-11 which was used as a positive control. As illustrated, SEQ ID NO:l treatment resulted in significant delay of tumour growth compared to saline control. SEQ ID NO:l is as effective as CPT-11 and in combination the efficacy was greater than either treatment alone (see Figure 18A, B & C). Results of SEQ ID NO:l treatment of drug-resistant tumours alone or in combination with various chemotherapeutics are summarised in Table 4.
  • Results shown are mean tumour weights presented as a percentage of saline treated controls.
  • EXAMPLE 3 In vivo Testing of SEQ ID NO:l Alone in Mouse Xenograft Models
  • HT-29 human colon cancer cells (3X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old CD-I female nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 4 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Treatments lasted for 14 days thereafter.
  • SEQ ID NO: 1 Antitumour effects of SEQ ID NO: 1 were shown to be dose-dependent.
  • HT-29 human colon cancer cells (2X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old CD-I female nude mice. After the size of tumour reached an approximate volume of 100 mm , 5 days post tumour cell injection, increasing concentrations (0.4-6.0 mg/kg, designated as T0.4 to T6.0) of SEQ ID NO:l were administered by bolus infusion into the tail vein every other day for 14 days. Control animals (control) received saline alone for the same period. At the end of the treatments, the animals were sacrificed, tumours were excised and their weights were measured. Each bar represents mean tumour weight calculated from 6 animals per experimental group. As illustrated in Figure 19B, SEQ ID NO:l exerted statistically significant inhibitory effects on the growth of human colon adenocarcinoma in a dose-dependent manner.
  • 3.3 MDA-MB-231 human breast cancer cells (1X10 7 cells in 200 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old CD-I female nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 7 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Treatments lasted for 24 days thereafter.
  • Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper at two-day intervals over the treatment period. Each point represents mean tumour volume calculated from 4 animals per experimental group.
  • SEQ ID NO:l exhibited strong inhibitory effects on the growth of human breast carcinoma.
  • 3.4 SK-OV-3 human ovarian cancer cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 5-6 week old Balb/c female nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 11 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period.
  • Treatments lasted for 22 days thereafter. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper on average at two-day intervals over the span of 22 days. Each point represents mean tumour volume calculated from 5 animals per experimental group. As illustrated in Figure 20C, SEQ ID NO:l exhibited statistically significant inhibitory effects on the growth of human ovary adenocarcinoma cells.
  • NCI-H460 human lung cancer cells (5X10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 week old CD-I female nude mice. After the size of tumour reached an approximate volume of 100 mm 3 , 3 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. Treatments lasted for 14 days thereafter. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper on four different occasions over 16-day period. Each point represents mean tumour volume calculated from 5 animals per experimental group. As illustrated in Figure 20D, SEQ ID NO:l treatment demonstrated strong inhibitory effects on the growth of human lung carcinoma.
  • Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper at two-day intervals over the treatment period. Each point represents mean tumour volume calculated from 5 animals per experimental group. As illustrated in Figure 21, SEQ ID NO:l exhibited strong inhibitory effects on the growth of human pancreatic carcinoma and no batch- specific effects were evident.
  • Hep G2 human liver cancer cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 7 week old CD-I female nude mice. After the size of the tumour reached an approximate volume of 100 mm 3 , each tumour mass was recovered and divided into approximately the same size before each piece (ca. 25 mg) was implanted into a new mouse. After 9 days of growth, SEQ ID NO:l and scrambled control oligonucleotide (SEQ ID NO:3) were administered by bolus infusion into the tail vein every day at 2.5 mg/kg. Control animals received saline alone for the same period. Treatments lasted for 30 days thereafter.
  • Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper at various time intervals over the span of 30 days. Each point represents mean tumour volume calculated from 6 animals per experimental group. As illustrated, SEQ ID NO:l demonstrated significant inhibitory effects on the growth of human hepatoma cells, while tumour growth in mice treated with scrambled oligonucleotide did not differ from that in saline treated mice (see Figure 22).
  • A498 human kidney cancer cells (1X10 7 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-7 weeks old female SCID mice. After the size of tumour reached an approximate volume of 100 mm 3 , 25 days post tumour cell injection, SEQ ID NO:l was administered by bolus infusion into the tail vein every other day at 10 mg/kg. Control animals received saline alone for the same period. The antitumour effect of SEQ ID NO:l was further compared to that of three chemotherapeutic agents: 5-FU, vinblastin and gemcitabine.
  • 5-FU was administered intraperitoneally at days 26-32, and 39-46 at a dose of 13 mg/kg/day, while vinblastin and gemcitabine were administered intraperitoneally at day 26, 32, 39, 46 and 52 at a dose of 0.6mg/kg/week or 80 mg/kg/week, respectively. All treatments were stopped at day 52. Mice treated with saline or one of the three chemotherapeutic agents were sacrificed at day 80 because the tumour became too large. Mice treated with SEQ ID NO:l were kept for 16 days further to observe possible resurgence of resistant tumour growth. Antitumour activities were estimated by the inhibition of tumour volume, which was measured with a caliper. Each point represents mean tumour volume calculated from 5 animals per experimental group. As illustrated in Figure 23B, SEQ ID NO: 1 exhibited powerful inhibitory effects on the growth of human renal carcinoma cells, resulting in total regression of 'the tumours.
  • EXAMPLE 4 In vivo Testing of SEQ ID NO: 1 in a Mouse Experimental Model of Metastasis
  • EXAMPLE 5 Prolonged Survival of SCID Mice Bearing Human Burkitt's Lymphoma
  • lymphomaNiable human Burkitt's lymphoma (Raji) cells (5 X 10 6 ) collected from subconfluent logarithmically growing cultures were injected i.v. via the tail vein of each animal and disease was allowed to establish for 2 days.
  • SEQ ID NO: 1 in normal saline was administered by tail vein injections every second day at a dose of 10 mg/kg. Control animals received saline alone, without oligonucleotide.
  • Treatment with SEQ ID NO:l was stopped at day 42.
  • Mice with established human Burkitt's lymphoma tumours were treated every second day with either saline or SEQ ID NO:l for 44 days as described above.
  • the control group mice all died from disease progression by day 20.
  • the SEQ ID NO: 1 mice continued treatment on a weekly basis until stopping the treatment at day 73. All mice survived to the end of the experimental period (140 days).
  • tumour volume in C3H mice by Growth Fibrosarcoma 1 approximately 80% on Day 15 after tumour implantation
  • EXAMPLE 7 In vivo Anti-tumour activity of SEQ ID NO:l
  • human tumour cell lines were purchased from the American Type Culture Collections (ATCC) (Rockville, MD).
  • FCS fetal calf serum
  • WI-38 human lung fibroblast
  • HUVEC human umbilical vein endothelial cells
  • murine R3 fibrosarcoma
  • L Ltk " fibroblast
  • C8161 metastatic melanoma cells were obtained from Dr. D.R. Welch, University of Pennsylvania, Hershey, PA, and were maintained as above (Welch, et al, IntJ Cancer 47:227-37 (1991)). All media used in these experiments contain an antibiotic- antimycotic solution at a final concentration of 100 units/ml penicillin and 100 ⁇ g/ml streptomycin (Invitrogen Canada Inc. Burlington, Ontario).
  • CD-I athymic female nude mice BALB/c nu/nu nude mice, SCID, and SCID beige mice were purchased from Charles River Laboratories (Montreal, Canada) and experiments were typically initiated when the mice were 6-7 weeks old.
  • Human tumour cells were grown in appropriate growth medium and 3 X 10 -1 X 10 cells suspended in 100 ⁇ l of PBS were subcutaneously injected into the right flank of the animals with a 23 gauge needle (cell number indicated in the figure legends). Each experimental group typically contained 10 mice. After the size of tumour reached a mean tumour volume of 50-100 mm , treatment was initiated.
  • Antisense oligonucleotides (dissolved in saline) were administered by bolus infusion into the tail vein of animal every other day at the indicated dose.
  • Viable human Burkitt's lymphoma (Raji) cells (5 X 10 6 ) collected from subconfluent logarithmically growing cultures were injected intravenously into SCID mice, via the tail vein of each animal, and disease was allowed to establish for 2 days.
  • Antisense oligonucleotides in normal saline were administered by tail vein injections every second day at a dose of 10 mg/kg.
  • Control animals received saline alone, without antisense oligonucleotide or with mismatched and scrambled control antisense oligonucleotides.
  • Each treatment group typically contained 10 animals.
  • the antitumour efficacy of treatment was assessed by the examination of the survival of the mice. Survival is reported as a percentage of the starting number of mice in the treatment group.
  • SEQ ID NO:l displays sequence-specific and dose-dependant anti-tumour activity in vivo
  • R3 mouse fibrosarcoma cells (1.5 X 10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6-8 week old C3H female mice. After the size of tumour reached an approximate volume of 100 mm 3 , 3 days post tumour cell injection, SEQ ID NO:l, SEQ ID NO:2 and SEQ ID NO:3 were administered by bolus infusion into the tail vein every other day at 5 mg/kg. Control animals received saline alone for the same period. Treatments lasted for 14 days thereafter. Anti- tumour activities were estimated by the inhibition of tumour volume, which was measured with a calliper at two-day intervals starting from day 9. Each point in Figure 29A represents the mean tumour volume calculated from 6-7 animals per experimental group.
  • Figure 29B shows the mean weight of tumours excised from the above animals at the end of treatment.
  • R3 mouse fibrosarcoma cells (2 X 10 6 cells in 100 ⁇ l of PBS) were subcutaneously injected into the right flank of 6 week old C3H female mice. After the size of tumour reached an approximate volume of 100 mm , 4 days post tumour cell injection, increasing concentrations (0.5-30 mg/kg, designated as 0.5 to 30) of SEQ ID NO:l were administered by bolus infusion into the tail vein every other day for 10 days. Control animals received saline alone for the same period. At the end of the treatments, the animals were sacrificed, tumours were excised and their weights were measured. Each bar in Figure 29C represents mean tumour weight and standard error calculated from 6-8 animals per experimental group.
  • Figure 29C shows that SEQ ID NO:l demonstrates dose-dependent anti-tumour activity against both murine (R3).
  • FIG. 30 shows the results from xenograft experiments with Caki-1 human kidney cancer cells in which 5 X 10 6 cells in 100 ⁇ l of PBS were subcutaneously injected into the right flank of 6-7 weeks old female SCID mice. After the size of tumour reached an approximate volume of 100 mm , 7 days post tumour cell injection, SEQ ID NO:l, SEQ ID NO:2, or SEQ ID NO:3 were administered (10 mg/kg/2days, i.v.). Three additional treatment groups received: 5-FU (13mg/kg/day x 5), vinblastine (1 mg/kg/week) and gemcitabine (lOOmg/kg/week). Control animals received saline alone for the same period.
  • Figure 30C & D show results using A498 human kidney cancer cells.
  • 5 X 10 6 cells in 100 ⁇ l of PBS were subcutaneously injected into the right flank of 6-7 weeks old female SCID mice. After the size of tumour reached an approximate volume of 100 mm 3 , 19 days post tumour cell injection, SEQ ID NO:l, SEQ ID NO:2, or SEQ ID NO:3 were administered (10 mg/kg/2days, i.v.).
  • SEQ ID NO: 1 treatment can be seen to result in dramatic inhibition in growth of Caki-1 and A498 renal tumours as assessed by both tumour volume and tumour weight at the experimental end point. Treatment resulted in a rapid tumour stabilization and shrinkage within the first week of treatment. Tumour regression was complete within 3 weeks of treatment (day 28, post implantation of tumour). At the same dose, treatment with the control oligonucleotides resulted in tumour growth that was indistinguishable from saline treatment.
  • SEQ ID NO: 1 displays anti-tumour activity against a wide range of solid tumours in vivo.
  • Figure 31 summarizes the results of a statistical analysis of data compiled from a number of experiments in which the effect of SEQ ID NO: 1 on tumour growth was assessed in human tumour cell xenografts in mice. Each data point represents tumour weight and standard error. The experiments were carried out as follows: Tumour cell suspensions (1.5X10 to 10 ) were injected s.c. into the right flank of the mice for the tumour lines shown (except for hepatocellular carcinoma cells, which were subcutaneously implanted as a tumour mass).
  • tumour mass was palpable, usually ranged between 50 to 100 mm3.
  • Oligonucleotide concentrations used in treatments ranged from 2.5 mg/kg to lOmg/kg, administered i.v., every second day.
  • Tumour weight is expressed in mg and was obtained approximately 2 weeks after the start of oligonucleotide treatments, except for slow growing tumours such as hepatocellular carcinoma where tumour weight was obtained after 4 weeks of treatment. Differences in tumour weight between control and oligonucleotide treated groups were statistically significant in all cases (p ⁇ 0.05).
  • SEQ ID NO:l appears to be active against a range of cancer types. In all cancer cell lines tested, SEQ ID NO:l treatment resulted in a significant decrease in tumour growth (volume over time) and endpoint weight.
  • SEQ ID NO:l has superior anti-tumour efficacy compared to RNR-based therapeutic compounds.
  • SEQ ID NO:l In order to assess the clinical potential of SEQ ID NO:l, its efficacy against human tumour cell xenografts in mice was compared to that of 5-FU, gemcitabine and vinblastine (Figure 30). These compounds are currently in clinical use and are thought to act, at least partially, by decreasing RNR activity. Without exception SEQ ID NO: 1 was superior to any of these compounds against two human renal tumours, Caki-1 and A498 tumours ( Figure 30).
  • SEQ ID NO:l was the only compound that displayed long term protection from tumour growth In A498 tumour xenografts, 5-FU, gemcitabine and vinblastine treatments were all ineffective in stabilization of the tumours.
  • 5-FU a compound that displayed long term protection from tumour growth
  • gemcitabine a compound that displayed long term protection from tumour growth
  • vinblastine and gemcitabine displayed better efficacy than 5-FU there was, at best, a delay in the rate of tumour growth (Figure 30C).
  • SEQ ID NO:l treatment dramatically prolongs survival in xenograft model
  • SEQ ID NO:l was administered to SCID mice bearing active Burkitt's lymphoma ( Figure 32).
  • Viable human Burkitt's lymphoma (Raji) cells (5 X 10 6 ) collected from subconfluent logarithmically growing cultures were injected i.v. into each animal and disease was allowed to establish for 2 days.
  • SEQ ID NO: 1 , SEQ ID NO:3 and SEQ ID NO:2, in normal saline were administered by tail vein injection every second day at a dose of 10 mg/kg.
  • the treatment schedule was reduced to lOmg/kg every 3 days.
  • Control animals received saline alone, without oligonucleotide. Survival is presented as a percentage of the starting number of mice over time. All the SEQ ID NO :1 -treated mice survived to the end of the study and were sacrificed at day 72 due to animal housing limitations.
  • SEQ ID NO:l treatment leads to a dramatic increase in survival time of mice well beyond the treatment period (up to 72 days post-end of treatment). In addition to prolonged survival, the SEQ ID NO :1 -treated mice appeared to recover from the symptoms associated with the lymphoma. As treatment progressed the SEQ ID NO:l-
  • mice changed from having rough coats and weight loss to smooth coats and weight gain. Although strictly qualitative, these observations would suggest that the disease is not only stabilizing, but also regressing, consistent with the prolonged survival post-end of treatment.
  • SEQ ID NO:l treatment dramatically decreases lung nodule formation in an experimental metastasis model.
  • C8161 human metastatic melanoma cells were seeded into 100 mm tissue culture dishes at a density of 2 X 10 6 and incubated overnight at 37 °C in ⁇ -MEM medium supplemented with 10% FBS.
  • the cells were trypsinized, collected by centrifugation and aliquots were removed from the suspension to determine the cell viability using trypan blue exclusion test.
  • Approximately 1 X 10 5 cells suspended in 0.1 ml of PBS were injected into the tail veins of 6-8 week old female SCID mice. Treatment was initiated after 2 days.
  • Estimates of the number of lung nodules were made 5-7 weeks later, after excised lungs from individual mice were stained with picric acid dye solution (75 % picric acid, 20 % formaldehyde, 5 % glacial acetic acid). The results are shown in Figure 33, bars represent the mean number of nodules per mouse with standard error. In the SEQ ID NO:l treatment group only one of the 9 mice had lung nodules.
  • Immune stimulation can be the result of un-methylated CpG di- nucleotides, which stimulate innate immune responses in vertebrates and can further augment acquired immune responses to both pathogens and tumour cells.
  • the presence of un-methylated CpGs in an oligonucleotide can have the same effect if in an optimal sequence context.
  • CpG-mediated immune stimulation is not a major contributor to the anti-tumour efficacy of SEQ ID NO:l
  • immune stimulation in addition to sequence specific anti-tumour activity may not be a negative side effect in immune competent individuals.
  • mild immune stimulation may off-set immunodeficiency brought about by chemotherapy.
  • CpG dinucleotide motifs must be unmethylated in order to stimulate immune responses in vertebrates. Therefore, one approach to addressing the question of CpG mediated immune stimulation is to methylate the C residue in the CpG motif, thereby abrogating a CpG mediated immune response (Krieg, A. M. Annu. Rev. Immunol. 20:709-760 (2002)).
  • SEQ ID NO:l target sequence in R2 mRNA is absolutely conserved across human, rat and monkey, as the sequences were determined by direct sequencing of R2 cDNAs prepared by RT-PCR amplifications of mRNA extracted from rat and monkey cells.
  • SEQ ID NO: 1 The toxicokinetics and tissue distribution of SEQ ID NO: 1 (and related oligonucleotide metabolites obtained from exonuclease-mediated chain shortening) were determined in rats and monkeys.
  • the purpose of the studies was to correlate adverse effects in the animal studies to the concentrations of SEQ ID NO:l (and metabolites) in tissue and blood.
  • a capillary electrophoresis (CE) method was used to measure the concentrations of SEQ ID NO: 1 (and metabolites) in plasma and tissues.
  • mice were administered one or two bolus i.v. injections (tail vein) of SEQ ID NO:l at a dose of 50 mg/kg (295 mg/m 2 ) and intravenously infused (abdominal vena cava) for up to 48 h at a dose of 50 mg/kg/day (295 mg/m 2 /day).
  • Blood samples were collected into EDTA-containing tubes from subsets of animals in each group at various sampling times up to 56 h after initiation of infusion or bolus injection.
  • Parent SEQ ID NO:l concentrations were measured in plasma by a validated CE method.
  • SEQ ID NO:l was administered to monkeys by continuous intravenous infusion for 21 days followed by a 21 -day recovery period. Twenty-eight monkeys were administered one of the following dose levels; vehicle control, 2, 10, or 50 mg/kg/day (24.6, 123, and 615 mg/m 2 /day). Toxicokinetic samples were collected prior to the initiation of infusion, approximately 8, 24, 48, and 96 hours following the start of infusion, and on Day 20 prior to the change in dose-syringes.
  • Plasma concentrations of SEQ ID NO: 1 in the 2 mg/kg dose group could not be detected. Plasma concentrations appear to reach steady state by eight hours after dosing.
  • Table 6 presents the pharmacokinetic parameters for the 10 mg/kg and 50 mg/kg dose groups. The median Tmax were 480 and 24 hours for the 10 mg/kg and the 50 mg/kg dose groups, respectively.
  • the concentrations of SEQ ID NO:l (and related n+1, n-1 to n-15 oligonucleotide metabolites) in the following rat and monkey tissues were measured by a validated CE method (Southwest Bio-Labs, Inc.): kidney, liver, spleen, heart, lung, bone marrow (rat), lymph nodes (monkey) and brain tissues.
  • the tissue samples were collected from the designated animals as described in the 21-day toxicity studies: rats, administered bolus intravenous injections of SEQ ID NO:l every other day at 2, 10, or 50 mg/kg; and monkeys administered SEQ ID NO:l by continuous intravenous infusion at 2, 10, or 50 mg/kg/day.
  • Table 7a Extraction of SEQ ID NO:l and metabolites (n+1 through n-13) from Rat and Monkey Tissues with Analysis by Capillary Electrophoresis
  • Table 7b Extraction of SEQ ID NO:l and metabolites (n+1 through n-13) from Rat and Monkey Tissues with Analysis by Capillary Electrophoresis Mean Concentration ( ⁇ g/g) total metabolites
  • SEQ ID NO:l was administered to Sprague-Dawley rats via intravenous bolus injection every second day up to 21 days, with a 21 -day recovery period.
  • the study design included 4 treatment groups; vehicle controls, 2 mg/kg, 10 mg/kg, and 50 mg/kg.
  • the main study group consisted of 10 animals/sex, and an additional 5 animals/sex were included in the control and high-dose groups for a recovery phase. Satellite animals (6/sex/group) were also included for toxicokinetic analysis.
  • a single mortality resulted from the 50 mg/kg dose as well as various adverse clinical signs, a decrement in body weight gain (associated with diminished food consumption), lymphocytosis, moderate anemia and thrombocytopenia, and various relatively mild changes in serum chemistry parameters.
  • the 10 mg/kg dose level there were no clinical signs, no effects on body weight or food consumption, a lesser degree of anemia and thrombocytopenia, and only minor changes in a few serum chemistry parameters.
  • the 2 mg/kg/dose level had no remarkable effects on any of the above parameters.
  • Histopathological findings indicated the lymphoid system, kidneys, and liver as target organs at 10 and 50 mg/kg, with evidence of recovery at the highest dose level. Again, these findings were consistent with the known toxicity profile of phosphorothioates.
  • Target organs were clearly identified through histopathologic changes as kidneys, liver, and lymph nodes.
  • SEQ ID NO: 1 -related changes were dose-dependent in severity and/or incidence, and were reversed either completely or partially (with the suggestion of eventual complete recovery) during the 3-week post-dosing treatment-free period.
  • the 2 mg/kg/day dose level was identified as the no-adverse- effect-level (NOAEL).
  • Intravenous Compatibility Assay SEQ ID NO: 1 injection was tested for its potential to cause haemolytic activity based on cell lysis and haemoglobin release in human whole blood and did not cause haemolysis under static or dynamic conditions.
  • EXAMPLE 11 Clinical Trials using SEQ ID NO:l in Combination with Various Chemotherapeutics Examples of potential designs for clinical trials to test SEQ ID NO:l in combination with various known chemotherapeutics are provided in Table 8.
  • PROTOCOL LO1-1409 (RENAL CELL CARCINOMA)
  • SEQ ID NO: land capecitabine combination therapy in patients with advanced or metastatic renal cell carcinoma (Phase I/II)
  • SEQ ID NO: 1 was administered as a continuous intravenous infusion for 21 days at a starting dose of 148.0 mg/m 2 /day (phase I) or 185 mg/m 2 /day (phase II) in combination with capecitabine administered orally at a fixed dose of 1660 mg/m 2 /day (divided into two daily doses for 21 days) followed by 7 days of rest.
  • PROTOCOL L6090 (SOLID TUMOURS)
  • Unresectable Colorectal Cancer Population Locally advanced or metastatic colorectal cancer (refractory, unresectable). Patients must have had at least one standard prior chemotherapy with no prior oxaliplatin-containing regimen.
  • AGC Absolute granulocyte count Allo SCT: Allogenic stem cell transplantation
  • Auto SCT Autologous stem cell transplantation
  • ALP Alkaline phosphatase
  • LVEF Left ventricular ejection fraction
  • PLT and pits Platelet count
  • Rx Therapy SGOT: Serum glutamic-oxalacetic transferase
  • EXAMPLE 12 Phase I Study Of SEQ ID NO:l Given By Continuous Intravenous Infusion (CVT) In Patients With Advanced Cancer
  • Second phase escalation At least 3 patients/cohort; dose escalation of 20-30% until DLT
  • MTD Definition The dose level at which at least one-third of the patients experience DLT .
  • a heparinized blood sample was collected at baseline and then at 1, 2, 3, 4 and 6 hours after start of infusion. On Days 8 and 15, a single heparinized sample was taken. On Day 22 a heparinized blood sample was collected prior to the end of infusion and at 0.25, 0.5, 1, 2, 4 and 6 hours following the end of infusion. A single sample was collected on day 1 of Cycle 2 just prior to the start of the Cycle 2 infusion. Plasma was decanted and stored at -80° C.
  • PPD Development performed pharmacokinetic (pK) analysis of SEQ ID NO: 1 in plasma.
  • Plasma concentration of SEQ ID NO: 1 was determined by capillary electrophoresis (CE) with ultraviolet absorbance detection, and metabolite concentrations for Nl through N4 were calculated using extinction coefficients, molecular weight and internal standard concentration. The method was validated to a lower limit of quantitation (LLOQ) for SEQ ID NO : 1 of 0.125 ⁇ g/mL.
  • LLOQ lower limit of quantitation
  • the intra-assay coefficients of variation (CV) were 1.9% at LLOQ and 4.9-10.4% at levels above LLOQ; and inter-assay CV were 15% at LLOQ and 5.6 to 11.7% at levels above LLOQ.
  • DLT dose-limiting toxicity
  • MTD maximum tolerated dose
  • DLT was defined as Grade 4 neutropenia lasting 3 days or with fever; Grade 4 thrombocytopenia (T) or Grade 3 T with Grade 1 bleeding; or Grade 3 non-hematologic toxicity.
  • Thirty-six patients [25 males, median age: 60 (29-78), median KPS 90 (70-100); 15 renal cell, 9 colorectal cancer and 12 other] received 49 cycles of therapy at doses ranging from 18.5 to 222 mg/m 2 /day (6 levels).
  • the other serious non-dose limiting adverse events observed on the trial include: two patients who developed Grade 3 infections (non-neutropenic) with gram-negative pathogens during treatment cycles 2 and 4, respectively.
  • One patient with extensive retroperitoneal disease developed a bowel obstruction during the second cycle of treatment that was eventually confirmed to have resulted from disease progression.
  • Hematologic toxicity was mild; common (Grade 1-2) non-hematologic toxicities were: fatigue (69%), anorexia (42%), and nausea (38%).
  • SD stable disease
  • one patient with pancreatic cancer had stable disease for 2 months (therapy discontinued in cycle 3 due to central line infection)
  • two patients with colorectal cancer experienced disease stabilization of 4 and 6 months duration, respectively
  • one patient with renal cell carcinoma experienced disease stabilization for 4 months.
  • SEQ ID NO:l was well tolerated by 11 patients at the recommended phase II dose of 185 mg/m 2 /day.
  • SEQ ID NO: 1 AUC verses actual dose is depicted in Figure 36 A. Plasma concentrations versus time at different dose levels of SEQ ID NO:l are shown in Figure 36B. Detectable levels of SEQ ID NO: 1 were observed in plasma at doses of 37 mg/m 2 /day and above, but detectable plasma concentrations for all 4 sampling days were only observed for the 3 highest dose levels. The pharmacokinetic parameters exhibited dose independence and high inter-individual variability. The terminal half-life of SEQ ID NO: 1 was approximately 3 hours at the recommended phase II dose.
  • N/A Parameters not available due to missing samples or values below level of detection
  • Phase I portion escalated the dose of SEQ ID NO: 1 in combination with a fixed dose of capecitabine in order to develop the recommended dose for the Phase II portion.
  • a Simon two-stage design was utilized for the Phase II portion with a target activity level of 25% and a lower activity level of 10% and scheduled first assessment after 18 patients are evaluable for efficacy in the Phase II portion.
  • Grade 4 pancytopenia (1), pulmonary embolism (1) and bone pain (1)
  • Grade 3 thrombocytopenia (4), lymphopenia (3), anemia (2), neutropenia (2), nausea/emesis (2), infections (3), fatigue (2); neuropathy, thrombosis, dehydration, hypophosphatemia, diarrhea (1 each).
  • Additional common grade 1/2 toxicities include: nausea (62%), fatigue (45%), emesis (35%), hematologic toxicity (28%), anorexia (28%), and diarrhea (24%).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention porte sur des oligonucléotides antisens dirigés contre un gène mammalien de la ribonucléotide réductase R2 et sur des combinaisons des oligonucléotides antisens avec un ou plusieurs agents chimiothérapeutiques destinés à être utilisés dans le traitement du cancer.
PCT/CA2004/000806 2003-05-31 2004-05-31 Oligonucleotides antisens diriges contre la ribonucleotide reductase r2 et leurs utilisations dans le traitement du cancer WO2004106518A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32024003P 2003-05-31 2003-05-31
US60/320,240 2003-05-31

Publications (1)

Publication Number Publication Date
WO2004106518A1 true WO2004106518A1 (fr) 2004-12-09

Family

ID=33489172

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2004/000806 WO2004106518A1 (fr) 2003-05-31 2004-05-31 Oligonucleotides antisens diriges contre la ribonucleotide reductase r2 et leurs utilisations dans le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2004106518A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998005769A2 (fr) * 1996-08-02 1998-02-12 Genesense Technologies, Inc. Sequences antisens et antitumorales dirigees conte les constituants r1 et r2 de la ribonucleotide reductase
WO1999002673A2 (fr) * 1997-07-10 1999-01-21 Genesense Technologies, Inc. Sequences oligonucleotidiques antisens servant d'inhibiteurs de micro-organismes
WO2000047733A1 (fr) * 1999-02-11 2000-08-17 Genesense Technologies Inc. Sequences antisens antitumorales contre les constituants r1 et r2 de la ribonucleotide reductase
WO2002085308A2 (fr) * 2001-04-24 2002-10-31 Epigenesis Pharmaceuticals, Inc. Compositions, formulations et trousses contenant des oligonucleotides anti-sens et des steroides anti-inflammatoires et/ou un ubiquinone pour le traitement de maladies respiratoires ou pulmonaires
WO2004070033A1 (fr) * 2003-02-10 2004-08-19 Genesense Technologies Inc. Oligonucleotides antisens se dirigeant vers la ribonucleotide reductase r2 et son utilisation dans le traitement du cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998005769A2 (fr) * 1996-08-02 1998-02-12 Genesense Technologies, Inc. Sequences antisens et antitumorales dirigees conte les constituants r1 et r2 de la ribonucleotide reductase
WO1999002673A2 (fr) * 1997-07-10 1999-01-21 Genesense Technologies, Inc. Sequences oligonucleotidiques antisens servant d'inhibiteurs de micro-organismes
WO2000047733A1 (fr) * 1999-02-11 2000-08-17 Genesense Technologies Inc. Sequences antisens antitumorales contre les constituants r1 et r2 de la ribonucleotide reductase
WO2002085308A2 (fr) * 2001-04-24 2002-10-31 Epigenesis Pharmaceuticals, Inc. Compositions, formulations et trousses contenant des oligonucleotides anti-sens et des steroides anti-inflammatoires et/ou un ubiquinone pour le traitement de maladies respiratoires ou pulmonaires
WO2004070033A1 (fr) * 2003-02-10 2004-08-19 Genesense Technologies Inc. Oligonucleotides antisens se dirigeant vers la ribonucleotide reductase r2 et son utilisation dans le traitement du cancer

Similar Documents

Publication Publication Date Title
US7968526B2 (en) Antisense oligonucleotides directed to ribonucleotide reductase R2 and uses thereof in the treatment of cancer
EP1786905B1 (fr) Molecules de petit arn interferant dirigees contre la ribonucleotide reductase et ses utilisations
US20080311126A1 (en) Antisense Oligonucleotides Directed to Ribonucleotide Reducatase R2 and Uses Thereof in Combination Therapies for the Treatment of Cancer
US20170016002A1 (en) Method of Treating Cancer by Inhibition of DNA Repair Proteins
EP3700538A1 (fr) Méthodes de traitement du cancer et/ou d'amélioration de la sensibilité à un traitement contre le cancer par accroissement de la charge de mutations tumorales ou des indels tumoraux
US20070274947A1 (en) Antisense Oligonucleotides Directed to Ribonucleotide Reductase R1 and Uses Thereof in the Treatment of Cancer
US20110003879A1 (en) Antisense oligonucleotides targeted to the coding region of thymidylate synthase and uses thereof
WO2005070469A1 (fr) Techniques de traitement du mesotheliome au moyen d'un oligonucleotide antisens d'une thymidylate synthase
US7465714B2 (en) Oligonucleotide inhibitors of MBD2/DNA demethylase and uses thereof
WO2004106518A1 (fr) Oligonucleotides antisens diriges contre la ribonucleotide reductase r2 et leurs utilisations dans le traitement du cancer
US20100204305A1 (en) Small interfering rna molecules against ribonucleotide reductase and uses thereof
WO2008124927A1 (fr) Siarn contre la thymidylate synthase et son utilisation dans les schémas de traitement du cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase