WO2013114339A1 - Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates - Google Patents

Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates Download PDF

Info

Publication number
WO2013114339A1
WO2013114339A1 PCT/IB2013/050882 IB2013050882W WO2013114339A1 WO 2013114339 A1 WO2013114339 A1 WO 2013114339A1 IB 2013050882 W IB2013050882 W IB 2013050882W WO 2013114339 A1 WO2013114339 A1 WO 2013114339A1
Authority
WO
WIPO (PCT)
Prior art keywords
hsp27
agent
cancer
erlotinib
cells
Prior art date
Application number
PCT/IB2013/050882
Other languages
French (fr)
Other versions
WO2013114339A4 (en
Inventor
Martin E Gleave
Amina ZOUBEIDI
Masafumi Kumano
Original Assignee
The Universityof British Columbia
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 The Universityof British Columbia filed Critical The Universityof British Columbia
Priority to EP13744042.6A priority Critical patent/EP2809325A4/en
Priority to CA2861415A priority patent/CA2861415A1/en
Priority to AU2013216361A priority patent/AU2013216361B2/en
Priority to US14/372,972 priority patent/US9717792B2/en
Publication of WO2013114339A1 publication Critical patent/WO2013114339A1/en
Publication of WO2013114339A4 publication Critical patent/WO2013114339A4/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • This application relates to combination therapy for the treatment of cancer using inhibitors of heat shock protein 27 (Hsp27) and an epidermal growth factor tyrosine kinase inhibitor (EGFR-TKI) such as erlotinib, or antifolates such as pemetrexed.
  • Hsp27 heat shock protein 27
  • EGFR-TKI epidermal growth factor tyrosine kinase inhibitor
  • antifolates such as pemetrexed.
  • Hsp27 is a cell survival protein found at elevated levels in many human cancers including prostate, lung, breast, ovarian, bladder, renal, pancreatic, multiple myeloma and liver cancer.
  • many anti-cancer therapies are known to further elevate Hsp27 levels.
  • Hsp27 levels increased four-fold in prostate cancer patients after treatment with chemo- or hormone therapy.
  • Increased levels of Hsp27 in some human cancers are associated with metastases, poor prognosis and resistance to radiation or chemotherapy.
  • Hsp27 has been disclosed as a therapeutic target in the treatment of cancer.
  • US Patent No. 7,101,991 discloses antisense oligonucleotides and siRNA that inhibit Hsp27 expression. Additional oligonucleotide sequences targeting Hsp27 expression are disclosed in WO2007/025229 and US Patent Publications Nos. 2009/0264502 and 2011/0144185.
  • Non- oligonucleotide compounds for inhibition of Hsp27 have also been disclosed, including berberine derivatives described in European Patent EP0813872, and compounds described in JP 10045572, JP 10045574, JP10036261 and JP 10036267, all assigned to Kureha Chemical Industries Co,. Ltd.
  • Paclitaxel has also been shown to be an inhibitor of Hsp27 expression. Tanaka et al., Int J Gynecol Cancer. 2004 Jul-Aug;14(4):616-20. Nucleoside inhibitors that binds to Hsp27 are also known. One of these, bromovinyldeoxyuridine (BRDU, Brivudine, RP101) has been tested in clinical trials and shown to enhance survival of patients with pancreatic cancer. Tuukanen et al. J Cancer Res Clin Oncol. 2011 Sep;137(9):1349-61.
  • BRDU bromovinyldeoxyuridine
  • OGX-427 an antisense oligonucleotide described in US Patent No. 7,101,991 (Seq. ID No. 1, OncoGenex Technologies Inc.), significantly decreases levels of Hsp27, induces apoptosis in several human cancer cell lines, has single agent anti-tumor activity, and acts as a chemosensitizer in combination with several cytotoxic drugs including docetaxel.
  • OGX-427 is being evaluated in a Phase 1 study in patients with breast, prostate, ovarian, non-small cell lung, or bladder cancer who have failed potentially curative treatments or for which a curative treatment does not exist.
  • a method for treating cancer in an individual comprising the steps of administering to the individual a therapeutically effective amount of a first active agent which is an inhibitor of Hsp27 activity, and administering to the individual a therapeutically effective amount of a second active agent which is an inhibitor of EGFR tyrosine kinase.
  • the first and second agents are administered such that both are present at therapeutically relevant levels during a common time period.
  • treatment with the Hsp27 inhibitor is commenced prior to treatment with the EGFR tyrosine kinase inhibitor.
  • a method for treating cancer in an individual comprising the steps of administering to the individual a therapeutically effective amount of a first active agent which is an inhibitor of Hsp27 activity, and administering to the individual a therapeutically effective amount of a second active agent which is an antifolate.
  • the first and second agents are administered such that both are present at therapeutically relevant levels during a common time period.
  • treatment with the Hsp27 inhibitor is commenced prior to treatment with the antifolate.
  • Fig. 1 shows induction of Hsp27 following treatment of A549 lung cancer cells with erlotinib.
  • Fig. 2 shows cell viability as a function of erlotinib treatment with and without treatment with siHsp27.
  • Fig. 3A shows cell viability as a function of erlotinib treatment with and without treatment with Hsp27 antisense.
  • Fig. 3B shows the combination index indicative of synergy for the combined treatment of erlotinib and Hsp27 antisense.
  • Fig. 3C shows cell cycle results for treatments with erlotinib and Hsp27 antisense.
  • Fig. 4A shows cell viability in A549 cells overexpressing Hsp27 following erlotinib treatment.
  • Figs. 4B and C show flow cytometry results for A549 cells overexpressing Hsp27 following erlotinib treatment.
  • Fig. 5A shows change in tumor volume in A549 xenograft tumors over time with various treatments.
  • Fig 5B shows this data in bar graph form.
  • Fig. 5C shows the average number of Tunel positive cells in cells treated with erlotinib, OGX-427 or both.
  • Fig. 6 shows cell viability as a function of pemetrexed treatment with and without treatment with Hsp27 antisense.
  • Fig. 7 shows expression of Hsp27 in HCC827 lung cancer cells with and without treatment with erlotinib.
  • Fig. 8A shows Hsp27 mRNA expression in parental and erlotinib resistant HCC827 cells.
  • Fig. 8B shows cell viability as a function of erlotinib concentration in parental and erlotinib resistant HCC827 cells.
  • Fig. 9A shows cell viability of parental HCC827 cells at different concentrations of erlotinib, with and without treatment with OGX-427.
  • Fig. 9B shows cell viability of erlotinib resistant HCC827 cells at different concentrations of erlotinib, with and without treatment with OGX-427.
  • Fig. 10A shows cell viability for individual and combination therapies using pemetrexed and OGX-427 at increasing concentrations of therapeutic.
  • Fig. 1 OB is a combination index (CI) plot of this data.
  • the present invention provides a method for treating cancer in a patient diagnosed as suffering from cancer.
  • the patient is a human patient, although the method can also be used in veterinary applications, for example in the treatment of cancer in dogs, cats and other pets.
  • cancer types that may be treated using the method of the invention include breast, prostate, ovarian, uterine, non-small cell lung, bladder, gastric, liver, endometrial, laryngeal and colorectal cancers; squamous cell carcinomas such as esophageal squamous cell carcinoma, glioma, glioblastoma, melanoma, multiple myeloma and lymphoma.
  • a second active agent is combined with an inhibitor of Hsp27.
  • the second active agent is an inhibitor of EGFR tyrosinase kinase or an antifolate.
  • the second active agent is selected to have independent therapeutic activity for the cancer to be treated in a particular individual.
  • treating refers to performing the method steps of the invention with intention and expectation of a therapeutic benefit to the patient. It would be understood in the art that not all patients respond favorably, or to the same extent to a given treatment. Furthermore, it will be understood in the art that the results obtained for any individual cannot be compared to results for that individual in the absence of the treatment. Thus, actual therapeutic benefit is not required to fall within the scope of the concept of "treating” nor is conclusive evidence that an observed benefit arose from the treatment.
  • Hsp27 refers to heat shock protein 27, an approximately 27 kilodalton stress- induced protein. Hsp27 is also sometimes referred to as heat shock protein beta-1 (HSPB1).
  • the sequences of Hsp27 are known in the art for Homo sapiens (AB020027, X54079, NM_006308, NM_001540 and NM_001541), dogs (NP_001003295), cattle (NP_001020740), mice
  • EGFR epidermal growth factor receptor
  • pEGFR phosphorylated EGFR
  • NSCLC non-small cell lung cancer
  • SCC refers to squamous cell carcinoma. SCC can occur in lung tissue or in other locations in the body. SCC originating in lung tissue is a recognized subset of NSCLC
  • AD or "lung AD” refers to lung adenocarcinoma, a different recognized subset of NSCLC.
  • LCC refers to large cell lung carcinoma, a different recognized subset of NSCLC.
  • lung cancer when used without further labeling refers generically to NSCLC, including lung SCC, lung-AD and LCC.
  • p-Hsp27 refers to hsp27 that is phosphorylated at serine 82.
  • Other serine residues may also be phosphorylated as described in "Regulation of HSP27 function is highly dependent on phosphorylation (Stetler et al., 2009, 2010).
  • Hsp27 is phosphorylated at several distinct serine residues (Serl5, Ser78, and Ser82 in humans). Phosphorylation at Ser82 leads to dissociation to lighter oligomers (Theriault et al., 2004) and loss of the chaperone function of HSP27 which is associated with the nonphosphorylated structure.
  • p-Hsp27 can suppress cell death-signaling (Bruey et al., 2000; Concannon et al., 2001 ; Benn et al., 2002; Rane et al., 2003; Lee et al., 2005; Voss et al., 2007).
  • Cell death suppression requires phosphorylation of Hsp27 (Benn et al., 2002).
  • measurement of increased levels p-Hsp27 is an indication that Hsp27 is acting in a
  • PARP refers to poly-(ADP-ribose) polymerase, a marker of apoptosis and a caspase substrate.
  • Caspase 3 refers to an enzyme that is a member of the cysteine-aspartic acid protease (caspase) family associated with the apoptotic pathway.
  • Cleaved caspase 3 is the large fragment (17/19 kDa) of activated caspase-3 resulting from cleavage adjacent to Aspl75.
  • Antibodies are used that bind to cleaved caspase-3 but that do not recognize full length caspase-3 or other cleaved caspases. This antibody detects non-specific caspase substrates by Western blot.
  • siRNA refers to double stranded RNA or RNA and DNA species that are active to reduce expression of targeted gene. These molecules are known variously as “small interfering RNA “, “short interfering RNA” or “silencing RNA.” siRNA strands are usually 20- 25 nucleotides long, although larger precursor molecules which are subject to cleavage in vivo to form the active species are within the scope of the term as used herein.
  • A549 cells ATCC No. CCL-185TM is an epithelial lung carcinoma (Giard DJ, et al. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J. Natl. Cancer Inst. 51 : 1417-1423, 1973.)
  • A549 cells are representative of NSCLC without an activating mutation in the EGFR tyrosine kinase domain. Such cells are not considered clinically sensitive to EGFR-TKI such as erlotinib and gefitinib.
  • HCC827 ATCC No. CRL-2868TM is an epithelial cell line derived from lung
  • CI or “combination index” is a quantitative analysis based on a theorem proposed by Chou-Talalay in the 1980s (Chou TC, Talalay PA. J. Biol. Chem (1977) 252: 6438-42; Chou TC. Talalay PA. Eur. J. Biochem. (1981) 115:207-216).
  • Software called CalcuSyn Biosoft, Cambridge UK
  • the software performs multiple drug dose-effect calculations using the Median Effect methods described by T-C Chou and P. Talalay (Trends Pharmacol. Sci. 4, 450-454).
  • TUNEL "TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining (1)".
  • TUNEL staining relies on the ability of the enzyme terminal deoxynucleotidyl transferase to incorporate labeled dUTP into free 3'-hydroxyl termini generated by the fragmentation of genomic DNA into low molecular weight double-stranded DNA and high molecular weight single stranded DNA.
  • Crystal violet is a triphenylmethane dye (4-[(4- dimethylaminophenyl)-phenyl-methyl]-N,N-dimethyl-aniline) also known as Gentian violet (or hexamethyl pararosaniline chloride).
  • Gentian violet or hexamethyl pararosaniline chloride.
  • the CV assay is used to determine cell viability or to determine cell proliferation under different testing conditions (Davey, Hazel M.; Kell, Douglas B. Microbial Reviews (1996) 60: 4: 641-696).
  • inhibitor of Hsp27 refers to a compound that reduces the activity of Hsp27, either by interaction with Hsp27 or its intended target, or through reduction in the amount of Hsp27 present in cells.
  • Inhibitors of Hsp27 expression of various different types are known in the art. Examples of inhibitors includes nucleotide compounds targeting Hsp27, peptide aptamers, flavonoid inhibitors of Hsp27, antibodies that interact with Hsp27, and interferon- ⁇ which has been shown to downregulate expression of Hsp27.
  • the specific route of administration, the dosage level and the treatment frequency will depend on the nature of the active agent employed. In general, the therapeutic agent may be administered by intravenous, intraperitoneal, subcutaneous, topical or oral routes, or direct local tumor injection.
  • the Hsp27 inhibitor is a nucleotide inhibitor.
  • nucleotide inhibitors include antisense sequences, which may be full-length antisense (see Horman et al., Int. J. Cancer (1999) 82: 574-582), or shorter oligonucleotide sequences, having a length of 100 bases or less, for example 12 to 30 bases.
  • antisense species are complementary to the target Hsp27 gene to an extent sufficient to achieve antisense inhibition in vivo, and may include degeneracy to take into account allelic variation.
  • Specific oligonucleotide antisense inhibitors of Hsp27 are known in the art from US Patent Publications 2004/0127441 and 2009/0264502 which are incorporated herein by reference.
  • the Hsp27 inhibitor is OGX-427, an antisense oligonucleotide made by OncoGenex that is currently in clinical trials for treatment of various types of cancer.
  • OGX-427 is a 4-12-4 2'-MOE gapmer oligonucleotide with phosphorothiolatedinternucleotide linkages which can be represented as
  • the nucleotide Hsp27 inhibitor is a double stranded RNA species (or precursor) that operates by an siRNA mechanism to reduce expression of Hsp27.
  • RNA species for this purpose are known from US Patent Publication 2004/0127441 and Chauhan, et al. (2003) Cancer Res.63, 6174-6177.
  • One specific siRNA inhibitor used in the examples below has the sequence GCU GCA AAA UCC GAU GAGA (Seq ID No. 2) and it is used with its complement to form the active double stranded inhibitor.
  • Peptide aptamers (Gilbert et al, Oncogene. 2011 Mar 21. [Epub ahead of print]); and antibodies (Tezel and Wax, J. Neuroscience 10:3553-3562 (2000) that interact with Hsp27 are also known and could serve as inhibitors of Hsp27 in accordance with the invention.
  • Other peptides that bind to Hsp27, such as CP91 or binding fragments thereof as described in US Patent Publication No. 2007/0003555 could also be employed.
  • Cytokines such as interferon- ⁇ are also known to inhibit Hsp27 and can be used as inhibitors in the present invention. Yonekura et al., Cell Death and Differentiation (2003) 10: 313-322.
  • Hsp27 Other inhibitors of Hsp27 are also known which are generally "small molecule" inhibitors. These include flavonoids such as quercetin, (Morino et al., in vivo (1997) 11: 265- 270; JP 10045572, JP 10045574, JP10036261 and JP 10036267), and biphenyl isooxazoles such as 5-(5-Ethyl-2-hydroxy-4-methoxyphenyl)-4-(4-methoxyphenyl)isoxazole (KRIBB3) (Shin et al. The Journal of Biological Chemistry VOL. 280, NO. 50, pp. 41439-41448, December 16, 2005).
  • flavonoids such as quercetin, (Morino et al., in vivo (1997) 11: 265- 270; JP 10045572, JP 10045574, JP10036261 and JP 10036267)
  • KRIBB3 is available commercially from Sigma- Aldrich is understood to reduce Hsp27 activity by acting as a specific inhibitor (IC50 of 50 nM) of PKC-dependent phosphorylation of Hsp27.
  • IC50 of 50 nM
  • Related compounds are described in Lee et al., Bioorg Med ChemLett. 2011 Feb l;21(3):977-9. Epub 2010 Dec 13.Berberine derivatives have also been shown to inhibit Hsp27. (EP 0 813 872)
  • Paclitaxel has also been shown to be an inhibitor of Hsp27 expression. (Tanaka et al., Int J Gynecol Cancer. 2004 Jul-Aug; 14(4): 616-20).
  • Nucleoside inhibitors such as brivudine also are within the scope of the invention.
  • EGFR epidermal growth factor receptor
  • inhibitors of EGFR tyrosine kinase activity include known therapeutic compounds such as erlotinib (TarcevaTM) and gefitinib (IressaTM) both of which are quinazoline compounds;.
  • EGFR-targeting antibodies and antibody fragments and their use in treatment of cancer are known and include cetuximab (ErbituxTM) and panitumumab (VectibixTM). Others are known from US Patent Publication No. 2011-0117110.
  • Oligonucleotide sequences for inhibition of wild-type or mutant forms of EGFR are known from US Patent Publication No. 2012-0022132; US Patent Publication No. 2009-0118208 and US Patent Publication No. 2003-0170891.
  • Antifolates are folate-analogue metabolic inhibitors. They may interfere with one or more of three enzymes involved in purine and pyrimidine synthesis— thymidylate synthase (TS), dihydrofolate reductase (DHFR), and glycinamide ribonucleotide formyltransferase (GARFT).
  • TS thymidylate synthase
  • DHFR dihydrofolate reductase
  • GARFT glycinamide ribonucleotide formyltransferase
  • Specific antifolates used in cancer therapy include methotrexate, pemetrexed, pralatrexate, and raltitrexed.
  • Each of the inhibitors of Hsp27 may be used individually or in combination with one or more of the other inhibitors.
  • Each of these inhibitors or combinations may be used with any second agent that is an inhibitor of EGFR tyrosine kinase activity or an antifolate, with the proviso that the second agent is selected for utility in the treatment of the particular cancer with which the individual treated is diagnosed.
  • combinations within the scope of the invention may be further combined with additional therapeutics effective
  • the therapeutic agents of the invention are suitably formulated in one or more pharmaceutically acceptable carriers of a type consistent with the intended mode of
  • the Hsp27 and second agents can be administered as an injectable liquid, an oral or aerosol composition, and may be administered systemically (for example intravenous, intramuscular, or oral) or regionally to an area harboring a cancer to be treated (for example intra-nasal, intra-tumoral, intra- tracheal, intrapleural and topical).
  • the Hsp27 inhibitor is an antisense
  • oligonucleotide Oligonucleotide therapeutics are commonly being tested at treatment levels of 1 to 500 mg/m2/day and preferred treatment levels are selected to balance toxicity with therapeutic benefit. Such levels are appropriate for use in the therapeutic combinations of the invention.
  • One specific oligonucleotide product is "OGX-427" (Seq ID No. 2) which is currently provided to human patients at about 600mg per patient in a 25 mg/niL concentration formulated as a mannitol-phosphate buffer solution (pH 7.4) for IV administration.
  • OGX-427 dosing solutions are administered intravenously using an infusion pump. In some situations, the administration will be preceded or accompanied by administration of an antihistamine.
  • Antisense oligonucleotides may also be administered in specific carrier, such as that described in US Patent Publication No. US 2009/0142413 Al.
  • Hsp27 Some treatment-naive cancers have been found to express Hsp27.
  • Hsp27 expression may arise as a response to treatment with chemotherapy or radiotherapy.
  • the method and the therapeutic combination of the invention are applicable to the treatment of cancers that inherently or as a consequence of prior or concurrent chemotherapy or radiation treatments express Hsp27.
  • Specific cancers include, without limitation, prostate, bladder, lung, breast, osteosarcoma, pancreatic, colon, testicular, colorectal, urothelial, renal cell,
  • hepatocellular, leukemia, lymphoma, and ovarian cancer hepatocellular, leukemia, lymphoma, and ovarian cancer, melanoma, central nervous system malignancies, and squamous cell carcinoma.
  • the cancer is a treatment-naive cancer that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer.
  • the level of Hsp27 expression in prostate cancer cells would be compared to an average value for non-cancerous prostate tissue.
  • the cancer is one that been previously treated in the individual by chemotherapy.
  • the cancer is preferably one that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer and/or that expresses Hsp27 as a level greater than levels in the individual prior to the treatment.
  • the cancer is one that been previously treated in the individual by radiotherapy.
  • the cancer is preferably one that that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer and/or that expresses Hsp27 as a level greater than levels in the individual prior to the treatment.
  • the therapeutic combination as described above can be used.
  • NSCLC non-small cell lung cancers in some patients possessing an activating mutation in the EGFR gene
  • NSCLC can be treated using EGFR-TKIs such as erlotinib and gefitinib.
  • EGFR-TKIs such as erlotinib and gefitinib.
  • these NSCLC patients develop acquired resista
  • the combination therapy of the EGFR-TKI and the Hsp27 inhibitor provides two benefits: First , it enhances the initial activity of the EGFR-TKI so that more effectiveness will be obtained before the onset of resistance. Second, it maintains the efficacy of the drug for a longer period of time, even after the resistance causing mutations have begun to appear. Erlotinib induces also expression of Hsp27 in HCC827 cells which are representative of NSCLC (lung).
  • Hsp27 results in enhanced efficacy of erlotinib in these cells. Greater levels of Hsp27 expression were observed in erlotinib resistant cell lines derived from HCC827 than in the parent strain. This may be drug resistance arising due to a second mutation in the EGFR thymidine kinase , reducing the sensitivity of the EGFR-TK to the drug, or may be a separate mechanism of drug resistance in which the cells produce higher levels of Hsp27. Regardless of mechanism, enhancement of drug activity continues in cancers that develop resistance to erlotinib. (See Experimental results, J, herein).
  • Hsp27 inhibitors can have low toxicity (for example OGX-427 (Seq. ID No. 1 ) was found to have no dose-limiting toxicity in a Phase I clinical study of patients with castrate resistant prostate cancer, breast cancer, ovarian cancer or non-small cell lung cancer), this r
  • Hsp27 inhibitors and the second agent have individual beneficial activity, the combined benefit was synergistic at least at some dosage levels.
  • the result of the invention is not merely the additive effect of two known therapeutics.
  • erlotinib induces Hsp27 expression in A549 lung cancer cells.
  • A549 lung cancer cells were exposed to 10 ⁇ Erlotinib and the amount of p- Hsp27 (ser82), Hsp27, p-EGFR (Tyr 1068), EGFR, cleaved PARP (an indicator of apoptosis) were determined by Western Blot after 1, 6, 24. 48 and 72 hours of exposure. Increasing amounts of EGFR, Hsp27 and p-Hsp27 were observed over time. p-EGFR was initially present but was not observed following treatment. Cleaved PARP was observed at all times following 24 hours of treatment.
  • Hsp27 mRNA expression was analyzed by quantitative RT-PCR. Hsp27 mRNA levels were normalized to levels of GAPDH mRNA. Bars, SD. ** and *, differ from control (P ⁇ 0.01 and P ⁇ 0.05, respectively). The results are shown in Fig. 1. As indicated, both levels of erlotinib resulted in increased Hsp27 expression.
  • Hsp27 knockdown enhances cytotoxic effect and apoptosis after treatment with erlotinib.
  • Cells were transfected with 20 nM Hsp27 siRNA (Seq ID No. 2) or a scrambled siRNA duplex containing 5 '-CAGCGCUGACAACAGUUUCAU-3' SEQ ID NO: 3 for 1 day, and treated with varying concentrations of erlotinib for 48 hours. Cell viability was determined by crystal violet assay. The results are summarized in Fig. 2. As shown, at each concentration, the reduction in cell viability was greater when both agents
  • Hsp27 and p-Hsp27 were both absent in cells treated with the siHsp27 but not the scrambled control.
  • P- EGFR was lower in cells treated with the combination that in cells treated with siHsp27 alone.
  • Cleaved PARP was greater in cells with the combined treatment than in cells with either treatment alone.
  • A549 cells were transfected with 50nM OGX-427 (Seq. ID No. 1) or a scrambled control on two consecutive days and then treated with 10 ⁇ erlotinib for 72 hours after the second transfection. Cell viability was determined using a crystal violet assay. The results are shown in Fig. 3 A.
  • the synergistic effect of combination treatment with Erlotinib and OGX-427 in vitro was assessed using combination index (CI) values calculated by CalcuSyn software were assessed in A549 cells treated for 72 hours with OGX-427 alone, Erlotinib alone or combined treatment.
  • the CI for inhibitory concentration (IC) 50, IC75 and IC90 was 0.9, 0.7 and 0.7, respectively, indicative of a synergistic effect of this combined treatment at all levels of erlotinib tested (See Fig. 3B).
  • Hsp27-overexpression protects from cell death induced by Erlotinib in A549 cells.
  • Hsp27 OE A549 cells overexpressing Hsp27 (Hsp27 OE A549) cells were established and protein expression was confirmed by western blotting. The effect of overexpression of Hsp27 protein on the sensitivity to Erlotinib in A549 cells was tested using the. Hsp27 OE A549 cells and control vector-transfected (Empty A549) cells were treated with the indicated cor
  • Hsp27 OE A549 and Empty A549 cells were treated with varying levels of erlotinib. Protein levels were analyzed by western blotting and higher levels of cleaved PARP were observed in the empty A549 cells. The cells undergoing apoptosis (subGO/Gl fraction) were quantified using flow cytometry. Figs. 4B and C. (Bars, SD. *, differ from Empty A549 (P ⁇ 0.05)).
  • Hsp27-overexpression protects from cell death induced by erlotinib in A549 cells.
  • A549 cells were inoculated s.c. and when tumors reached 50-100 mm3, mice were treated with ScrB control oligodeoxynucleotide (ScrB) + diluent, ScrB + Erlotinib, OGX-427 + diluent or OGX-427 + Erlotinib Tumor volume was monitored for 7 weeks after treatment. As shown in Fig. 5A, the combined treatment substantially reduced tumor growth as compared to either treatment individually. Each data point represents the mean tumor volume in each group containing 8 mice + SEM.
  • mice were sacrificed and tumors excised for assessment. Tissue preparations were subjected to TUNEL staining, and the results are shown in Figure 5C More signs of apoptosis were observed for OGX-427 and erlotinib combined than any of the other three groups.
  • A549 cells were cultured as previously described by others (Van Schaeybroeck, Sandra; Kyula, Joan; Kelly, Donal M; et al. Mol Cancer Ther (2006) 5; 1154), and treated with pemetrexed at a
  • OGX-427 was also shown to enhance the antitumor effect of premetrexed in A549 cells.
  • A549 cells were treated 50nM OGX-427 on 2 consecutive days and incubated with varying concentrations of Pemetrexed for 72 h . 72 h later, cell viability was determined by crystal violet assay. The results are shown in Fig. 6. (** p ⁇ 0.01) Reduction in cell viability comparable to 1 ⁇ pemetrexed alone were obtained using the combination therapy and only 0.1 ⁇ , a tenfold reduction. Protein levels were also determined in these cells by immunoblott
  • Hsp27 positive and negative cells were assessed in a variety of human lung cancer tissues, namely squamous cell lung carcinoma (SCC), lung adenocarcinoma (AD), and large-cell lung carcinoma (LCC).
  • SCC squamous cell lung carcinoma
  • AD lung adenocarcinoma
  • LCC large-cell lung carcinoma
  • Assays for Hsp27 and p-Hsp27 are available commercially from Cell Signalling, Danvers USA, and Perkin-Elmer (USA).
  • Hsp27 generally and in combination with a second agent used in the treatment of particular types of lung cancer is beneficial in a majority of human NSCLC patients.
  • Lung cancer cell line HCC827 was cultured and exposed, under otherwise normal conditions, to from 0 to 50 micromolar erlotinib, and the cells were lysed under non-denaturing conditions and protein content was subjected to Western blotting technique.
  • HCC827cells erlotinib at 2.5 nanomolar concentration generated a decrease in p- EGFR (Tyr068) and an increase in p-Hsp27(ser82) as compared to non-treated cells, while protein levels for P-Hsp27(ser82), Hsp27 were increased.
  • the increase is in Hsp27 is shown graphically in Fig. 7.
  • Erlonitib-resistant HCC827 cells (Re-HCC827 #l,-#2, and -#3) were established by exposure to erlotinib and protein expression was confirmed by Western blotting.
  • the three new cell lines were resistant to erlotinib treatment and overexpressed Hsp27 (Fig. 8A), and had elevated levels of p-Hsp27 as compared to the parent cell lines.
  • the graph in Fig.8 B shows cell viability for the parental HCC827 cell line and the three resistant derivations (Re#l, #2, and #3) under conditions of increasing concentrations of erlotinib (0.1 to 1000 nanoMole).
  • the resistant cell lines are able to overcome the effects of erlotinib to a greater degree than the parental cell line can.
  • HCC827 parental and resistant HCC827 #3
  • OGX-427 was added to
  • ScrB control oligodeoxynucleotide was used as a control for the OGX-427.
  • Cell viability was measured using crystal violet technique. The results were expressed as a percentage of the cell viability in the presence of OligofectamineTM control in Figs 9A and 9B for the parental and resistant cell lines respectively.
  • the second, paler bars in the graphs are the OGX-427 treated cells, while the darker bars represent the cell viability of cells treated with ScrB control oligodeoxynucleotide.
  • the synergistic effect of combination treatment with pemetrexed and OGX-427 was assessed in A549 cells.
  • Cells were treated with ScrB control oligodeoxynucleotide, OGX-427 alone, pemetrexed with ASO control, and OGX-427 and pemetrexed together.
  • An additional control was OligofectamineTM transfecting agent alone.
  • the cells were treated with OGX-427 for 2 consecutive days, and then incubated with pemetrexed for 72 hours. Another 72 hours later, cell viability was determined by crystal violet assay.
  • the relative amounts of OGX-427 and pemetrexed was maintaed constant as the total amount of treatment agent was increased.
  • the concentrations were:
  • Fig. 10A The results are summarized in Fig. 10A. As shown, cell viability after was lowest in tne ceil to which combined OGX-427 and erlotinib treatment was applied.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Endocrinology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Combination therapy for cancer makes use of HSP27 inhibitors and EGFR tyrosine kinase inhibitors or antifolates.

Description

COMBINATION THERAPY FOR CANCER USING HSP27 INHIBITOR AND EGFR TYROSINE KINASE INHIBITORS OR ANTI-FOLATES
Field of the Invention
This application relates to combination therapy for the treatment of cancer using inhibitors of heat shock protein 27 (Hsp27) and an epidermal growth factor tyrosine kinase inhibitor (EGFR-TKI) such as erlotinib, or antifolates such as pemetrexed.
Background of the Invention
Hsp27 is a cell survival protein found at elevated levels in many human cancers including prostate, lung, breast, ovarian, bladder, renal, pancreatic, multiple myeloma and liver cancer. In addition, many anti-cancer therapies are known to further elevate Hsp27 levels. For example, Hsp27 levels increased four-fold in prostate cancer patients after treatment with chemo- or hormone therapy. Increased levels of Hsp27 in some human cancers are associated with metastases, poor prognosis and resistance to radiation or chemotherapy.
Hsp27 has been disclosed as a therapeutic target in the treatment of cancer. For example, US Patent No. 7,101,991 discloses antisense oligonucleotides and siRNA that inhibit Hsp27 expression. Additional oligonucleotide sequences targeting Hsp27 expression are disclosed in WO2007/025229 and US Patent Publications Nos. 2009/0264502 and 2011/0144185. Non- oligonucleotide compounds for inhibition of Hsp27 have also been disclosed, including berberine derivatives described in European Patent EP0813872, and compounds described in JP 10045572, JP 10045574, JP10036261 and JP 10036267, all assigned to Kureha Chemical Industries Co,. Ltd. Paclitaxel has also been shown to be an inhibitor of Hsp27 expression. Tanaka et al., Int J Gynecol Cancer. 2004 Jul-Aug;14(4):616-20. Nucleoside inhibitors that binds to Hsp27 are also known. One of these, bromovinyldeoxyuridine (BRDU, Brivudine, RP101) has been tested in clinical trials and shown to enhance survival of patients with pancreatic cancer. Tuukanen et al. J Cancer Res Clin Oncol. 2011 Sep;137(9):1349-61.
Preclinical studies show that OGX-427, an antisense oligonucleotide described in US Patent No. 7,101,991 (Seq. ID No. 1, OncoGenex Technologies Inc.), significantly decreases levels of Hsp27, induces apoptosis in several human cancer cell lines, has single agent anti-tumor activity, and acts as a chemosensitizer in combination with several cytotoxic drugs including docetaxel. OGX-427 is being evaluated in a Phase 1 study in patients with breast, prostate, ovarian, non-small cell lung, or bladder cancer who have failed potentially curative treatments or for which a curative treatment does not exist.
Summary of the Invention
The present inventors have discovered that combination therapy using Hsp27 inhibitors and EGFR tyrosine kinase inhibitors leads to superior therapeutic effects by reducing tumor growth rates and enhancing the cytotoxic effect of the EGFR tyrosine kinase inhibitors. Thus, in accordance with one aspect of the invention, a method is provided for treating cancer in an individual, including a human individual, comprising the steps of administering to the individual a therapeutically effective amount of a first active agent which is an inhibitor of Hsp27 activity, and administering to the individual a therapeutically effective amount of a second active agent which is an inhibitor of EGFR tyrosine kinase. The first and second agents are administered such that both are present at therapeutically relevant levels during a common time period. In some embodiments of the invention, treatment with the Hsp27 inhibitor is commenced prior to treatment with the EGFR tyrosine kinase inhibitor.
The present inventors have also discovered that combination therapy using Hsp27 inhibitors and an antifolate leads to superior therapeutic effects by reducing tumor growth rates and enhancing the cytotoxic effect of the antifolate. Thus, in accordance with one aspect of the invention, a method is provided for treating cancer in an individual, including a human individual, comprising the steps of administering to the individual a therapeutically effective amount of a first active agent which is an inhibitor of Hsp27 activity, and administering to the individual a therapeutically effective amount of a second active agent which is an antifolate. The first and second agents are administered such that both are present at therapeutically relevant levels during a common time period. In some embodiments of the invention, treatment with the Hsp27 inhibitor is commenced prior to treatment with the antifolate. Brief Description of the Drawings
Fig. 1 shows induction of Hsp27 following treatment of A549 lung cancer cells with erlotinib.
Fig. 2 shows cell viability as a function of erlotinib treatment with and without treatment with siHsp27.
Fig. 3A shows cell viability as a function of erlotinib treatment with and without treatment with Hsp27 antisense.
Fig. 3B shows the combination index indicative of synergy for the combined treatment of erlotinib and Hsp27 antisense.
Fig. 3C shows cell cycle results for treatments with erlotinib and Hsp27 antisense.
Fig. 4A shows cell viability in A549 cells overexpressing Hsp27 following erlotinib treatment.
Figs. 4B and C show flow cytometry results for A549 cells overexpressing Hsp27 following erlotinib treatment.
Fig. 5A shows change in tumor volume in A549 xenograft tumors over time with various treatments. Fig 5B shows this data in bar graph form.
Fig. 5C shows the average number of Tunel positive cells in cells treated with erlotinib, OGX-427 or both.
Fig. 6 shows cell viability as a function of pemetrexed treatment with and without treatment with Hsp27 antisense.
Fig. 7 shows expression of Hsp27 in HCC827 lung cancer cells with and without treatment with erlotinib.
Fig. 8A shows Hsp27 mRNA expression in parental and erlotinib resistant HCC827 cells.
Fig. 8B shows cell viability as a function of erlotinib concentration in parental and erlotinib resistant HCC827 cells.
Fig. 9A shows cell viability of parental HCC827 cells at different concentrations of erlotinib, with and without treatment with OGX-427.
Fig. 9B shows cell viability of erlotinib resistant HCC827 cells at different concentrations of erlotinib, with and without treatment with OGX-427. Fig. 10A shows cell viability for individual and combination therapies using pemetrexed and OGX-427 at increasing concentrations of therapeutic. Fig. 1 OB is a combination index (CI) plot of this data.
Detailed Description of the Invention
The present invention provides a method for treating cancer in a patient diagnosed as suffering from cancer. In preferred embodiments, the patient is a human patient, although the method can also be used in veterinary applications, for example in the treatment of cancer in dogs, cats and other pets.
The occurrence of elevated levels of Hsp27 in various types of cancer and the
demonstrated efficacy of Hsp27 inhibitors in multiple types of cancers is indicative of the general applicability of the present invention to cancers of many types. In general, the method will be employed with cancer types which are considered to be targets for Hsp27 therapy, including in particular those where there has been a previous determination of Hsp27
overexpression for the patient's cancer or where a selected treatment induces hHsp27 expression. Specific non-limiting examples of cancer types that may be treated using the method of the invention include breast, prostate, ovarian, uterine, non-small cell lung, bladder, gastric, liver, endometrial, laryngeal and colorectal cancers; squamous cell carcinomas such as esophageal squamous cell carcinoma, glioma, glioblastoma, melanoma, multiple myeloma and lymphoma.
In the combination therapy of the present invention, a second active agent is combined with an inhibitor of Hsp27. The second active agent is an inhibitor of EGFR tyrosinase kinase or an antifolate. The second active agent is selected to have independent therapeutic activity for the cancer to be treated in a particular individual.
Definitions
As used in the specification and claims of the present application, the term "treating" refers to performing the method steps of the invention with intention and expectation of a therapeutic benefit to the patient. It would be understood in the art that not all patients respond favorably, or to the same extent to a given treatment. Furthermore, it will be understood in the art that the results obtained for any individual cannot be compared to results for that individual in the absence of the treatment. Thus, actual therapeutic benefit is not required to fall within the scope of the concept of "treating" nor is conclusive evidence that an observed benefit arose from the treatment.
The term "Hsp27" refers to heat shock protein 27, an approximately 27 kilodalton stress- induced protein. Hsp27 is also sometimes referred to as heat shock protein beta-1 (HSPB1). The sequences of Hsp27 are known in the art for Homo sapiens (AB020027, X54079, NM_006308, NM_001540 and NM_001541), dogs (NP_001003295), cattle (NP_001020740), mice
(NP_038588) and other species.
The term "EGFR" refers to epidermal growth factor receptor (EGFR). Its activated form, phosphorylated EGFR (pEGFR), is correlated with poor prognosis in lung cancer. Selvaggi G, Novello S, Torri V. et al. Ann Oncol. 2004 Jan;15(l):28-32.
The term "NSCLC" refers to non-small cell lung cancer, a recognized subset of lung cancer.
The term "SCC" refers to squamous cell carcinoma. SCC can occur in lung tissue or in other locations in the body. SCC originating in lung tissue is a recognized subset of NSCLC
The term "AD" or "lung AD" refers to lung adenocarcinoma, a different recognized subset of NSCLC.
The term "LCC" refers to large cell lung carcinoma, a different recognized subset of NSCLC.
The term "lung cancer" when used without further labeling refers generically to NSCLC, including lung SCC, lung-AD and LCC.
The term "p-Hsp27 (Ser82)" refers to hsp27 that is phosphorylated at serine 82. Other serine residues may also be phosphorylated as described in "Regulation of HSP27 function is highly dependent on phosphorylation (Stetler et al., 2009, 2010). In response to cellular stimuli, such as oxidative stress, Hsp27 is phosphorylated at several distinct serine residues (Serl5, Ser78, and Ser82 in humans). Phosphorylation at Ser82 leads to dissociation to lighter oligomers (Theriault et al., 2004) and loss of the chaperone function of HSP27 which is associated with the nonphosphorylated structure. p-Hsp27 can suppress cell death-signaling (Bruey et al., 2000; Concannon et al., 2001 ; Benn et al., 2002; Rane et al., 2003; Lee et al., 2005; Voss et al., 2007). Cell death suppression requires phosphorylation of Hsp27 (Benn et al., 2002). Thus, measurement of increased levels p-Hsp27 is an indication that Hsp27 is acting in a
cytoprotective manner, while a decrease in p-Hsp27 in targeted cells is indicative of a therapeutic enhancement.
The term "PARP" refers to poly-(ADP-ribose) polymerase, a marker of apoptosis and a caspase substrate.
Caspase 3 refers to an enzyme that is a member of the cysteine-aspartic acid protease (caspase) family associated with the apoptotic pathway. Cleaved caspase 3 is the large fragment (17/19 kDa) of activated caspase-3 resulting from cleavage adjacent to Aspl75. Antibodies are used that bind to cleaved caspase-3 but that do not recognize full length caspase-3 or other cleaved caspases. This antibody detects non-specific caspase substrates by Western blot.
The term siRNA refers to double stranded RNA or RNA and DNA species that are active to reduce expression of targeted gene. These molecules are known variously as "small interfering RNA ", "short interfering RNA" or "silencing RNA." siRNA strands are usually 20- 25 nucleotides long, although larger precursor molecules which are subject to cleavage in vivo to form the active species are within the scope of the term as used herein.
A549 cells: ATCC No. CCL-185™ is an epithelial lung carcinoma (Giard DJ, et al. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J. Natl. Cancer Inst. 51 : 1417-1423, 1973.) A549 cells are representative of NSCLC without an activating mutation in the EGFR tyrosine kinase domain. Such cells are not considered clinically sensitive to EGFR-TKI such as erlotinib and gefitinib.
HCC827: ATCC No. CRL-2868™ is an epithelial cell line derived from lung
adenocarcinoma. Growth and subculturing conditions are provided at
www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/tabid/452/Default.aspx7ATCCNu m=CRL-2868&Template=cellBiology. HCC827 is representative of NSCLC (lung
adenocarcinoma) with an activating mutation in the EGFR tyrosine kinase domain (E746 - A750 deletion). Methods
The method referred to herein as "CI" or "combination index" is a quantitative analysis based on a theorem proposed by Chou-Talalay in the 1980s (Chou TC, Talalay PA. J. Biol. Chem (1977) 252: 6438-42; Chou TC. Talalay PA. Eur. J. Biochem. (1981) 115:207-216). Software called CalcuSyn (Biosoft, Cambridge UK) is available and was used for these studies to quantify phenomena such as synergism and inhibition. The software performs multiple drug dose-effect calculations using the Median Effect methods described by T-C Chou and P. Talalay (Trends Pharmacol. Sci. 4, 450-454).
CI<1 indicates synergism
CI=1 indicates additive effect
CI>1 indicates antagonism
TUNEL: "TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining (1)". TUNEL staining relies on the ability of the enzyme terminal deoxynucleotidyl transferase to incorporate labeled dUTP into free 3'-hydroxyl termini generated by the fragmentation of genomic DNA into low molecular weight double-stranded DNA and high molecular weight single stranded DNA.
Western blots: To perform Westerns, cell ly sates were prepared under non-denaturing conditions. Briefly, the cell lysates were to immunoprecipition, and subsequently
immunoblotted using antibody and visualized via electrophoresis and autoradiogram.
Crystal Violet: Crystal violet (CV) is a triphenylmethane dye (4-[(4- dimethylaminophenyl)-phenyl-methyl]-N,N-dimethyl-aniline) also known as Gentian violet (or hexamethyl pararosaniline chloride). The CV assay is used to determine cell viability or to determine cell proliferation under different testing conditions (Davey, Hazel M.; Kell, Douglas B. Microbial Reviews (1996) 60: 4: 641-696). Inhibitors o†Hsp27
As used in the present application, the term "inhibitor of Hsp27" refers to a compound that reduces the activity of Hsp27, either by interaction with Hsp27 or its intended target, or through reduction in the amount of Hsp27 present in cells. Inhibitors of Hsp27 expression of various different types are known in the art. Examples of inhibitors includes nucleotide compounds targeting Hsp27, peptide aptamers, flavonoid inhibitors of Hsp27, antibodies that interact with Hsp27, and interferon-γ which has been shown to downregulate expression of Hsp27. The specific route of administration, the dosage level and the treatment frequency will depend on the nature of the active agent employed. In general, the therapeutic agent may be administered by intravenous, intraperitoneal, subcutaneous, topical or oral routes, or direct local tumor injection.
In accordance with some embodiments, the Hsp27 inhibitor is a nucleotide inhibitor. Examples of nucleotide inhibitors include antisense sequences, which may be full-length antisense (see Horman et al., Int. J. Cancer (1999) 82: 574-582), or shorter oligonucleotide sequences, having a length of 100 bases or less, for example 12 to 30 bases. Such antisense species are complementary to the target Hsp27 gene to an extent sufficient to achieve antisense inhibition in vivo, and may include degeneracy to take into account allelic variation. Specific oligonucleotide antisense inhibitors of Hsp27 are known in the art from US Patent Publications 2004/0127441 and 2009/0264502 which are incorporated herein by reference.
In specific embodiments, the Hsp27 inhibitor is OGX-427, an antisense oligonucleotide made by OncoGenex that is currently in clinical trials for treatment of various types of cancer. OGX-427 is a 4-12-4 2'-MOE gapmer oligonucleotide with phosphorothiolatedinternucleotide linkages which can be represented as
5'-GGGAMeCGMeCGGMeCGMeCTMeCGGMeUMeCAMeU-3' (Seq. ID No. 1) where G, A, MeC, and T represent the nucleosides 2'-deoxyguanosine, 2'-deoxyadenosine, 2'- deoxy-5-methylcytidine, and 2'-deoxythymidine, the underlined nucleosides denote 2'-0- methoxyethyl (2'-MOE) modifications of the nucleosides, and the internucleotide linkages are phosphothioatediester, sodium salts.
In other embodiments, the nucleotide Hsp27 inhibitor is a double stranded RNA species (or precursor) that operates by an siRNA mechanism to reduce expression of Hsp27. Specific RNA species for this purpose are known from US Patent Publication 2004/0127441 and Chauhan, et al. (2003) Cancer Res.63, 6174-6177. One specific siRNA inhibitor used in the examples below has the sequence GCU GCA AAA UCC GAU GAGA (Seq ID No. 2) and it is used with its complement to form the active double stranded inhibitor.
Peptide aptamers (Gilbert et al, Oncogene. 2011 Mar 21. [Epub ahead of print]); and antibodies (Tezel and Wax, J. Neuroscience 10:3553-3562 (2000) that interact with Hsp27 are also known and could serve as inhibitors of Hsp27 in accordance with the invention. Other peptides that bind to Hsp27, such as CP91 or binding fragments thereof as described in US Patent Publication No. 2007/0003555 could also be employed.
Cytokines such as interferon-γ are also known to inhibit Hsp27 and can be used as inhibitors in the present invention. Yonekura et al., Cell Death and Differentiation (2003) 10: 313-322.
Other inhibitors of Hsp27 are also known which are generally "small molecule" inhibitors. These include flavonoids such as quercetin, (Morino et al., in vivo (1997) 11: 265- 270; JP 10045572, JP 10045574, JP10036261 and JP 10036267), and biphenyl isooxazoles such as 5-(5-Ethyl-2-hydroxy-4-methoxyphenyl)-4-(4-methoxyphenyl)isoxazole (KRIBB3) (Shin et al. The Journal of Biological Chemistry VOL. 280, NO. 50, pp. 41439-41448, December 16, 2005). KRIBB3 is available commercially from Sigma- Aldrich is understood to reduce Hsp27 activity by acting as a specific inhibitor (IC50 of 50 nM) of PKC-dependent phosphorylation of Hsp27. Related compounds are described in Lee et al., Bioorg Med ChemLett. 2011 Feb l;21(3):977-9. Epub 2010 Dec 13.Berberine derivatives have also been shown to inhibit Hsp27. (EP 0 813 872) Paclitaxel has also been shown to be an inhibitor of Hsp27 expression. (Tanaka et al., Int J Gynecol Cancer. 2004 Jul-Aug; 14(4): 616-20). Nucleoside inhibitors such as brivudine also are within the scope of the invention. Inhibitors of EGFR tyrosine kinase
As used in the present application, the term "inhibitor of EGFR tyrosine Kinase rerers to inhibitors of the tyrosine kinase activity of epidermal growth factor receptor (EGFR), either by interaction with EGFR tyrosine kinase or its intended target, or through reduction in the amount of EGFR tyrosine kinase present in cells.
Examples of inhibitors of EGFR tyrosine kinase activity include known therapeutic compounds such as erlotinib (Tarceva™) and gefitinib (Iressa™) both of which are quinazoline compounds;. PKI-166; EGFR-specific and irreversible inhibitors, such as EKI-569; a PAN-HER (human EGF receptor family) reversible inhibitor, such as GW2016 (targets both EGFR and Her2/neu); and a PAN-HER irreversible inhibitor, such as CI-1033 (4-anilinoquinazoline).
EGFR-targeting antibodies and antibody fragments, and their use in treatment of cancer are known and include cetuximab (Erbitux™) and panitumumab (Vectibix™). Others are known from US Patent Publication No. 2011-0117110.
Oligonucleotide sequences (antisense, siRNA and the like) for inhibition of wild-type or mutant forms of EGFR are known from US Patent Publication No. 2012-0022132; US Patent Publication No. 2009-0118208 and US Patent Publication No. 2003-0170891.
Antifolates
Antifolates are folate-analogue metabolic inhibitors. They may interfere with one or more of three enzymes involved in purine and pyrimidine synthesis— thymidylate synthase (TS), dihydrofolate reductase (DHFR), and glycinamide ribonucleotide formyltransferase (GARFT). Specific antifolates used in cancer therapy include methotrexate, pemetrexed, pralatrexate, and raltitrexed.
Combinations of Agents
Each of the inhibitors of Hsp27 may be used individually or in combination with one or more of the other inhibitors. Each of these inhibitors or combinations may be used with any second agent that is an inhibitor of EGFR tyrosine kinase activity or an antifolate, with the proviso that the second agent is selected for utility in the treatment of the particular cancer with which the individual treated is diagnosed. In addition, combinations within the scope of the invention may be further combined with additional therapeutics effective
treated.
Formulation and Administration
The therapeutic agents of the invention are suitably formulated in one or more pharmaceutically acceptable carriers of a type consistent with the intended mode of
administration and the specific Hsp27 and second agent. In general, the Hsp27 and second agents can be administered as an injectable liquid, an oral or aerosol composition, and may be administered systemically (for example intravenous, intramuscular, or oral) or regionally to an area harboring a cancer to be treated (for example intra-nasal, intra-tumoral, intra- tracheal, intrapleural and topical).
Inhalation strategies for antisense therapeutics are known, for example from Karras et al, Drug Discovery Today: Therapeutic Strategies (2006) 3(3): 335-341 and Crosby et al, J Pharmacol, and Exp. Therapeutics,. (2007) 321: 938-946. See also, US Patent Publication No. 2006/0003954. Other modes of regional administration to the lungs include intra-pleural injection and intra-tracheal administration.
In some embodiments of the invention, the Hsp27 inhibitor is an antisense
oligonucleotide. Oligonucleotide therapeutics are commonly being tested at treatment levels of 1 to 500 mg/m2/day and preferred treatment levels are selected to balance toxicity with therapeutic benefit. Such levels are appropriate for use in the therapeutic combinations of the invention. One specific oligonucleotide product is "OGX-427" (Seq ID No. 2) which is currently provided to human patients at about 600mg per patient in a 25 mg/niL concentration formulated as a mannitol-phosphate buffer solution (pH 7.4) for IV administration. OGX-427 dosing solutions are administered intravenously using an infusion pump. In some situations, the administration will be preceded or accompanied by administration of an antihistamine.
Antisense oligonucleotides may also be administered in specific carrier, such as that described in US Patent Publication No. US 2009/0142413 Al.
Other components used in the therapeutic combinations are used at levels consistent with usages for other purposes. Lower levels may be possible because of the effectiveness of the combination, or where more than one inhibitor of a given type is included. Treatable Cancers
Some treatment-naive cancers have been found to express Hsp27. In addition, Hsp27 expression may arise as a response to treatment with chemotherapy or radiotherapy. The method and the therapeutic combination of the invention are applicable to the treatment of cancers that inherently or as a consequence of prior or concurrent chemotherapy or radiation treatments express Hsp27. Specific cancers include, without limitation, prostate, bladder, lung, breast, osteosarcoma, pancreatic, colon, testicular, colorectal, urothelial, renal cell,
hepatocellular, leukemia, lymphoma, and ovarian cancer, melanoma, central nervous system malignancies, and squamous cell carcinoma.
In some embodiments of the invention, the cancer is a treatment-naive cancer that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer. Thus, by way of example only, the level of Hsp27 expression in prostate cancer cells would be compared to an average value for non-cancerous prostate tissue.
In some embodiments of the invention, the cancer is one that been previously treated in the individual by chemotherapy. In this case, the cancer is preferably one that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer and/or that expresses Hsp27 as a level greater than levels in the individual prior to the treatment.
In some embodiments of the invention, the cancer is one that been previously treated in the individual by radiotherapy. In this case, the cancer is preferably one that that expresses Hsp27 at a level that is greater to a statistically significant extent than the amount of expression of Hsp27 in non-cancerous cells of the same type as the cancer and/or that expresses Hsp27 as a level greater than levels in the individual prior to the treatment.
In each of these embodiments, the therapeutic combination as described above can be used.
As shown further in the examples, specific cancers for which the invention is particularly useful are non-small cell lung cancers in some patients possessing an activating mutation in the EGFR gene, NSCLC can be treated using EGFR-TKIs such as erlotinib and gefitinib. However, almost without exception, these NSCLC patients develop acquired resista
treatment proceeds, apparently as a result of the development of a second mutation. ( ao, w ; Miller, VA; Politi, KA; Riely, GJ; Somwar, R; Zakowski, MF; et al. Acquired resistance of NSCLC to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med (2005);2:e73.) Treatment of A549 NSCLC cells with erlotinib induces Hsp27 expression and the knock down of this expression results in enhanced cytotoxic effect and apoptosis. (See, Experimental results, A and B, herein) Thus, for treatment of NSCLC, the combination therapy of the EGFR-TKI and the Hsp27 inhibitor provides two benefits: First , it enhances the initial activity of the EGFR-TKI so that more effectiveness will be obtained before the onset of resistance. Second, it maintains the efficacy of the drug for a longer period of time, even after the resistance causing mutations have begun to appear. Erlotinib induces also expression of Hsp27 in HCC827 cells which are representative of NSCLC (lung
adenocarcinoma). (Experimental Results, G, herein). Suppression of Hsp27 results in enhanced efficacy of erlotinib in these cells. Greater levels of Hsp27 expression were observed in erlotinib resistant cell lines derived from HCC827 than in the parent strain. This may be drug resistance arising due to a second mutation in the EGFR thymidine kinase , reducing the sensitivity of the EGFR-TK to the drug, or may be a separate mechanism of drug resistance in which the cells produce higher levels of Hsp27. Regardless of mechanism, enhancement of drug activity continues in cancers that develop resistance to erlotinib. (See Experimental results, J, herein).
Beneficial Effects of the Invention
Compounds such as inhibitors of EGFR tyrosine kinase activity and antifolates are effective as chemotherapy agents because they disrupt important metabolic functions of the living cells and they can therefore have substantial detrimental side effects. While these side effects are tolerable to some extent because of the seriousness of the condition being treated (cancer), reduction in side effects is always desirable and the dosage used often reflects a balancing between these two competing interests. The present invention permits the usage of substantially lower dosages of the second agent with the same degree of efficacy. Since Hsp27 inhibitors can have low toxicity (for example OGX-427 (Seq. ID No. 1 ) was found to have no dose-limiting toxicity in a Phase I clinical study of patients with castrate resistant prostate cancer, breast cancer, ovarian cancer or non-small cell lung cancer), this r
receive comparable therapeutic effect with fewer side effects, or greater therapeutic Denerit rrom a high dose with the same level of side effects.
Furthermore, while both Hsp27 inhibitors and the second agent have individual beneficial activity, the combined benefit was synergistic at least at some dosage levels. Thus, the result of the invention is not merely the additive effect of two known therapeutics.
Experimental Results
A. Erlotinib induces expression of Hsp27
Experimental results showed that erlotinib induces Hsp27 expression in A549 lung cancer cells. A549 lung cancer cells were exposed to 10 μΜ Erlotinib and the amount of p- Hsp27 (ser82), Hsp27, p-EGFR (Tyr 1068), EGFR, cleaved PARP (an indicator of apoptosis) were determined by Western Blot after 1, 6, 24. 48 and 72 hours of exposure. Increasing amounts of EGFR, Hsp27 and p-Hsp27 were observed over time. p-EGFR was initially present but was not observed following treatment. Cleaved PARP was observed at all times following 24 hours of treatment.
The experiment was repeated using 48 hours of treatment time and erlotinib at levels of 2.5, 5 and 10 μΜ. Qualitatively similar results were observed at all concentration.
Finally cells were treated with 5 or 10 M Erlotinib for 24 or 48 hours. Hsp27 mRNA expression was analyzed by quantitative RT-PCR. Hsp27 mRNA levels were normalized to levels of GAPDH mRNA. Bars, SD. ** and *, differ from control (P < 0.01 and P < 0.05, respectively). The results are shown in Fig. 1. As indicated, both levels of erlotinib resulted in increased Hsp27 expression.
B. Hsp27 knockdown enhances erlotonib effectiveness
It was also shown that Hsp27 knockdown enhances cytotoxic effect and apoptosis after treatment with erlotinib. Cells were transfected with 20 nM Hsp27 siRNA (Seq ID No. 2) or a scrambled siRNA duplex containing 5 '-CAGCGCUGACAACAGUUUCAU-3' SEQ ID NO: 3 for 1 day, and treated with varying concentrations of erlotinib for 48 hours. Cell viability was determined by crystal violet assay. The results are summarized in Fig. 2. As shown, at each concentration, the reduction in cell viability was greater when both agents
of the combination decreased cell viability with 2.5 μΜ erlotinib more that enotiniD alone at 4 times the concentration.
Protein expression was also analyzed after 48 hours exposure to 10 μΜ erlotinib. Hsp27 and p-Hsp27 were both absent in cells treated with the siHsp27 but not the scrambled control. P- EGFR was lower in cells treated with the combination that in cells treated with siHsp27 alone. Cleaved PARP was greater in cells with the combined treatment than in cells with either treatment alone.
Combined treatment of A549 cells with OGX-427 (Seq ID No. 1) and erlotinib was found to be synergistic. A549 cells were transfected with 50nM OGX-427 (Seq. ID No. 1) or a scrambled control on two consecutive days and then treated with 10 μΜ erlotinib for 72 hours after the second transfection. Cell viability was determined using a crystal violet assay. The results are shown in Fig. 3 A.
The synergistic effect of combination treatment with Erlotinib and OGX-427 in vitro was assessed using combination index (CI) values calculated by CalcuSyn software were assessed in A549 cells treated for 72 hours with OGX-427 alone, Erlotinib alone or combined treatment. The CI for inhibitory concentration (IC) 50, IC75 and IC90 was 0.9, 0.7 and 0.7, respectively, indicative of a synergistic effect of this combined treatment at all levels of erlotinib tested (See Fig. 3B).
To evaluate the apoptotic rate after combination treatment with Erlotinib and OGX-427, cells were treated as with described above. Protein expression was analyzed by western blotting. Cleaved caspase-3 and cleaved PARP were elevated in cells with the combined treated, while p- EGFR, Hsp27 and p-Hsp27 were reduced. Flow cytometry was used to quantify the percentage of cells in each cell cycle phase. (Fig. 3C; Bars, SD. **, differ from ScrB (P < 0.01).)
C. Hsp27-overexpression protects from cell death induced by Erlotinib in A549 cells.
A547 cells overexpressing Hsp27 (Hsp27 OE A549) cells were established and protein expression was confirmed by western blotting. The effect of overexpression of Hsp27 protein on the sensitivity to Erlotinib in A549 cells was tested using the. Hsp27 OE A549 cells and control vector-transfected (Empty A549) cells were treated with the indicated cor
for 72 hours. The cell viability was determined by crystal violet assay. Fig. A tsars, su.
and *, differ from Empty A549 (P < 0.01 and P < 0.05, respectively).
A decreased apoptotic rate after Erlotinib treatment in cells with Hsp27 overexpression was also observed. Hsp27 OE A549 and Empty A549 cells were treated with varying levels of erlotinib. Protein levels were analyzed by western blotting and higher levels of cleaved PARP were observed in the empty A549 cells. The cells undergoing apoptosis (subGO/Gl fraction) were quantified using flow cytometry. Figs. 4B and C. (Bars, SD. *, differ from Empty A549 (P < 0.05)).
D. Hsp27-overexpression protects from cell death induced by erlotinib in A549 cells.
Western blotting technique was used to assess relative levels of certain proteins in A549 and A549 over-expressing (OE) cell lines cultured in the present of erlotinib at concentrations of 0 to 20 micromolar after 72 hours. Cleaved PARP, p-EGFR(Tyrl068), EGFR, p-Hsp27(Ser82), Hsp27, and Beta-actin were assayed. A decrease in p-EGFR(Tyrl068) in the presence of erlotinib for both cell lines, and a dose-dependent increase in p-Hsp27(Ser82) in the A549 OE cell line were observed. Cleaved PARP increased in both cell lines as doses of erlotinib increased, indicating an increase in apoptosis.
The in vitro results were confirmed using an in vivo test on A549 xenograft tumors. A549 cells were inoculated s.c. and when tumors reached 50-100 mm3, mice were treated with ScrB control oligodeoxynucleotide (ScrB) + diluent, ScrB + Erlotinib, OGX-427 + diluent or OGX-427 + Erlotinib Tumor volume was monitored for 7 weeks after treatment. As shown in Fig. 5A, the combined treatment substantially reduced tumor growth as compared to either treatment individually. Each data point represents the mean tumor volume in each group containing 8 mice + SEM. *, differ from ScrB + diluent, ScrB + Erlotinib or OGX-427 + diluent treatment group (P<0.05). B, tumors were collected after 49 days. Hsp27, ph-Hsp27 (Ser82), EGFR, p-EGFR, Ki-67 and TUNEL were evaluated by immunohistochemical analysis (original magnification: x200). A bar graph recapitulating this same information illustrates the percent of volume change showing the greatest effects when erlotinib and OGX-427 were used together (Fig 5 B). In each cluster, the 8 lines represent the % tumor volume change on the 1st through 8 days. As can be seen, tumor size begins to decrwase arlier in the com
erlotinib treatment.
After the seven week treatment, mice were sacrificed and tumors excised for assessment. Tissue preparations were subjected to TUNEL staining, and the results are shown in Figure 5C More signs of apoptosis were observed for OGX-427 and erlotinib combined than any of the other three groups.
E. Pemetrexed induces Hsp27 expression
Experimental results showed that Hsp27 is induced in A549 cells. A549 cells were cultured as previously described by others (Van Schaeybroeck, Sandra; Kyula, Joan; Kelly, Donal M; et al. Mol Cancer Ther (2006) 5; 1154), and treated with pemetrexed at a
concentration of 0, 0.25, 0.5, and 1.0 microMoles for a 48 hour period. Western gels were run on cell protein prepared under standard non-denaturing conditions, and relative levels of p- Hsp27(Ser82), Hsp27, cleaved PARP, and beta-actin were detected using commercial antibodies from Cell Signalling (Danvers, USA). Protein levels from zero to 48 hours for a 1 microMolar pemetrexed exposure were measured. The signals for pHsp27 and cleaved PARP increase with dose and are greatest at 48 hours, providing evidence that apoptosis and Hsp27 activation are increased in the presence of pemetrexed.
The dose effect of pemetrexed at concentrations of from 0 to 1 micromole was measured on the same proteins as assayed in the above time course experiment. pHsp27 and cleaved PARP levels show similar patterns.
F. Hsp27 knockdown enhances pemetrexed effectiveness
OGX-427 was also shown to enhance the antitumor effect of premetrexed in A549 cells. A549 cells were treated 50nM OGX-427 on 2 consecutive days and incubated with varying concentrations of Pemetrexed for 72 h . 72 h later, cell viability was determined by crystal violet assay. The results are shown in Fig. 6. (** p<0.01) Reduction in cell viability comparable to 1 μΜ pemetrexed alone were obtained using the combination therapy and only 0.1 μΜ, a tenfold reduction. Protein levels were also determined in these cells by immunoblott
PARP was observed, indicating that the Hsp27 inhibitor increases apoptotic rates as comparea to pemetrexed alone.
G. Hsp27 is overexpressed in NSCLC of different subtypes
Distribution of Hsp27 positive and negative cells (n=440) was assessed in a variety of human lung cancer tissues, namely squamous cell lung carcinoma (SCC), lung adenocarcinoma (AD), and large-cell lung carcinoma (LCC). Assays for Hsp27 and p-Hsp27 are available commercially from Cell Signalling, Danvers USA, and Perkin-Elmer (USA).
Figure imgf000020_0001
Thus, the reduction in Hsp27 generally and in combination with a second agent used in the treatment of particular types of lung cancer is beneficial in a majority of human NSCLC patients.
H. Erlotinib Induces Hsp27 in HCC827 Cells
Lung cancer cell line HCC827 was cultured and exposed, under otherwise normal conditions, to from 0 to 50 micromolar erlotinib, and the cells were lysed under non-denaturing conditions and protein content was subjected to Western blotting technique.
In HCC827cells, erlotinib at 2.5 nanomolar concentration generated a decrease in p- EGFR (Tyr068) and an increase in p-Hsp27(ser82) as compared to non-treated cells, while protein levels for P-Hsp27(ser82), Hsp27 were increased. The increase is in Hsp27 is shown graphically in Fig. 7. I. Overexpression of Hsp27 in HCC827 Cell Line Coincides witn esistance to Erlotinib
Erlonitib-resistant HCC827 cells (Re-HCC827 #l,-#2, and -#3) were established by exposure to erlotinib and protein expression was confirmed by Western blotting. The three new cell lines were resistant to erlotinib treatment and overexpressed Hsp27 (Fig. 8A), and had elevated levels of p-Hsp27 as compared to the parent cell lines. The graph in Fig.8 B shows cell viability for the parental HCC827 cell line and the three resistant derivations (Re#l, #2, and #3) under conditions of increasing concentrations of erlotinib (0.1 to 1000 nanoMole). The resistant cell lines are able to overcome the effects of erlotinib to a greater degree than the parental cell line can.
J. Hsp27 Suppression Sensitizes both HCC827 Parental and Resistant Cells to Erlotinib
Two lung cancer cell lines, HCC827 parental and resistant (HCC827 #3) were cultured according to supplier's directions in RPMI 1640 and 10% FCS. OGX-427 was added to
HCC827 and Re-HCC827 #3 cell cultures in the presence of increasing doses of erlotinib (0, 2.5, 5, 10 and 50 nM). ScrB control oligodeoxynucleotide was used as a control for the OGX-427. Cell viability was measured using crystal violet technique. The results were expressed as a percentage of the cell viability in the presence of Oligofectamine™ control in Figs 9A and 9B for the parental and resistant cell lines respectively. The second, paler bars in the graphs are the OGX-427 treated cells, while the darker bars represent the cell viability of cells treated with ScrB control oligodeoxynucleotide.
Western blotting technique was used to assess the mechanisms of sensitization for both the parental and resistant HCC827 cell lines, under four conditions: ScrB control
oligodeoxynucleotide only, ScrB control oligodeoxynucleotide with erlotinib, OGX-427 alone, and OGX-427 with erlotinib. Levels of p-EGFR(Tyrl068), EGFR, Hsp27, Cleaved PARP, Cleaved and whole Caspase-3, and beta-actin (cell protein control) were detected. Similar results were observed for both parental and resistant cell lines treated with both OGX-427 and erlotinib, with decreases in p-EGFR (Tyrl068) and Hsp27 as compared t
while protein levels for clraved Parp andf claved caspase-3 were increasea
K. Hsp27 suppression sensitizes A549 mock and OE to erlotinib
A Western blot was performed on A549 cells treated with ScrB control
oligodeoxynucleotide, ScrB control oligodeoxynucleotide with erlotinib, OGX-427 alone, and OGX-427 with erlotinib. Western blotting technique was used to assay levels of P- EGFR(Typrl068), EGFR, p-Hsp27(Ser82), HSP27, cleaved PARP, cleaved and whole Caspase- 3, and beta-actin. Cleaved PARP and cleaved Caspase-3 levels were highest under the influence of the OGX-427 and erlotinib combination, suggesting that the apoptotic effect was greatest under those conditions.
L. Hsp27 Suppression with Pemetrexed Treatment
The synergistic effect of combination treatment with pemetrexed and OGX-427 was assessed in A549 cells. Cells were treated with ScrB control oligodeoxynucleotide, OGX-427 alone, pemetrexed with ASO control, and OGX-427 and pemetrexed together. An additional control was Oligofectamine™ transfecting agent alone. The cells were treated with OGX-427 for 2 consecutive days, and then incubated with pemetrexed for 72 hours. Another 72 hours later, cell viability was determined by crystal violet assay. The relative amounts of OGX-427 and pemetrexed was maintaed constant as the total amount of treatment agent was increased.
The concentrations were:
Figure imgf000022_0001
The results are summarized in Fig. 10A. As shown, cell viability after was lowest in tne ceil to which combined OGX-427 and erlotinib treatment was applied.
When the values were graphed using combination index (CI) values calculated by CalcuSyn™ software. The combination index (CI) for effective concentrations (EC) ED75 and ED90 were indicative of a synergistic effect of this combined treatment (See Fig. 10B).
All of the publications referred to herein are incorporated herein by reference in their entirety.

Claims

Claims:
1. A method for treating cancer in an individual, including a human individual, comprising the steps of
administering to the individual a therapeutically effective amount of a first active agent which is an inhibitor of Hsp27 activity, and
administering to the individual therapeutically effective amount of a second active agent which is an inhibitor of EGFR tyrosine kinase activity or an antifolate.
2. The method of claim 1 , wherein the second agent is an inhibitor of EGFR tyrosine kinase activity.
3. The method of claim 2, wherein the second agent is a quinazoline.
4. The method of claim 3, wherein the second agent is erlotinib.
5. The method of claim 3, wherein the second agent is gefitinib.
6. The method of claim 2, wherein the second agent is a monoclonal antibody.
7. The method of claim 1 wherein the second agent is an antifolate.
8. The method of claim 7, wherein the second agent is pemetrexed.
9. The method of any one of claims 1 to 8, wherein the first agent is administered before the second agent.
10. The method of any one of claims 1 to 9, wherein the first agent is an oligonucleotide therapeutic.
11. The method of claim 10, wherein the first agent is an antisense oli
12 The method of claim 11, wherein the first agent is OGX-427 (Seq ID No. 1).
13. The method of claim 10, wherein the first agent is an siRNA.
14. The method of claim 13, wherein the first agent comprises Seq ID no. 2 and a complementary strand.
15. The method of any one of claims 1 to 14, wherein the cancer is prostate, bladder, lung, breast, osteosarcoma, pancreatic, colon, testicular, colorectal, urothelial, renal cell,
hepatocellular, leukemia, lymphoma, ovarian, melanoma, central nervous system malignancies, or squamous cell carcinoma.
16. The method of claim 15, wherein the cancer is lung cancer.
17. The method of any one of claims 1 to 16, wherein the individual is human.
18. A therapeutic combination for use in the method of any one of claims 1 to 17.
PCT/IB2013/050882 2012-02-02 2013-02-01 Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates WO2013114339A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP13744042.6A EP2809325A4 (en) 2012-02-02 2013-02-01 Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates
CA2861415A CA2861415A1 (en) 2012-02-02 2013-02-01 Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates
AU2013216361A AU2013216361B2 (en) 2012-02-02 2013-02-01 Combination therapy for cancer using HSP27 inhibitor and EGFR tyrosine kinase inhibitors or anti-folates
US14/372,972 US9717792B2 (en) 2012-02-02 2013-02-01 Combination therapy for cancer using HSP27 inhibitor and EGFR tyrosine kinase inhibitors or anti-folates

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261594173P 2012-02-02 2012-02-02
US61/594,173 2012-02-02

Publications (2)

Publication Number Publication Date
WO2013114339A1 true WO2013114339A1 (en) 2013-08-08
WO2013114339A4 WO2013114339A4 (en) 2013-10-03

Family

ID=48904496

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2013/050882 WO2013114339A1 (en) 2012-02-02 2013-02-01 Combination therapy for cancer using hsp27 inhibitor and egfr tyrosine kinase inhibitors or anti-folates

Country Status (5)

Country Link
US (1) US9717792B2 (en)
EP (1) EP2809325A4 (en)
AU (1) AU2013216361B2 (en)
CA (1) CA2861415A1 (en)
WO (1) WO2013114339A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016016267A1 (en) 2014-07-28 2016-02-04 Technische Universität Dresden Efficient inhibition of hsp27
EP3819006A1 (en) 2019-11-05 2021-05-12 Technische Universität Dresden Compounds with thymine skeleton for use in medicine

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007041294A2 (en) * 2005-09-29 2007-04-12 The Trustees Of Boston University Methods for sensitizing cancer cells to inhibitors
US20090281166A1 (en) * 2008-04-14 2009-11-12 The University Of British Columbia Treatment of Cancer by Inhibition of HSP27
US20090292008A1 (en) * 2002-10-02 2009-11-26 The University Of British Columbia Compositions and Methods for Treatment of Prostate and Other Cancers
WO2012080509A1 (en) * 2010-12-17 2012-06-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nucleic acids targeting tctp for use in the treatment of chemo-or hormone-resistant cancers

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH107559A (en) 1996-06-18 1998-01-13 Kureha Chem Ind Co Ltd Suppressant for synthesis of protein belonging to hsp27 family containing berberine derivative
JPH1036261A (en) 1996-07-25 1998-02-10 Kureha Chem Ind Co Ltd Aconitine-containing synthesis inhibitor of protein belonging to hsp27 family
JPH1036267A (en) 1996-07-25 1998-02-10 Kureha Chem Ind Co Ltd Synthetic suppressant containing evodiamine derivative for protein belonging to hsp27 family
JPH1045574A (en) 1996-07-30 1998-02-17 Kureha Chem Ind Co Ltd Shikonin-containing synthetic suppressor of protein belonging to hsp27 family
JPH1045572A (en) 1996-07-31 1998-02-17 Kureha Chem Ind Co Ltd Magnolol-containing synthetic suppressor of protein belonging to hsp27 family
US7087613B2 (en) * 1999-11-11 2006-08-08 Osi Pharmaceuticals, Inc. Treating abnormal cell growth with a stable polymorph of N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine hydrochloride
US7396823B2 (en) 1999-12-15 2008-07-08 Nath Rahul K Therapy inhalation involving antisense oligonucleotides for treating idiopathic pulmonary fibrosis
US20030170891A1 (en) 2001-06-06 2003-09-11 Mcswiggen James A. RNA interference mediated inhibition of epidermal growth factor receptor gene expression using short interfering nucleic acid (siNA)
WO2003099205A2 (en) * 2002-05-20 2003-12-04 Abgenix, Inc. Treatment of renal carcinoma using antibodies against the egfr
DE60332363D1 (en) 2002-10-02 2010-06-10 Univ British Columbia OLIGONUCLEOTIDES FOR THE TREATMENT OF PROSTATE CANCER AND OTHER CANCER DISEASES
WO2004033657A2 (en) 2002-10-07 2004-04-22 Antigenics, Inc. Heat shock protein binding fragments of cd91, and uses thereof
NZ551431A (en) * 2004-06-03 2010-04-30 Hoffmann La Roche Treatment with cisplatin and an EGFR-inhibitor
BRPI0610499A2 (en) 2005-04-12 2010-06-22 Intradigm Corp nucleic acid molecules, compositions and uses of said molecules
WO2007025229A2 (en) 2005-08-25 2007-03-01 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hsp27
US20090130017A1 (en) 2007-11-19 2009-05-21 Searete Llc Targeted short-lived drug delivery
BRPI0914123A2 (en) 2008-07-02 2015-10-20 Enzon Pharmaceuticals Inc rna antagonists targeting hsp27
EP2690175B1 (en) 2008-09-02 2016-12-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
EP2493929B1 (en) 2009-10-28 2017-08-16 AbbVie Biotherapeutics Inc. Anti-egfr antibodies and their uses
CN102713606A (en) * 2009-11-13 2012-10-03 无限制药股份有限公司 Compositions, kits, and methods for identification, assessment, prevention, and therapy of cancer
WO2013066485A2 (en) * 2011-08-31 2013-05-10 Asea Alexzander A Compositions and methods for treatment of metastatic cancer

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090292008A1 (en) * 2002-10-02 2009-11-26 The University Of British Columbia Compositions and Methods for Treatment of Prostate and Other Cancers
WO2007041294A2 (en) * 2005-09-29 2007-04-12 The Trustees Of Boston University Methods for sensitizing cancer cells to inhibitors
US20090281166A1 (en) * 2008-04-14 2009-11-12 The University Of British Columbia Treatment of Cancer by Inhibition of HSP27
WO2012080509A1 (en) * 2010-12-17 2012-06-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nucleic acids targeting tctp for use in the treatment of chemo-or hormone-resistant cancers

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PECTASIDES, D. ET AL.: "Testicular function in poor-risk nonseminomatous germ cell tumors treated with methotrexate, paclitaxel, ifosamide, and cisplatin combination chemotherapy.", JOURNAL OF ANDROLOGY, vol. 30, no. 3, May 2009 (2009-05-01), pages 280 - 286, XP003031299 *
See also references of EP2809325A4 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016016267A1 (en) 2014-07-28 2016-02-04 Technische Universität Dresden Efficient inhibition of hsp27
US10940150B2 (en) 2014-07-28 2021-03-09 Technische Universitaet Dresden Thymine derivatives and quinazoline-dione derivatives for the inhibition of HSP27
EP3819006A1 (en) 2019-11-05 2021-05-12 Technische Universität Dresden Compounds with thymine skeleton for use in medicine
US11214564B2 (en) 2019-11-05 2022-01-04 Technische Universität Dresden Compounds with thymine skeleton for use in medicine

Also Published As

Publication number Publication date
EP2809325A4 (en) 2015-04-01
AU2013216361A1 (en) 2014-08-21
EP2809325A1 (en) 2014-12-10
US9717792B2 (en) 2017-08-01
CA2861415A1 (en) 2013-08-08
US20150050285A1 (en) 2015-02-19
WO2013114339A4 (en) 2013-10-03
AU2013216361B2 (en) 2017-09-07

Similar Documents

Publication Publication Date Title
Lee et al. microRNA-7 increases radiosensitivity of human cancer cells with activated EGFR-associated signaling
Marshall et al. A phase I trial of a Bcl-2 antisense (G3139) and weekly docetaxel in patients with advanced breast cancer and other solid tumors
US20080145313A1 (en) Compositions and Methods for the Treatment and Prevention of Neoplastic Disorders
Pino et al. Transforming growth factor α expression drives constitutive epidermal growth factor receptor pathway activation and sensitivity to gefitinib (Iressa) in human pancreatic cancer cell lines
CN105008394B (en) Methods of treating colorectal cancer
Iwasa et al. Inhibition of insulin-like growth factor 1 receptor by CP-751,871 radiosensitizes non–small cell lung cancer cells
CN113924365A (en) Compositions and methods for treating KRAS-related diseases or disorders
US20170016002A1 (en) Method of Treating Cancer by Inhibition of DNA Repair Proteins
García-Parra et al. Poly (ADP-ribose) polymerase inhibition enhances trastuzumab antitumour activity in HER2 overexpressing breast cancer
US8026225B2 (en) Short nucleic acid molecule-mediated modulation of Aurora B kinase expression and combinations for use in anticancer therapy
Syed et al. Progesterone induces apoptosis in TRAIL‐resistant ovarian cancer cells by circumventing c‐FLIPL overexpression
AU2013216361B2 (en) Combination therapy for cancer using HSP27 inhibitor and EGFR tyrosine kinase inhibitors or anti-folates
US9822363B2 (en) Replication factor C-40 (RFC40/RFC2) as a prognostic marker and target in estrogen positive and negative and triple negative breast cancer
Pitari et al. Interruption of homologous desensitization in cyclic guanosine 3′, 5′-monophosphate signaling restores colon cancer cytostasis by bacterial enterotoxins
US10907159B2 (en) Methods of treating cancer by inhibition of DNA repair proteins using antisense based therapies
US20080318891A1 (en) Antisense oligonucleotides against thymidylate synthase
US9970012B2 (en) Replication factor C-40 (RFC40/RFC2) as a prognostic marker and target in estrogen positive and negative and triple negative breast cancer
Spankuch et al. Combinatorial application of nucleic acid-based agents targeting protein kinases for cancer treatment
WO2017091196A1 (en) Replication factor c-40 as a prognostic marker and target in breast cancer
Folly-Kossi et al. DNA2 nuclease inhibition confers synthetic lethality in cancers with mutant p53 and synergizes with PARP inhibitors
WO2022212905A1 (en) Combinations of sirnas with sirnas against sulf2 or gpc3 for use in treating cancer
Vassilakos et al. GTI-2040 displays cooperative anti-tumor activity when combined with interferon α against human renal carcinoma xenografts
WO2016132103A1 (en) Cancer prognostic

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13744042

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2861415

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 14372972

Country of ref document: US

Ref document number: 2013744042

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2013216361

Country of ref document: AU

Date of ref document: 20130201

Kind code of ref document: A