WO2006063707A2 - Novel pharmaceutical composition containing at least one dolastatin 10 derivative - Google Patents

Novel pharmaceutical composition containing at least one dolastatin 10 derivative Download PDF

Info

Publication number
WO2006063707A2
WO2006063707A2 PCT/EP2005/012992 EP2005012992W WO2006063707A2 WO 2006063707 A2 WO2006063707 A2 WO 2006063707A2 EP 2005012992 W EP2005012992 W EP 2005012992W WO 2006063707 A2 WO2006063707 A2 WO 2006063707A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
pharmaceutical composition
composition according
cancer
Prior art date
Application number
PCT/EP2005/012992
Other languages
French (fr)
Other versions
WO2006063707A3 (en
Inventor
Yuko Aoki
Masanori Miwa
Fumie Sawamura
Hiromi Tanimura
Toshikazu Yamazaki
Original Assignee
F. Hoffmann-La Roche Ag
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 F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Priority to JP2007544793A priority Critical patent/JP2008523002A/en
Priority to EP05813987A priority patent/EP1827603A2/en
Priority to AU2005315912A priority patent/AU2005315912A1/en
Priority to BRPI0519023-1A priority patent/BRPI0519023A2/en
Priority to MX2007006430A priority patent/MX2007006430A/en
Priority to CA002590431A priority patent/CA2590431A1/en
Publication of WO2006063707A2 publication Critical patent/WO2006063707A2/en
Publication of WO2006063707A3 publication Critical patent/WO2006063707A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/08Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon radicals, substituted by hetero atoms, attached to ring carbon atoms
    • 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
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • Novel pharmaceutical composition containing at least one Dolastatin 10 Derivative
  • Dolastatin 10 is known to be a potent antimitotic peptide, isolated from the marine mollusk Dolabella auricularia, which inhibits tubulin polymerization and is a different chemical class from taxanes and vincas (Curr. Pharm. Des. 1999, 5: 139-162). Preclinical studies of Dolastatin 10 have demonstrated activities against a variety of murine and human tumor cell lines in culture and in xenograft tumor models in animals. Dolastatin 10 and two synthetic dolastatin derivatives, Cemadotin and TZT- 1027 are described in Drugs of the future 1999, 24(4): 404-409.
  • Chemotherapy and more particularly combined chemotherapy is one of the means well accepted to fight cancers.
  • the combination of different antitumor agents may be a way to increase the antitumoral efficacy when a more than additive effect is revealed and/or less toxicity is observed.
  • the present invention is directed to a pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
  • trastuzumab in combination with capecitabine; trastuzumab; pertuzumab; irinotecan or a pharmaceutically acceptable salt thereof; or cisplatin for simultaneous, sequential or separate administration in the treatment of cancer;
  • R 1 and R 2 are methyl; ethyl; propyl; isopropyl or butyl;
  • R 3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having (Q-C 4 )- alkylene and wherein the phenyl group optionally may be substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; alkoxy; phenyl; phenoxy; trifluoromethyl; trifiuoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyl; amino and benzyl.
  • the present invention is directed to a pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
  • trastuzumab or pertuzumab for simultaneous, sequential or separate administration in the treatment of cancer;
  • R 1 and R 2 are methyl; ethyl; propyl; isopropyl or butyl;
  • R 3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having (C 1 -C 4 )- alkylene and wherein the phenyl group optionally may be substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; alkoxy; phenyl; phenoxy; trifiuoromethyl; trifluoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyl; amino and benzyl.
  • HerceptinTM (Trastuzumab) is a recombinant DNA-derived, humanized monoclonal antibody that selectively binds with high affinity to the extracellular domain of the human epidermal growth factor receptor 2 protein, HER2.
  • OmnitargTM (Pertuzumab; 2C4) is a humanized antibody known as HER dimerization inhibitor (HDIs).
  • HDIs block the ability of the HER2 receptor to collaborate with other HER receptor family members (HER1/EGFR, HER3, and HER4) .
  • HER1/EGFR HER1/EGFR
  • HER3, and HER4 HER dimerization inhibitor
  • interfering with HER2's ability to collaborate with other HER family receptors blocks cell signaling and may ultimately lead to cancer cell growth inhibition and death of the cancer cell.
  • HDIs because of their unique mode of action, have the potential to work in a wide variety of tumors, including those that do not overexpress HER2.
  • XelodaTM (capecitabine) is a fluoropyrimidine carbamate with antineoplastic activity.
  • CamptosarTM (TopotecinTM; CamptoTM; irinotecan hydrochloride; CPT-Il) is a semisynthetic, water-soluble derivative of camptothecin, which is a cytotoxic alkaloid extracted from plants such as Camptotheca acuminata.
  • PZ ⁇ ftnoZTM (RandaTM; BriplatinTM; czs-diamminedichloroplatinum; CDDP; cisplatin) is an inorganic Platinum (II) complex. It is widely prescribed for a variety of tumors (germ-cell, advanced bladder carcinoma, adrenal cortex carcinoma, breast cancer, head and neck carcinoma, lung carcinoma). It is administered intravenously for one to 5 days in a row, followed by a rest period of 2-3 weeks.
  • pharmaceutically acceptable salt refers to conventional acid- or base-addition salts that retain the biological effectiveness and properties of the parent compounds, for example the compounds of formula (I).
  • acid- addition salts are especially preferred and are formed from suitable non- toxic organic or inorganic acids.
  • Sample acid-addition salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, phosphoric acid and nitric acid, and those derived from organic acids such as p-toluenesulfonic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, methanesulfonic acid, ethanesulfonic acid, trifiuoro acetic acid and the like.
  • the chemical modification of a pharmaceutical compound i.e.
  • a drug) into a salt is a technique well known to pharmaceutical chemists to obtain improved physical and chemical stability, hygroscopicity, flowability and solubility of compounds. See, e.g., Ansel, H., et. al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 6th ed., 1995.
  • the pharmaceutically acceptable salts which are formed with trifiuoro acetic acid or hydrochloric acid are especially preferred.
  • a preferred embodiment of the present invention is the pharmaceutical composition as described above, comprising at least one compound of formula (I), wherein
  • R 1 and R 2 are methyl; and R 3 is as defined above.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is the compound of formula (I-A)
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with trastuzumab.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with pertuzumab.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with capecitabine.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with cisplatin.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with irinotecan hydrochloride.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab or pertuzumab.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with irinotecan hydrochloride.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab or pertuzumab.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with irinotecan hydrochloride.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab or pertuzumab.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from irinotecan hydrochloride.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of solid tumors.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
  • lung cancer preferably non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above, containing a compound of formula (I) or (I-A) together with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
  • lung cancer preferably non-small cell lung cancer (NSCLC).
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above, containing a compound of formula (I) or (I-A) together with irinotecan hydrochloride for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
  • lung cancer preferably non-small cell lung cancer (NSCLC).
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of solid tumors.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of lung cancer.
  • Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of non-small cell lung cancer.
  • the present invention relates to the treatment of cancer with therapeutic, more than additive combinations of N-[l-( ⁇ l-sec-Butyl-4-[2-(2- ⁇ [2-(3-hydroxy-phenyl)- ethyl] - methyl- carb amoyl ⁇ - 1 -methylsulfanyl-pr opyl) -pyrr olidin- 1 -yl] - 2 -methoxy-4- oxo- butyl ⁇ -methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyr amide (compound of formula (1-A)) and capecitabine, trastuzumab, pertuzumab, cisplatin and irinotecan or a pharmaceutically acceptable salt thereof; and the use of such combinations for an improved treatment of cancer, especially solid tumors, more particularly those defined above.
  • the present invention relates to the treatment of cancer with therapeutic, more than additive combinations ofN-[l-( ⁇ l-sec-But7l-4-[2-(2- ⁇ [2-(3-hydrox7-phenyl)- ethyl]-methyl-carbamoyl ⁇ -l-methylsulfanyl-propyl)-pyrrolidin-l-yl]-2-methoxy-4-oxo- butyl ⁇ -methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyramide
  • the present invention relates to the treatment of cancer with therapeutic, more than additive combinations of N-[l-( ⁇ l-sec-Butyl-4-[2-(2- ⁇ [2-(3-hydroxy-phenyl)- ethyl] -methyl-carb amoyl ⁇ - 1 -methylsulfanyl-pr opyl) -pyrr olidin- 1 -yl] -2-methoxy-4- oxo - butyl ⁇ -methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyr amide (compound of formula (1-A)) and cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; and the use of such combinations for an improved treatment of cancer, especially solid tumors, more particularly those defined above.
  • the present invention relates to the treatment of lung cancer with therapeutic, more than additive combinations of N-[l-( ⁇ l-sec-Butyl-4-[2-(2- ⁇ [2-(3- hydrox7-phenyl)-ethyl]-methyl-carbamoyl ⁇ -l-methylsulfanyl-propyl)-pyrrolidin-l-yl]-2- methoxy-4-oxo-butyl ⁇ -methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl- butyramide (compound of formula (1-A)) and irinotecan hydrochloride; and the use of such combinations for an improved treatment of lung cancer, especially non-small cell lung cancer.
  • antineoplastic effects are significantly superior to the results obtained by simple addition of the effects of each compound alone.
  • administration of each combination in accordance with the present invention resulted in an improved therapeutic index (that is, superior efficacy) in comparison to either component alone without a significant increase in toxicity, as i.e. judged by the body weight control during the studies.
  • the invention permits reduction of the amount of at least one component (in comparison the amount typically given in monotherapy) while retaining a desirable therapeutic index.
  • the amount of both components is reduced affording reduced toxicity while still retaining a desirable therapeutic index.
  • the invention relates more particularly to a method of treating cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; said method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
  • the invention relates to a method of treating cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of capecitabine, trastuzumab or pertuzumab; said, method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
  • the invention relates more particularly to a method of treating non-small cell lung cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; said method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
  • the two components: the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof may be administered simultaneously, separately or sequenced over time.
  • the compound of formula (I) or (I-A) maybe administered first or capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof may be administered first.
  • compositions as defined above comprise effective therapeutic quantities of at least the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable supports to form together or separately liquid composition(s) such as solutions or suspension, as such or in a capsule; or solid composition(s) such as compressed tablets, pills, powders and the like.
  • the preferred doses and dosage forms correspond to those typically recommended in monotherapy of each of the above-mentioned product, and are readily available to the skilled artisan from the corresponding publications.
  • the administration of the compound of formula (I) or (I-A) and the second additional component is carried out according to a regimen dependent on the type of cancer and more particularly on the type of tumors.
  • the dosage ranges for the administration of the combination of the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof according to the present invention may also vary with the administration routes, as well as the state of the patient (age, extent of the disease).
  • the two components can be administered, by identical or different administration routes. They can be administered by identical or different administration routes when they are present in separated form, and by identical administration routes when they are present in unseparated form.
  • trastuzumab pertuzumab, cisplatin or irinotecan hydrochloride are known to the skilled artisan. Their preferred doses and dosage forms are well documented and readily available to the one skilled in the art. Preferred doses according to the present invention are 5 to 50 mg/kg, preferably 15 to 30 mg/kg trastuzumab; 5 to 60 mg/kg, preferably 20 to 40 mg/kg pertuzumab; and 50 to 550 mg/kg, preferably 150 to
  • Cisplatin is used at doses between 0.5 and 10 mg/kg, preferably between 4 and 7 mg/kg; and irinotecan hydrochloride at a doses between 10 to 120 mg/kg, preferably 30 to 100 mg/kg, more preferably 60 to 90 mg/kg.
  • the maximum tolerated dose (MTD) was determined to be 6 mg/kg in the studies according to the present invention. Therefore the compound of formula (I-A) can be administered at doses between 0.1 and 6 mg/kg, preferably at doses between 1 and 6 mg/kg, more preferably at doses of 3, 4 and 6 mg/kg. It is understood that some compounds of formula (I) can show higher MTD's and therefore applicable doses of more than 6 mg/kg without departing from the spirit of the present invention.
  • the combination of 6 mg/kg (MTD) of the compound of formula (I A) plus 20 mg/kg (MTD) trastuzumab also resulted in tumor regression. Furthermore, complete tumor regression was observed in 2 out of 5 mice at 3 mg/kg and all 5 mice at 4 or 6 mg/kg of the compound of formula (I A) plus 20 mg/kg trastuzumab.
  • a particularly preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab.
  • Another particularly preferred embodiment of the present invention is the combination of 4 or 6 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab.
  • Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer.
  • the volume of tumor treated with the compound of formula (I-A) at 3, 4, or 6 mg/kg decreased significantly in comparison with that of the control; significant differences were observed on day 21 using the Tukey test (P 0.0038 at 3 mg/kg, and P ⁇ 0.0001 for 4 and 6 mg/kg).
  • tumor volumes in combination therapies of 3, 4, or 6 mg/kg of the compound of formula (I-A) with pertuzumab were compared to those of the monotherapy of the compound of formula (I-A) on day 21: every combination showed superior antitumor activity compared to that of the compound of formula (I-A) monotherapy.
  • a particularly preferred embodiment of the present invention is the combination of 4 mg/kg of the compound of formula (I-A) with 30 mg/kg pertuzumab.
  • Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer.
  • each dose (3, 4, or 6 mg/kg) of the compound of formula (I-A) in combination with 359 mg/kg capecitabine showed to be equal to that of either of the single agents in the MAXF-401 human breast carcinoma xenograft model.
  • the combination of each dose of the compound of formula (I-A) plus 539 mg/kg (MTD) capecitabine showed less favourable effects.
  • statistical analyses were performed using results on tumor volume assessed at one week after the final injection.
  • the volume of tumor treated with the compound of formula (I-A) at 3, 4, or 6 mg/kg decreased significantly (or more than that observed) in the 2.5 % DMSO control.
  • tumor volumes in combination therapies of 3, 4, or 6 mg/kg of the compound of formula (I-A) with capecitabine were compared to those of the monotherapy of the compound of formula (I-A) on day 21: combinations resulted in similar antitumor activity to that observed with monotherapy. However, the duration of observed regression was longer with combination therapy than with capecitabine monotherapy.
  • a particularly preferred embodiment of the present invention is the combination of 4 mg/kg of the compound of formula (I-A) with 359 mg/kg capecitabine.
  • Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer.
  • Antitumor activity the compound of formula (I-A) in combination with CDDP in the NCI-H460 human large cell lung carcinoma xenograft model was examined.
  • the compound of formula (I-A) was administered weekly for 3 consecutive weeks and CDDP was administered at the first day of the treatment.
  • MTD maximum tolerated dose
  • 3 mg/kg 1/2 MTD
  • the compound of formula (I-A) reduced the tumor growth by 45% and 93% respectively at day 12.
  • the dose of 5 mg/kg (1/2 MTD) and 6.7 mg/kg (2/3 MTD) CDDP reduced the tumor growth by 56% and 66% respectively at day 12.
  • Combination of 2 mg/kg of the compound of formula (I-A) and 6.7 mg/kg CDDP reduced the tumor growth by 104% which was greater than that observed with the corresponding dose of the compound of formula (I-A) or CDDP alone without toxicity judged by the body weight loss at day 12.
  • Combination of 3 mg/kg of the compound of formula (I-A) and 5 mg/kg CDDP reduced the tumor growth by 108% which was greater than the corresponding dose of the compound of formula (I-A) or CDDP alone without toxicity judged by the body weight loss at day 12.
  • a preferred embodiment of the present invention is the combination of 2 mg/kg of the compound of formula (I-A) together with 6.7 mg/kg CDDP.
  • Yet another preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) and 5 mg/kg CDDP.
  • Still another preferred embodiment of the present invention is a combination as defined above for the manufacture of medicaments for the treatment of non-small cell lung cancer.
  • Antitumor activity of the compound of formula (I-A) in combination with irinotecan was determined using the Calu-6 human lung cancer xenograft model. According to previous experiments, the maximum tolerated dose (MTD) of compound (I-A) was defined as 6 mg/kg and of irinotecan as 120 mg/kg. The compound of formula (I-A) was administered weekly for 3 consecutive weeks and irinotecan was administered weekly for 2 consecutive weeks followed by rest for a week.
  • MTD maximum tolerated dose
  • Antitumor activity of 0.375 mg/kg (1/16 MTD) or 0.75 mg/kg (1/8 MTD) of the compound of formula (I-A) in combination with 80 mg/kg irinotecan was consistent with those of the irinotecan alone at day 32.
  • the combination of the 3 mg/kg (1/2 MTD) of the compound of formula (I-A) and 80 mg/kg (2/3 MTD) irinotecan was toxic.
  • a preferred embodiment of the present invention is the combination of 1.5 mg/kg of the compound of formula (I-A) together with 80 mg/kg irinotecan hydrochloride.
  • Yet another preferred embodiment of the present invention is the use of the combination as defined above for the manufacture of medicaments for the treatment of non-small lung cancer.
  • Antitumor activity of the compound of formula (I-A) in combination with capecitabine in the HT-29 human colorectal adenocarcinoma xenograft model was examined.
  • the compound of formula (I-A) was administered weekly for 3 consecutive weeks and capecitabine was administered daily for 14 days.
  • MTD maximum tolerance dose
  • 4 mg/kg (2/3 MTD) the compound of formula (I-A) reduced the tumor growth by 76% and 101% respectively at day 31.
  • capecitabine reduced the tumor growth by 32% and 50% respectively at day 31.
  • a preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) together with 539 mg/kg capecitabine.
  • Fig. Ia Mean Tumor Volume (T. V.; mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (3.0 mg/kg) alone and in combination with trastuzumab (HerceptinTM; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
  • Fig. Ib Mean Tumor Volume (T.V.; mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (4.0 mg/kg) alone and in combination with trastuzumab (HerceptinTM; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
  • Fig. Ic Mean Tumor Volume (mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (6.0 mg/kg) alone and in combination with trastuzumab (HerceptinTM; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
  • Fig. 2a Mean Tumor Volume (T. V., mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (3.0 mg/kg) alone and in combination with pertuzumab (OmnitargTM; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30 mg/kg) as well as for the vehicle are also given.
  • Fig. 2b Mean Tumor Volume (T. V., mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (4.0 mg/kg) alone and in combination with pertuzumab (OmnitargTM; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30mg/kg) as well as for the vehicle are also given.
  • Fig. 2c Mean Tumor Volume (T.V., mm 3 ) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (6.0 mg/kg) alone and in combination with pertuzumab (OmnitargTM; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30mg/kg) as well as for the vehicle are also given.
  • Fig. 3a Tumor Volume (mm 3 ) of NCI-H460 tumor cells versus days after inoculation with the compound of formula I-A (2mg/kg) alone and in combination with cisplatin (RandaTM; CDDP; 6.7mg/kg). The data for the treatment with cisplatin alone (RandaTM; CDDP; 6.7mg/kg) as well as for the vehicle are also given.
  • Pig. 3b Tumor Volume (mm 3 ) of NCI-H460 tumor cells versus days after inoculation with the compound of formula I-A (3mg/kg) alone and in combination with cisplatin (RandaTM; CDDP; 5mg/kg).
  • RandaTM; CDDP; 5mg/kg The data for the treatment with cisplatin alone (RandaTM; CDDP; 5mg/kg) as well as for the vehicle are also given.
  • Fig. 4 Tumor Volume (mm 3 ) of Calu-6 tumor cells versus days after inoculation with the compound of formula I-A (1.5 mg/kg) alone and in combination with irinotecan hydrochloride (TopotecinTM; CPT-Il; 80 mg/kg).
  • irinotecan hydrochloride TopictecinTM; CPT-Il; 80 mg/kg.
  • the data for the treatment with irinotecan hydrochloride alone (TopotecinTM; CPT-Il; 80 mg/kg) as well as for the vehicle are also given.
  • Fig. 5a Tumor Volume (mm 3 ) of HT-29 tumor cells versus days after inoculation with the compound of formula I-A (3 mg/kg) alone and in combination with capecitabine (XelodaTM; 539 mg/kg).
  • the data for the treatment with capecitabine alone (XelodaTM; 539 mg/kg) as well as for the vehicle are also given.
  • Fig. 5b Tumor Volume (mm 3 ) of HT-29 tumor cells versus days after inoculation with the compound of formula I-A (4 mg/kg) alone and in combination with capecitabine (XelodaTM; 359 mg/kg).
  • the data for the treatment with capecitabine alone (XelodaTM; 359 mg/kg) as well as for the vehicle are also given.
  • vehicle as used herein means the solution wherein the compound of formula I-A, alone or in combination with the respective medicament, or the respective medicament alone is dissolved for subsequent application. Further specification of such solutions as well as their use and effects according to the present invention is illustrated by the following examples, which must in no way be considered as limiting the scope of the present invention.
  • the KPL-4 human breast carcinoma cell line was kindly donated by Prof. Kurebayashi, Kawasaki Medical Center.
  • the cells were cultured in Dulbecco's MEM (Sigma Chemical Co.) supplemented with 5% (v/v) fetal bovine serum (Roche Diagnostics).
  • mice One hundred forty (140) 5-week-old female athymic nude mice (BALBVc nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 ⁇ 2 0 C, relative humidity 55 ⁇ 10%) under 12 h light/ dark cycles. The mice were given sterilized CE-2 and tap water with 25 ⁇ g/mL gentamycin ad libitum. After 14 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
  • the title compound (compound of formula (1-A)) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Trastuzumab (Herceptin) was purchased from Chugai Pharmaceutical Co., Ltd. The title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration. Trastuzumab was dissolved and diluted with saline before administration. Purified Human IgG (ICN Pharmaceuticals, Inc.) was dissolved and diluted into 20 mg/mL with saline for a stock solution. Before administration, the stock solution was diluted with saline for a dosage of 20 mg/kg. Determination of antitumor activity
  • mice A single cell suspension of KPL-4 (4 x 10 6 cells per mouse), conventionally cultured in flasks, was implanted orthotopicaUy at a volume of 100 ⁇ L under the nipple of the second mammary fat pad on the right of athymic nude mice.
  • Mice bearing tumors of between 225 and 485 mm 3 were selected on day 43 after tumor implantation and were randomly divided into 8 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 340 mm 3 and 18.6 g respectively.
  • Drug administration was initiated after grouping the mice.
  • the title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2 mL/mouse) intravenously once a week for 3 consecutive weeks.
  • Trastuzumab or the IgG solution was administered (0.2 mL/mouse) intraperitoneally twice a week for 3 weeks on days 0, 4, 7, 11, 14, and 18.
  • Mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.2 mL of 20 mg/kg human IgG for trastuzumab.
  • the tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab 2 /2, where a and b represent tumor length and width, respectively.
  • TGI tumor growth inhibition
  • the KPL-4 human breast carcinoma cell line was kindly donated by Prof. Kurebayashi, Kawasaki Medical Center.
  • the cells were cultured in Dulbecco's MEM (Sigma Chemical Co.) supplemented with 5% (v/v) fetal bovine serum (Roche Diagnostics).
  • mice One hundred (100) 5-week-old female athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 + 2 0 C, relative humidity 55 + 10%) under 12 h light/ dark cycles. The mice were given sterilized CE-2 and tap water with 25 ⁇ g/mL gentamycin ad libitum. After 14 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
  • the title compound (compound of formula (1-A)) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Pertuzumab was kindly given by Roche Diagnostics GmbH. The title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration. Pertuzumab was dissolved and diluted with buffer solution before administration. Purified Human IgG (ICN Pharmaceuticals, Inc.) was dissolved and diluted into 25 mg/mL with saline for a stock solution. Before administration, the stock solution was diluted with saline for a dosage of 30 mg/kg.
  • DMSO dimethyl sulfoxide
  • saline for injection, Ohtsuka Pharmaceutical Co., Ltd.
  • Purified Human IgG ICN Pharmaceuticals, Inc.
  • a single cell suspension of KPL-4 (4.5 x 10 6 cells per mouse), conventionally cultured in flasks, was implanted orthotopically at a volume of 100 ⁇ L under the nipple of the second mammary fat pad on the right of athymic nude mice.
  • Mice bearing tumors of between 190 and 356 mm 3 were selected on day 18 after tumor implantation and were randomly divided into 9 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 226 mm 3 and 20 g, respectively.
  • Drug administration was initiated after grouping the mice.
  • the title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2mL/mouse) intravenously once a week for 3 consecutive weeks.
  • Pertuzumab or the IgG solution was administered (0.2 mL/mouse) intraperitoneally once a week for 3 weeks on days 0, 7, and 14.
  • Mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.2 mL of 25 mg/kg human IgG for pertuzumab.
  • the tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab 2 /2, where a and b represent tumor length and width, respectively.
  • TGI tumor growth inhibition
  • the MAXF-401 human breast carcinoma cell line was obtained from Prof. Fiebig (University of Freiburg, Germany). This cell line was maintained in mice; the tumor fragments were implanted subcutaneously and serially passaged in mice prior to use.
  • mice One hundred (100) 5-week-old female athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 ⁇ 2 0 C, relative humidity 55 + 10%) under 12 h light/dark cycles. The mice were given sterilized CE-2 and tap water with 25 ⁇ g/mL gentamycin ad libitum. After 5 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
  • the title compound and capecitabine were synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.
  • the title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration.
  • Capecitabine was dissolved and diluted with 5% Gum Arabic - 40 mM Citrate buffer (pH 6) before administration.
  • mice Ten to twenty mg (ca. 2x2x2 mm 3 ) of tumor fragments of MAXF401 were implanted subcutaneously in the right flank of each mouse. Mice bearing tumors of between 245 and 588 mm 3 were selected on day 35 after tumor implantation and were randomly divided into 11 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 401 mm 3 and 22 g respectively. Drug administration was initiated after grouping the mice. The title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2 mL/mouse) intravenously once a week for 3 consecutive weeks. Capecitabine or its vehicle was administered (0.5 mL/mouse) orally once a day for 14 consecutive days.
  • mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.5 mL of 5% Gum Arabic - 40 mM Citrate buffer (pH 6) for capecitabine.
  • the tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab 2 l2, where a and b represent tumor length and width, respectively.
  • tumor growth inhibition (TGI) was calculated using the formula (1-T/C) x 100 (%), where T (treated group) and C (control group) represent the mean tumor volume change.
  • ED 50 values were calculated from values of tumor growth inhibition on day 21 using the XL fit program.
  • MTD was defined as the dose with neither lethality nor more than 20% body weight loss.
  • the NCI-H460 human large cell lung carcinoma cell line was purchased from American Type Culture Collection (Rockville, MD, U.S.A.). The cells were cultured in RPMI- 1640 medium (cat. # R8758, lot 12K2310, exp 01/03; lot 82K2497, exp 08/03; Sigma- Aldrich corporation, MO, U.S.A.) supplemented with 10% (v/v) fetal bovine serum (cat. # 047115, lot # 31300124; Roche Diagnostics KK, Tokyo, Japan).
  • mice Sixty of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 8 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
  • a single cell suspension of NCI-H460 (5.6 x 106 cells per mouse) was inoculated subcutaneously into the right flank of each mouse.
  • the tumor volume was estimated by using the equation ab 2 /2, where a and b represent tumor length and width, respectively.
  • Mice bearing tumors of between 150 and 245 mm 3 were selected on day 7 after tumor inoculation and were randomly divided into 8 groups, consisting of 5 mice each.
  • Mean tumor volume and body weight of the selected mice were 187 mm 3 and 24.3 g, respectively.
  • Drug administration was initiated after mice were divided into groups.
  • the title compound (2 mg/kg or 3 mg/kg), CDDP (5 mg/kg or 6.7 mg/kg) and each vehicle were administered intravenously. Tumor volume and body weight of each mouse were measured twice a week.
  • That dose was defined as toxic when at least one dead mouse was observed during the administration period or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period.
  • Tumor growth inhibition was calculated using the equation (1- ⁇ T/ ⁇ C) x 100, where ⁇ T represents the difference in tumor volume from the initial treatment date of the treated group and ⁇ C represents the difference in tumor volume from the initial treatment date of the vehicle group.
  • the CaIu- 6 human lung carcinoma cell line was purchased from American Type Culture Collection (MD, U.S.A.). The cells were cultured in Eagle's MEM ( Sigma- Aldrich corporation, MO, U.S.A.) supplemented with 0.1 milli mol/L Non-essential amino acids (Invitrogen corporation, CA, U.S.A.), 1 milli mol/L sodium pyruvate (Invitrogen corporation, CA, U.S.A.), and 10% (v/v) fetal bovine serum (Roche Diagnostics KK, Tokyo, Japan).
  • Eagle's MEM Sigma- Aldrich corporation, MO, U.S.A.
  • 0.1 milli mol/L Non-essential amino acids Invitrogen corporation, CA, U.S.A.
  • 1 milli mol/L sodium pyruvate Invitrogen corporation, CA, U.S.A.
  • 10% (v/v) fetal bovine serum (Roche Diagnostics KK, Tokyo, Japan).
  • mice 60 of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 8 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
  • the title compound was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Irinotecan (TopotecinTM, lot. No. EQAZF13) was obtained from Daiichi Pharmaceutical Co., Ltd. (Tokyo, Japan). The title compound was dissolved in dimethyl sulfoxide (DMSO) and diluted into 2.5 % DMSO solution with saline (For Injection; Ohtsuka Pharmaceutical Industry Co., Ltd., Tokushima, Japan) at the date of administration. Irinotecan was diluted with saline at the date of administration.
  • DMSO dimethyl sulfoxide
  • saline Formula Injection; Ohtsuka Pharmaceutical Industry Co., Ltd., Tokushima, Japan
  • a single cell suspension of Calu-6 (5.7 x 10 6 cells per mouse) was inoculated subcutaneously into the right flank of each mouse.
  • the tumor volume was estimated by using the equation ah 2 12, where a and b represent tumor length and width, respectively.
  • Mice bearing tumors of between 181 and 326 mm 3 were selected on day 11 after tumor inoculation and were randomly divided into 10 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 251 mm 3 and 24.7 g respectively.
  • Drug administration was initiated after mice were randomized and divided into groups. The title compound (0.375 mg/kg; 0.75 mg/kg; 1.5 mg/kg or 3 mg/kg), irinotecan (80 mg/kg) and each vehicles were administered intravenously.
  • Tumor volume and body weight of each mouse were measured twice a week. That dose was defined as toxic when at least one dead mouse was observed during the administration period or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period.
  • Tumor growth inhibition was calculated by using the equation (1- ⁇ T/ ⁇ C) x 100, where ⁇ T represents the tumor volume difference from the initial treatment date of treated group and ⁇ C represents the tumor volume difference from the initial treatment date of vehicle group.
  • the HT-29 human colorectal adenocarcinoma cell line was purchased from American Type Culture Collection (MD, U.S.A.). The cells were cultured in McCoy's 5a medium (Sigma- Aldrich Corporation, MO, U.S.A.) supplemented with 10% (v/v) fetal bovine serum (Roche Diagnostics KK, Tokyo, Japan). Animals
  • mice Sixty of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 9 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
  • the title compound (compound of formula I-A) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Capecitabine (Lot. No. 26954-190A- MIL) was purchased from F. Hoffmann-La Roche Ltd (Basel, Switzerland). The compound of formula I-A was dissolved in dimethyl sulfoxide (DMSO; Wako Pure
  • a single cell suspension of HT-29 (1.1 x 10 7 cells per mouse) was inoculated subcutaneously into the right flank of each mouse.
  • the tumor volume was estimated by using the equation ab 2 /2, where a and b represent tumor length and width, respectively.
  • Mice bearing tumors of between 172 and 282 mm 3 were selected on day 10 after tumor inoculation and were randomly divided into 7 groups, consisting of 6 mice each. Mean tumor volume and body weight of the selected mice were 217 mm 3 and 25.0 g respectively.
  • Drug administration was initiated after mice were divided into groups.
  • the compound of formula I-A (3 mg/kg or 4 mg/kg) and its vehicle were administered intravenously.
  • Capecitabine (359 mg/kg or 539 mg/kg) and its vehicle were administered orally.
  • Tumor volume and body weight of each mouse were measured twice a week.
  • the dose was defined as toxic when at least one dead mouse was observed during the administration period, or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period.
  • Tumor growth inhibition was calculated using the equation (1- ⁇ T/ ⁇ C) x 100, where ⁇ T represents the difference in tumor volume from the initial treatment date of the treated group and ⁇ C represents the difference in tumor volume from the initial treatment date of the vehicle group.

Abstract

The present invention is directed to a pharmaceutical composition, comprising at least one compound of formula (I), in combination with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan for simultaneous, sequential or separate administration in the treatment of cancer.

Description

Novel pharmaceutical composition containing at least one Dolastatin 10 Derivative
Dolastatin 10 is known to be a potent antimitotic peptide, isolated from the marine mollusk Dolabella auricularia, which inhibits tubulin polymerization and is a different chemical class from taxanes and vincas (Curr. Pharm. Des. 1999, 5: 139-162). Preclinical studies of Dolastatin 10 have demonstrated activities against a variety of murine and human tumor cell lines in culture and in xenograft tumor models in animals. Dolastatin 10 and two synthetic dolastatin derivatives, Cemadotin and TZT- 1027 are described in Drugs of the future 1999, 24(4): 404-409.
Subsequently it had been found that the Dolastatin 10 derivatives of formula (I), having various thio-groups at the dolaproine part show significantly improved anti-tumor activity and therapeutic index in human cancer xenograft models (WO 03/008378).
Chemotherapy and more particularly combined chemotherapy is one of the means well accepted to fight cancers. Thus the combination of different antitumor agents may be a way to increase the antitumoral efficacy when a more than additive effect is revealed and/or less toxicity is observed.
It has now been found that a combination of at least one Dolastatin 10 derivative, in particular at least one of formula (I) as defined below, shows more than additive effects when administered together with the known anti cancer drugs Xeloda™, Herceptin™, Omnitarg™, Camptosar™ (Topotecin™ in Japan) or Platinol™ (Randa™ in Japan). Therefore, the present invention is directed to a pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000003_0001
in combination with capecitabine; trastuzumab; pertuzumab; irinotecan or a pharmaceutically acceptable salt thereof; or cisplatin for simultaneous, sequential or separate administration in the treatment of cancer;
wherein
R1 and R2 are methyl; ethyl; propyl; isopropyl or butyl;
R3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having (Q-C4)- alkylene and wherein the phenyl group optionally may be substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; alkoxy; phenyl; phenoxy; trifluoromethyl; trifiuoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyl; amino and benzyl.
In a preferred embodiment, the present invention is directed to a pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
in combination with capecitabine, trastuzumab or pertuzumab for simultaneous, sequential or separate administration in the treatment of cancer; wherein
R1 and R2 are methyl; ethyl; propyl; isopropyl or butyl;
R3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having (C1-C4)- alkylene and wherein the phenyl group optionally may be substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; alkoxy; phenyl; phenoxy; trifiuoromethyl; trifluoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyl; amino and benzyl.
Herceptin™ (Trastuzumab) is a recombinant DNA-derived, humanized monoclonal antibody that selectively binds with high affinity to the extracellular domain of the human epidermal growth factor receptor 2 protein, HER2.
Omnitarg™ (Pertuzumab; 2C4) is a humanized antibody known as HER dimerization inhibitor (HDIs). HDIs block the ability of the HER2 receptor to collaborate with other HER receptor family members (HER1/EGFR, HER3, and HER4) . In cancer cells, interfering with HER2's ability to collaborate with other HER family receptors blocks cell signaling and may ultimately lead to cancer cell growth inhibition and death of the cancer cell. HDIs, because of their unique mode of action, have the potential to work in a wide variety of tumors, including those that do not overexpress HER2.
Xeloda™ (capecitabine) is a fluoropyrimidine carbamate with antineoplastic activity.
Camptosar™ (Topotecin™; Campto™; irinotecan hydrochloride; CPT-Il) is a semisynthetic, water-soluble derivative of camptothecin, which is a cytotoxic alkaloid extracted from plants such as Camptotheca acuminata. Irinotecan and its active metabolite, SN-38, inhibit the action of Topoisomerase I, an enzyme that produces reversible single- strand breaks in DNA during DNA replication.
PZαftnoZ™ (Randa™; Briplatin™; czs-diamminedichloroplatinum; CDDP; cisplatin) is an inorganic Platinum (II) complex. It is widely prescribed for a variety of tumors (germ-cell, advanced bladder carcinoma, adrenal cortex carcinoma, breast cancer, head and neck carcinoma, lung carcinoma). It is administered intravenously for one to 5 days in a row, followed by a rest period of 2-3 weeks. The term "pharmaceutically acceptable salt" as used herein refers to conventional acid- or base-addition salts that retain the biological effectiveness and properties of the parent compounds, for example the compounds of formula (I). According to the present invention, acid- addition salts are especially preferred and are formed from suitable non- toxic organic or inorganic acids. Sample acid-addition salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, phosphoric acid and nitric acid, and those derived from organic acids such as p-toluenesulfonic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, methanesulfonic acid, ethanesulfonic acid, trifiuoro acetic acid and the like. The chemical modification of a pharmaceutical compound (i.e. a drug) into a salt is a technique well known to pharmaceutical chemists to obtain improved physical and chemical stability, hygroscopicity, flowability and solubility of compounds. See, e.g., Ansel, H., et. al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 6th ed., 1995.
With respect to the compounds of formula (I), the pharmaceutically acceptable salts which are formed with trifiuoro acetic acid or hydrochloric acid are especially preferred.
A preferred embodiment of the present invention is the pharmaceutical composition as described above, comprising at least one compound of formula (I), wherein
R1 and R2 are methyl; and R3 is as defined above.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is the compound of formula (I-A)
Figure imgf000005_0001
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with trastuzumab. Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with pertuzumab.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with capecitabine.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with cisplatin.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, wherein said compound of formula (I) is administered in combination with irinotecan hydrochloride.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab or pertuzumab.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered simultaneously with irinotecan hydrochloride. Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab or pertuzumab.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered sequentially with irinotecan hydrochloride.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab or pertuzumab.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the present invention is the pharmaceutical composition as defined above, whereby said compound of formula (I) is administered separately from irinotecan hydrochloride.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of cancer. Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of solid tumors.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above, containing a compound of formula (I) or (I-A) together with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above, containing a compound of formula (I) or (I-A) together with irinotecan hydrochloride for the treatment of lung cancer, preferably non-small cell lung cancer (NSCLC).
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of cancer.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of solid tumors.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer. Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of lung cancer.
Another preferred embodiment of the present invention is the use of a pharmaceutical composition as described above for the production of a medicament for the treatment of non-small cell lung cancer.
In particular, the present invention relates to the treatment of cancer with therapeutic, more than additive combinations of N-[l-({l-sec-Butyl-4-[2-(2-{[2-(3-hydroxy-phenyl)- ethyl] - methyl- carb amoyl} - 1 -methylsulfanyl-pr opyl) -pyrr olidin- 1 -yl] - 2 -methoxy-4- oxo- butyl}-methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyr amide (compound of formula (1-A)) and capecitabine, trastuzumab, pertuzumab, cisplatin and irinotecan or a pharmaceutically acceptable salt thereof; and the use of such combinations for an improved treatment of cancer, especially solid tumors, more particularly those defined above.
More particularly, the present invention relates to the treatment of cancer with therapeutic, more than additive combinations ofN-[l-({l-sec-But7l-4-[2-(2-{[2-(3-hydrox7-phenyl)- ethyl]-methyl-carbamoyl}-l-methylsulfanyl-propyl)-pyrrolidin-l-yl]-2-methoxy-4-oxo- butyl}-methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyramide
(compound of formula (1-A)) and capecitabine, trastuzumab or pertuzumab; and the use of such combinations for an improved treatment of cancer, especially solid tumors, more particularly those defined above.
More particularly, the present invention relates to the treatment of cancer with therapeutic, more than additive combinations of N-[l-({l-sec-Butyl-4-[2-(2-{[2-(3-hydroxy-phenyl)- ethyl] -methyl-carb amoyl} - 1 -methylsulfanyl-pr opyl) -pyrr olidin- 1 -yl] -2-methoxy-4- oxo - butyl}-methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl-butyr amide (compound of formula (1-A)) and cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; and the use of such combinations for an improved treatment of cancer, especially solid tumors, more particularly those defined above. More particularly, the present invention relates to the treatment of lung cancer with therapeutic, more than additive combinations of N-[l-({l-sec-Butyl-4-[2-(2-{[2-(3- hydrox7-phenyl)-ethyl]-methyl-carbamoyl}-l-methylsulfanyl-propyl)-pyrrolidin-l-yl]-2- methoxy-4-oxo-butyl}-methyl-carbamoyl)-2-methyl-propyl]-2-dimethylamino-3-methyl- butyramide (compound of formula (1-A)) and irinotecan hydrochloride; and the use of such combinations for an improved treatment of lung cancer, especially non-small cell lung cancer.
It was unexpectedly found that administration of each of capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof with N- [ 1 - ({ l-sec-Butyl-4- [2- (2- { [2-(3-hydroxy-phenyl) -ethyl] -methyl-carbamoyl}- 1- methylsulfanyl-propyl)-pyrrolidin-l-yl]-2-methoxy-4-oxo-butyl}-methyl-carbamoyl)-2- methyl-propyl]-2-dimethylamino-3-memyl-butyramide in accordance with the present invention results in improved, more than additive antineoplastic effects. This means that said antineoplastic effects are significantly superior to the results obtained by simple addition of the effects of each compound alone. Namely, administration of each combination in accordance with the present invention resulted in an improved therapeutic index (that is, superior efficacy) in comparison to either component alone without a significant increase in toxicity, as i.e. judged by the body weight control during the studies. Alternatively, the invention permits reduction of the amount of at least one component (in comparison the amount typically given in monotherapy) while retaining a desirable therapeutic index. In preferred embodiments, the amount of both components (in comparison to the amount typically given in monotherapy) is reduced affording reduced toxicity while still retaining a desirable therapeutic index.
The invention relates more particularly to a method of treating cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; said method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
Further to this, the invention relates to a method of treating cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of capecitabine, trastuzumab or pertuzumab; said, method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
The invention relates more particularly to a method of treating non-small cell lung cancer, said method comprising administering to a patient in need thereof an effective amount of a therapeutic composition containing at least a first component consisting of the compound of formula (I) or (I-A), and a second component consisting of cisplatin or irinotecan or a pharmaceutically acceptable salt thereof; said method being characterized in that said composition shows a more than additive effect with respect to the effect of each of its active components alone.
According to the method of the present invention, the two components: the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof may be administered simultaneously, separately or sequenced over time. When administered separately, the compound of formula (I) or (I-A) maybe administered first or capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof may be administered first.
Therapeutic compositions as defined above comprise effective therapeutic quantities of at least the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable supports to form together or separately liquid composition(s) such as solutions or suspension, as such or in a capsule; or solid composition(s) such as compressed tablets, pills, powders and the like. The preferred doses and dosage forms correspond to those typically recommended in monotherapy of each of the above-mentioned product, and are readily available to the skilled artisan from the corresponding publications. Preferably, the administration of the compound of formula (I) or (I-A) and the second additional component is carried out according to a regimen dependent on the type of cancer and more particularly on the type of tumors. The dosage ranges for the administration of the combination of the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof according to the present invention may also vary with the administration routes, as well as the state of the patient (age, extent of the disease).
Irrespective of the use of the administration, i.e. simultaneously, separately or sequenced over time, the two components, the compound of formula (I) or (I-A) and capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof, can be administered, by identical or different administration routes. They can be administered by identical or different administration routes when they are present in separated form, and by identical administration routes when they are present in unseparated form.
Capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan hydrochloride are known to the skilled artisan. Their preferred doses and dosage forms are well documented and readily available to the one skilled in the art. Preferred doses according to the present invention are 5 to 50 mg/kg, preferably 15 to 30 mg/kg trastuzumab; 5 to 60 mg/kg, preferably 20 to 40 mg/kg pertuzumab; and 50 to 550 mg/kg, preferably 150 to
400 mg/kg, more preferably from 300 to 400 mg/kg capecitabine. Cisplatin is used at doses between 0.5 and 10 mg/kg, preferably between 4 and 7 mg/kg; and irinotecan hydrochloride at a doses between 10 to 120 mg/kg, preferably 30 to 100 mg/kg, more preferably 60 to 90 mg/kg.
For the compound of formula (I-A) the maximum tolerated dose (MTD) was determined to be 6 mg/kg in the studies according to the present invention. Therefore the compound of formula (I-A) can be administered at doses between 0.1 and 6 mg/kg, preferably at doses between 1 and 6 mg/kg, more preferably at doses of 3, 4 and 6 mg/kg. It is understood that some compounds of formula (I) can show higher MTD's and therefore applicable doses of more than 6 mg/kg without departing from the spirit of the present invention.
In particular the following results were observed for each of the three combinations according to the present invention:
Compound of formula (I-A) with trastuzumab (Herceptin™; KPL-4 xenograft model):
Contrary to the results of monotherapy with either the compound of formula (I-A) or trastuzumab, the combination studies of 3, 4 and 6 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab resulted in significant tumor regression of longer duration (at least until day 70) at all doses, in the KPL-4 human breast carcinoma model. Palpable tumors were not present after day 28 at 3 mg/kg (1/2 MTD) or 4 mg/kg (2/3 MTD) of the compound of formula (I-A) plus 20 mg/kg (MTD) trastuzumab. Observation of the antitumor activity in mice with the combination of the compound of formula (I A) and trastuzumab was continued until day 252. The combination of 6 mg/kg (MTD) of the compound of formula (I A) plus 20 mg/kg (MTD) trastuzumab also resulted in tumor regression. Furthermore, complete tumor regression was observed in 2 out of 5 mice at 3 mg/kg and all 5 mice at 4 or 6 mg/kg of the compound of formula (I A) plus 20 mg/kg trastuzumab.
To quantify the potency of each drug in combination, statistical analyses were performed with data collected at one week after the final injection. The volume of tumor treated with the compound of formula (I A) at 3, 4, or 6 mg/kg decreased significantly in comparison with that of the 2.5 % DMSO-saline plus 20mg/kg human IgG control. In addition, tumor volumes in combination therapies of 3, 4, or 6 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab were compared to those of the monotherapy of the compound of formula (I-A) on day 21: every combination showed superior antitumor activity compared to that of monotherapy.
Based on these findings, a particularly preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab.
Another particularly preferred embodiment of the present invention is the combination of 4 or 6 mg/kg of the compound of formula (I-A) with 20 mg/kg trastuzumab.
Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer.
Compound of formula (I-A) with pertuzumab (Omnitarg™; KPL-4 xenograft model):
In these combination studies, using the KPL-4 human breast carcinoma xenograft model, significant tumor regression was observed at all dosages of the compound of formula (I-A). No evidence of tumors was detected by palpation after day 28 at 4 mg/kg of the compound of formula (I-A) plus 30 mg/kg pertuzumab. Observation of the antitumor activity in mice with the combination of the compound of formula (I-A) and pertuzumab was continued until day 133. The combination of 4 mg/kg of the compound of formula (I-A) plus 30 mg/kg pertuzumab showed potent tumor regression. Furthermore, the tumors in 3 out of 5 mice at 4 mg/kg of the compound of formula (I-A) plus 30 mg/kg pertuzumab completely regressed. To assess the potency of each drug in combination, statistical analyses were performed using results on tumor volume assessed at one week after the final injection. The volume of tumor treated with the compound of formula (I-A) at 3, 4, or 6 mg/kg decreased significantly in comparison with that of the control; significant differences were observed on day 21 using the Tukey test (P = 0.0038 at 3 mg/kg, and P < 0.0001 for 4 and 6 mg/kg). In addition, tumor volumes in combination therapies of 3, 4, or 6 mg/kg of the compound of formula (I-A) with pertuzumab were compared to those of the monotherapy of the compound of formula (I-A) on day 21: every combination showed superior antitumor activity compared to that of the compound of formula (I-A) monotherapy.
Based on these findings, a particularly preferred embodiment of the present invention is the combination of 4 mg/kg of the compound of formula (I-A) with 30 mg/kg pertuzumab.
Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer.
Compound of formula (I-A) with capecitabine (Xeloda™; MAXF-401 xenograft model):
The antitumor activity of each dose (3, 4, or 6 mg/kg) of the compound of formula (I-A) in combination with 359 mg/kg capecitabine showed to be equal to that of either of the single agents in the MAXF-401 human breast carcinoma xenograft model. The combination of each dose of the compound of formula (I-A) plus 539 mg/kg (MTD) capecitabine showed less favourable effects. To evaluate the antitumor effects, statistical analyses were performed using results on tumor volume assessed at one week after the final injection. The volume of tumor treated with the compound of formula (I-A) at 3, 4, or 6 mg/kg decreased significantly (or more than that observed) in the 2.5 % DMSO control. In addition, tumor volumes in combination therapies of 3, 4, or 6 mg/kg of the compound of formula (I-A) with capecitabine were compared to those of the monotherapy of the compound of formula (I-A) on day 21: combinations resulted in similar antitumor activity to that observed with monotherapy. However, the duration of observed regression was longer with combination therapy than with capecitabine monotherapy.
Based on these findings, a particularly preferred embodiment of the present invention is the combination of 4 mg/kg of the compound of formula (I-A) with 359 mg/kg capecitabine.
Yet another preferred embodiment of the present invention is the combination as defined above for the manufacture of medicaments for the treatment of breast cancer. Compound of formula (I- A) with cisplatin fPlatinol™, CDDP: NCT-H460 xenograft model):
Antitumor activity the compound of formula (I-A) in combination with CDDP in the NCI-H460 human large cell lung carcinoma xenograft model was examined. The compound of formula (I-A) was administered weekly for 3 consecutive weeks and CDDP was administered at the first day of the treatment. At the dose of 2 mg/kg (1/3 of maximum tolerated dose, MTD) and 3 mg/kg (1/2 MTD), the compound of formula (I-A) reduced the tumor growth by 45% and 93% respectively at day 12. At the dose of 5 mg/kg (1/2 MTD) and 6.7 mg/kg (2/3 MTD), CDDP reduced the tumor growth by 56% and 66% respectively at day 12. Combination of 2 mg/kg of the compound of formula (I-A) and 6.7 mg/kg CDDP reduced the tumor growth by 104% which was greater than that observed with the corresponding dose of the compound of formula (I-A) or CDDP alone without toxicity judged by the body weight loss at day 12. Combination of 3 mg/kg of the compound of formula (I-A) and 5 mg/kg CDDP reduced the tumor growth by 108% which was greater than the corresponding dose of the compound of formula (I-A) or CDDP alone without toxicity judged by the body weight loss at day 12.
Therefore, a preferred embodiment of the present invention is the combination of 2 mg/kg of the compound of formula (I-A) together with 6.7 mg/kg CDDP.
Yet another preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) and 5 mg/kg CDDP.
Still another preferred embodiment of the present invention is a combination as defined above for the manufacture of medicaments for the treatment of non-small cell lung cancer.
Compound of formula (I-A) with irinotecan (Camptosar™; Calu-6 xenograft model):
Antitumor activity of the compound of formula (I-A) in combination with irinotecan was determined using the Calu-6 human lung cancer xenograft model. According to previous experiments, the maximum tolerated dose (MTD) of compound (I-A) was defined as 6 mg/kg and of irinotecan as 120 mg/kg. The compound of formula (I-A) was administered weekly for 3 consecutive weeks and irinotecan was administered weekly for 2 consecutive weeks followed by rest for a week. Doses of 0.375 mg/kg, 0.75 mg/kg, 1.5 mg/kg, and 3mg/kg of the compound of formula (I-A) inhibited tumor growth, dependent on the dose by 11%, 21%, 41%, and 117% respectively, and measured at day 32 after the inoculation. At 80 mg/kg (2/3 MTD) of irinotecan, growth of the tumor was inhibited by 68% at day 32. Antitumor activity of 1.5 mg/kg (1/4 MTD) of the compound of formula (I-A) in combination with 80 mg/kg (2/3 MTD) of irinotecan was 110% at day 32. This effect is more than additive with respect to the single agents. No toxicity, as judged by body weight loss, was observed with this combination. Antitumor activity of 0.375 mg/kg (1/16 MTD) or 0.75 mg/kg (1/8 MTD) of the compound of formula (I-A) in combination with 80 mg/kg irinotecan was consistent with those of the irinotecan alone at day 32. The combination of the 3 mg/kg (1/2 MTD) of the compound of formula (I-A) and 80 mg/kg (2/3 MTD) irinotecan was toxic.
Therefore, a preferred embodiment of the present invention is the combination of 1.5 mg/kg of the compound of formula (I-A) together with 80 mg/kg irinotecan hydrochloride.
Yet another preferred embodiment of the present invention is the use of the combination as defined above for the manufacture of medicaments for the treatment of non-small lung cancer.
Compound of formula (I-A) with capecitabine (Xeloda™; HT-29 xenograft model):
Antitumor activity of the compound of formula (I-A) in combination with capecitabine in the HT-29 human colorectal adenocarcinoma xenograft model was examined. The compound of formula (I-A) was administered weekly for 3 consecutive weeks and capecitabine was administered daily for 14 days. At the dose of 3 mg/kg (1/2 of maximum tolerance dose, MTD) and 4 mg/kg (2/3 MTD), the compound of formula (I-A) reduced the tumor growth by 76% and 101% respectively at day 31. At the dose of 359 mg/kg (2/3 MTD) and 539 mg/kg (MTD) , capecitabine reduced the tumor growth by 32% and 50% respectively at day 31. Combination of 3 mg/kg (1/2 MTD) of the compound of formula (I-A) and 539 mg/kg (MTD) capecitabine reduced the tumor growth by 111%. Combination of 4 mg/kg (2/3 MTD) of the compound of formula (I-A) and 359 mg/kg (2/3 MTD) capecitabine reduced the tumor growth by 115% at day 31. Increased antitumor activity was observed with these combinations than with the corresponding dose of the compound of formula (I-A) or capecitabine alone without toxicity judged by minimum body weight loss at day 31.
Therefore, a preferred embodiment of the present invention is the combination of 3 mg/kg of the compound of formula (I-A) together with 539 mg/kg capecitabine.
Another preferred embodiment of the present invention is the combination of 4 mg/kg of the compound of formula (I-A) together with 359 mg/kg capecitabine. Yet another preferred embodiment of the present invention is the use of either of the two combinations as defined above for the manufacture of medicaments for the treatment of colorectal cancer.
Unless defined in another manner, all the technical and scientific terms and abbreviations used herein have the meanings as commonly understood by a person of ordinary skill in the field to which the invention belongs. Similarly, all publications, Patent Applications, all Patents and all other references mentioned here are incorporated by way of reference.
Figures:
Fig. Ia: Mean Tumor Volume (T. V.; mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (3.0 mg/kg) alone and in combination with trastuzumab (Herceptin™; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
Fig. Ib: Mean Tumor Volume (T.V.; mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (4.0 mg/kg) alone and in combination with trastuzumab (Herceptin™; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
Fig. Ic: Mean Tumor Volume (mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (6.0 mg/kg) alone and in combination with trastuzumab (Herceptin™; Her; 20 mg/kg). The data for the treatment with trastuzumab alone (20mg/kg) as well as for the vehicle are also given.
Fig. 2a: Mean Tumor Volume (T. V., mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (3.0 mg/kg) alone and in combination with pertuzumab (Omnitarg™; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30 mg/kg) as well as for the vehicle are also given.
Fig. 2b: Mean Tumor Volume (T. V., mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (4.0 mg/kg) alone and in combination with pertuzumab (Omnitarg™; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30mg/kg) as well as for the vehicle are also given.
Fig. 2c: Mean Tumor Volume (T.V., mm3) versus days after the initial treatment of KPL-4 tumor cells with the compound of formula I-A (6.0 mg/kg) alone and in combination with pertuzumab (Omnitarg™; 2C4; 30 mg/kg). The data for the treatment with pertuzumab alone (30mg/kg) as well as for the vehicle are also given. Fig. 3a: Tumor Volume (mm3) of NCI-H460 tumor cells versus days after inoculation with the compound of formula I-A (2mg/kg) alone and in combination with cisplatin (Randa™; CDDP; 6.7mg/kg). The data for the treatment with cisplatin alone (Randa™; CDDP; 6.7mg/kg) as well as for the vehicle are also given.
Pig. 3b: Tumor Volume (mm3) of NCI-H460 tumor cells versus days after inoculation with the compound of formula I-A (3mg/kg) alone and in combination with cisplatin (Randa™; CDDP; 5mg/kg). The data for the treatment with cisplatin alone (Randa™; CDDP; 5mg/kg) as well as for the vehicle are also given.
Fig. 4: Tumor Volume (mm3) of Calu-6 tumor cells versus days after inoculation with the compound of formula I-A (1.5 mg/kg) alone and in combination with irinotecan hydrochloride (Topotecin™; CPT-Il; 80 mg/kg). The data for the treatment with irinotecan hydrochloride alone (Topotecin™; CPT-Il; 80 mg/kg) as well as for the vehicle are also given.
Fig. 5a: Tumor Volume (mm3) of HT-29 tumor cells versus days after inoculation with the compound of formula I-A (3 mg/kg) alone and in combination with capecitabine (Xeloda™; 539 mg/kg). The data for the treatment with capecitabine alone (Xeloda™; 539 mg/kg) as well as for the vehicle are also given.
Fig. 5b: Tumor Volume (mm3) of HT-29 tumor cells versus days after inoculation with the compound of formula I-A (4 mg/kg) alone and in combination with capecitabine (Xeloda™; 359 mg/kg). The data for the treatment with capecitabine alone (Xeloda™; 359 mg/kg) as well as for the vehicle are also given.
The term "vehicle" as used herein means the solution wherein the compound of formula I-A, alone or in combination with the respective medicament, or the respective medicament alone is dissolved for subsequent application. Further specification of such solutions as well as their use and effects according to the present invention is illustrated by the following examples, which must in no way be considered as limiting the scope of the present invention.
In the following examples, the Determination of antitumor activity of the present compounds, especially the compound of formula I-A, in a variety of xenograft models was carried out according to methods described by Plowman J, Dykes DJ, Hollingshead M, Simpson-Herren L and Alley MC, in Anticancer Drug Development Guide: Preclinical Screening, Clinical Trials and Approval, Teicher B (ed), Humana Press Inc, Totowa (NJ), 1997, pp 101-125 (Chapter 6: Human Tumor Xenograft Models in NCI Drug Development). Examples
Example 1:
N-ri-({l-sec-Butyl-4-r2-(2-{r2-(3-hvdroxy-phenyl)-ethyll-methyl-carbamovU-l- methylsulfanyl-propyl)-pyrroUdin-l-yll-2-methoxy-4-oxo-butyl}-ineAyl-carbamoyl)--2- methyl-propyl1-2-dimethylamino-3-metiLyl-butyramide (compound of formula (1-A)) in combination with trastuzumab (Herceptin™)
Human tumor cell line
The KPL-4 human breast carcinoma cell line was kindly donated by Prof. Kurebayashi, Kawasaki Medical Center. The cells were cultured in Dulbecco's MEM (Sigma Chemical Co.) supplemented with 5% (v/v) fetal bovine serum (Roche Diagnostics).
Animals
One hundred forty (140) 5-week-old female athymic nude mice (BALBVc nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 ± 20C, relative humidity 55 ± 10%) under 12 h light/ dark cycles. The mice were given sterilized CE-2 and tap water with 25μg/mL gentamycin ad libitum. After 14 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
Drugs
The title compound (compound of formula (1-A)) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Trastuzumab (Herceptin) was purchased from Chugai Pharmaceutical Co., Ltd. The title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration. Trastuzumab was dissolved and diluted with saline before administration. Purified Human IgG (ICN Pharmaceuticals, Inc.) was dissolved and diluted into 20 mg/mL with saline for a stock solution. Before administration, the stock solution was diluted with saline for a dosage of 20 mg/kg. Determination of antitumor activity
A single cell suspension of KPL-4 (4 x 106 cells per mouse), conventionally cultured in flasks, was implanted orthotopicaUy at a volume of 100 μL under the nipple of the second mammary fat pad on the right of athymic nude mice. Mice bearing tumors of between 225 and 485 mm3 were selected on day 43 after tumor implantation and were randomly divided into 8 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 340 mm3 and 18.6 g respectively. Drug administration was initiated after grouping the mice. The title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2 mL/mouse) intravenously once a week for 3 consecutive weeks. Trastuzumab or the IgG solution was administered (0.2 mL/mouse) intraperitoneally twice a week for 3 weeks on days 0, 4, 7, 11, 14, and 18. Mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.2 mL of 20 mg/kg human IgG for trastuzumab. The tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab2/2, where a and b represent tumor length and width, respectively. One week after the final injection, tumor growth inhibition (TGI) was calculated using the formula (1-T/C) x 100 (%), where T (treated group) and C (control group) represent the mean tumor volume change. ED50 values were calculated from values of tumor growth inhibition on day 21 using the XL fit program. The maximum tolerated dose (MTD) was defined as the dose with neither lethality nor more than 20% body weight loss.
Statistical analysis
Packaged software (SAS preclinical package version 5.0,SAS Institute Japan, Ltd.) was used for the statistical analysis. To evaluate the potency of the drugs, values of individual tumor volumes with treatments were compared to those of the controls using the Tukey test. ED50S were calculated from values of tumor growth inhibition percentage using XL fit program #509 (MS Excel Add Version 3.0.5 Build -In: XLfit3 12, ID Business Solutions Ltd.) Example 2:
N-ri-({l-sec-Butyl-4-[2-(2-{r2-('3-hvdroxy-phenyl)-ethγl1-methyl-carbamoyl}-l- methylsulfanyl-propyl)-pyrroHdin-l-yll-2-methoxy-4-oxo-butyl}-methyl-carbamoyl)-2- methyl-propγll-2-dimeth.ylamino-3-methyl-butyramid.e (compound of formula (1-A)) in combination with pertuzumab (Omnitarg™)
Human tumor cell line
The KPL-4 human breast carcinoma cell line was kindly donated by Prof. Kurebayashi, Kawasaki Medical Center. The cells were cultured in Dulbecco's MEM (Sigma Chemical Co.) supplemented with 5% (v/v) fetal bovine serum (Roche Diagnostics).
Animals
One hundred (100) 5-week-old female athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 + 2 0C, relative humidity 55 + 10%) under 12 h light/ dark cycles. The mice were given sterilized CE-2 and tap water with 25μg/mL gentamycin ad libitum. After 14 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
Drugs
The title compound (compound of formula (1-A)) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Pertuzumab was kindly given by Roche Diagnostics GmbH. The title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration. Pertuzumab was dissolved and diluted with buffer solution before administration. Purified Human IgG (ICN Pharmaceuticals, Inc.) was dissolved and diluted into 25 mg/mL with saline for a stock solution. Before administration, the stock solution was diluted with saline for a dosage of 30 mg/kg.
Determination of antitumor activity
A single cell suspension of KPL-4 (4.5 x 106 cells per mouse), conventionally cultured in flasks, was implanted orthotopically at a volume of 100 μL under the nipple of the second mammary fat pad on the right of athymic nude mice. Mice bearing tumors of between 190 and 356 mm3 were selected on day 18 after tumor implantation and were randomly divided into 9 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 226 mm3 and 20 g, respectively. Drug administration was initiated after grouping the mice. The title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2mL/mouse) intravenously once a week for 3 consecutive weeks. Pertuzumab or the IgG solution was administered (0.2 mL/mouse) intraperitoneally once a week for 3 weeks on days 0, 7, and 14. Mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.2 mL of 25 mg/kg human IgG for pertuzumab. The tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab2/2, where a and b represent tumor length and width, respectively. One week after the final injection, tumor growth inhibition (TGI) was calculated using the formula (1-T/C) x 100 (%), where T (treated group) and C (control group) represent the mean tumor volume change. ED50 values were calculated from values of tumor growth inhibition on day 21 using the XL fit program. The maximum tolerated dose (MTD) was defined as the dose with neither lethality nor more than 20% body weight loss.
Statistical analysis
Packaged software (SAS preclinical package version 5.0, SAS Institute Japan, Ltd.) was used for the statistical analysis. To evaluate the potency of the drugs, values of individual tumor volumes with treatments were compared to those of the controls using the Tukey test. ED50S were calculated from values of tumor growth inhibition percentage using XL fit program #509 (MS Excel Add-In: XLfit3 Version 3.0.5 Build 12, ID Business Solutions Ltd.)
Example 3:
N-ri-({l-sec-Butyl-4-[2-f2-{[2-('3-hγdroxy-phenyl')-ethvn-methyl-carbamoyl}-l- methγlsulfanyl-propyl)-pyrroUdin-l-yl1-2-inethoxy-4-oxo-butyl}-rnethγl-carbamoyl)-2- methyl-propyll-2-dimethylamino-3-methγl-butyramide ("compound of formula (1-A)) in combination with capecitabine (Xeloda™)
Human tumor cell line
The MAXF-401 human breast carcinoma cell line was obtained from Prof. Fiebig (University of Freiburg, Germany). This cell line was maintained in mice; the tumor fragments were implanted subcutaneously and serially passaged in mice prior to use.
Animals
One hundred (100) 5-week-old female athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). The mice were housed in an air-conditioned room (temperature 22 ± 2 0C, relative humidity 55 + 10%) under 12 h light/dark cycles. The mice were given sterilized CE-2 and tap water with 25μg/mL gentamycin ad libitum. After 5 days quarantine in the animal facility of Nippon Roche Research Center (presently Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd.), mice were subjected to the experiment.
Drugs
The title compound and capecitabine were synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. The title compound was dissolved in dimethyl sulfoxide (DMSO, Wako Pure Chemical Industries Ltd.) and diluted into 2.5% DMSO with saline (for injection, Ohtsuka Pharmaceutical Co., Ltd.) before administration. Capecitabine was dissolved and diluted with 5% Gum Arabic - 40 mM Citrate buffer (pH 6) before administration.
Determination of antitumor activity
Ten to twenty mg (ca. 2x2x2 mm3) of tumor fragments of MAXF401 were implanted subcutaneously in the right flank of each mouse. Mice bearing tumors of between 245 and 588 mm3 were selected on day 35 after tumor implantation and were randomly divided into 11 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 401 mm3 and 22 g respectively. Drug administration was initiated after grouping the mice. The title compound (3 mg/kg, 4 mg/kg, or 6 mg/kg) or its vehicle was administered (0.2 mL/mouse) intravenously once a week for 3 consecutive weeks. Capecitabine or its vehicle was administered (0.5 mL/mouse) orally once a day for 14 consecutive days. Mice in the control group were given the same volume of vehicle as those in the treatment groups: 0.2 mL of 2.5% DMSO in saline for the title compound and 0.5 mL of 5% Gum Arabic - 40 mM Citrate buffer (pH 6) for capecitabine. The tumor volume and body weight of each mouse were measured twice a week. Tumor volume was estimated by using the equation ab2l2, where a and b represent tumor length and width, respectively. One week after the final injection, tumor growth inhibition (TGI) was calculated using the formula (1-T/C) x 100 (%), where T (treated group) and C (control group) represent the mean tumor volume change. ED50 values were calculated from values of tumor growth inhibition on day 21 using the XL fit program. MTD was defined as the dose with neither lethality nor more than 20% body weight loss.
Statistical analysis
Packaged software (SAS preclinical package version 5.0,SAS Institute Japan, Ltd.) was used for the statistical analysis. To evaluate the potency of the drugs, values of individual tumor volumes with treatments were compared to those of the controls using the Tukey test. ED50S were calculated from values of tumor growth inhibition percentage using XL fit program #509 (MS Excel Add Version 3.0.5 Build -In: XLfit3 12, ID Business Solutions Ltd.)
Example 4:
N-[l-f{l-sec-Butyl-4-r2-(2-{r2-r3-hydroxy-phenyl)-ethyll-methyl-carbanioyl}-l- nie&ylsulfanyl-propyl)-pyrroUdin-l-γll-2-methoxy-4-oxo-butyl}-methyl-carbamoyl)-2- methyl-propyll-2-dimethylamino-3-methyl-butyramide (compound of formula (1-A)) in combination with, cisplatin (Randa™, CDDP)
Human tumor cell line
The NCI-H460 human large cell lung carcinoma cell line was purchased from American Type Culture Collection (Rockville, MD, U.S.A.). The cells were cultured in RPMI- 1640 medium (cat. # R8758, lot 12K2310, exp 01/03; lot 82K2497, exp 08/03; Sigma- Aldrich corporation, MO, U.S.A.) supplemented with 10% (v/v) fetal bovine serum (cat. # 047115, lot # 31300124; Roche Diagnostics KK, Tokyo, Japan).
Animals
Sixty of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 8 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
Drugs
The title compound was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. CDDP (Randa™, lot # 924150) was obtained from Nippon
Kayaku Co., Ltd. (Tokyo, Japan). The title compound was dissolved in dimethyl sulfoxide (DMSO) and diluted into 2.5 % DMSO solution with saline (for Injection; Ohtsuka Pharmaceutical Industry Co., Ltd., Tokushima, Japan) on the date of administration. CDDP was diluted with saline at the date of administration.
Determination of antitumor activity
A single cell suspension of NCI-H460 (5.6 x 106 cells per mouse) was inoculated subcutaneously into the right flank of each mouse. The tumor volume was estimated by using the equation ab2/2, where a and b represent tumor length and width, respectively. Mice bearing tumors of between 150 and 245 mm3 were selected on day 7 after tumor inoculation and were randomly divided into 8 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 187 mm3 and 24.3 g, respectively. Drug administration was initiated after mice were divided into groups. The title compound (2 mg/kg or 3 mg/kg), CDDP (5 mg/kg or 6.7 mg/kg) and each vehicle were administered intravenously. Tumor volume and body weight of each mouse were measured twice a week. That dose was defined as toxic when at least one dead mouse was observed during the administration period or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period. Tumor growth inhibition was calculated using the equation (1- ΔT/ΔC) x 100, where ΔT represents the difference in tumor volume from the initial treatment date of the treated group and ΔC represents the difference in tumor volume from the initial treatment date of the vehicle group.
Example 5:
N-[l-("{l-sec-Butyl-4-r2-(2-{r2-('3-hvdroxy-phenyl)-ethyn-methyl-carbamoyl}-l- rnethylsuh%nyl-propyl)-pyrrolidin-l-yll-2-methoxy-4-oxo-butyll-methyl-carbamoyl)-2- rnethyl-propyl1-2-diπieτhylarnino-3-rnethyl-butyramide (compound of formula (1-A)) in combination with irinotecan (Topotecin™)
Human tumor cell line
The CaIu- 6 human lung carcinoma cell line was purchased from American Type Culture Collection (MD, U.S.A.). The cells were cultured in Eagle's MEM ( Sigma- Aldrich corporation, MO, U.S.A.) supplemented with 0.1 milli mol/L Non-essential amino acids (Invitrogen corporation, CA, U.S.A.), 1 milli mol/L sodium pyruvate (Invitrogen corporation, CA, U.S.A.), and 10% (v/v) fetal bovine serum (Roche Diagnostics KK, Tokyo, Japan).
Animals
60 of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 8 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
Drugs
The title compound was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Irinotecan (Topotecin™, lot. No. EQAZF13) was obtained from Daiichi Pharmaceutical Co., Ltd. (Tokyo, Japan). The title compound was dissolved in dimethyl sulfoxide (DMSO) and diluted into 2.5 % DMSO solution with saline (For Injection; Ohtsuka Pharmaceutical Industry Co., Ltd., Tokushima, Japan) at the date of administration. Irinotecan was diluted with saline at the date of administration.
Determination of antitumor activity
A single cell suspension of Calu-6 (5.7 x 106 cells per mouse) was inoculated subcutaneously into the right flank of each mouse. The tumor volume was estimated by using the equation ah212, where a and b represent tumor length and width, respectively. Mice bearing tumors of between 181 and 326 mm3 were selected on day 11 after tumor inoculation and were randomly divided into 10 groups, consisting of 5 mice each. Mean tumor volume and body weight of the selected mice were 251 mm3 and 24.7 g respectively. Drug administration was initiated after mice were randomized and divided into groups. The title compound (0.375 mg/kg; 0.75 mg/kg; 1.5 mg/kg or 3 mg/kg), irinotecan (80 mg/kg) and each vehicles were administered intravenously. Tumor volume and body weight of each mouse were measured twice a week. That dose was defined as toxic when at least one dead mouse was observed during the administration period or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period. Tumor growth inhibition was calculated by using the equation (1-ΔT/ΔC) x 100, where ΔT represents the tumor volume difference from the initial treatment date of treated group and ΔC represents the tumor volume difference from the initial treatment date of vehicle group.
Example 6:
N- r 1- (\ 1 -sec-Butyl-4- [2- (2-1 [2- (3-hvdroxy-phenyl)-ethyll -methyl-carbamoyll- 1- methylsulfanyl-propyl)-pyrrolidin-l-yll-2-methoxy-4-oxo-butyl}-methyl-carbarnoyl)-2- niethyl-propyl1-2-ά^methylarnino-3-methyl-butyrarnide (compound of formula (1-A)) in combination with capecitabine (Xeloda™)
Human tumor cell line
The HT-29 human colorectal adenocarcinoma cell line was purchased from American Type Culture Collection (MD, U.S.A.). The cells were cultured in McCoy's 5a medium (Sigma- Aldrich Corporation, MO, U.S.A.) supplemented with 10% (v/v) fetal bovine serum (Roche Diagnostics KK, Tokyo, Japan). Animals
Sixty of 5-week-old male athymic nude mice (BALB/c nu/nu) were purchased from Charles River Japan, Inc. (Yokohama, Japan). All mice were kept for 9 days in the animal facility of Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. before subjection to the experiment.
Drugs
The title compound (compound of formula I-A) was synthesized at Kamakura Research Laboratories of Chugai Pharmaceutical Co., Ltd. Capecitabine (Lot. No. 26954-190A- MIL) was purchased from F. Hoffmann-La Roche Ltd (Basel, Switzerland). The compound of formula I-A was dissolved in dimethyl sulfoxide (DMSO; Wako Pure
Chemical Industries, Ltd., Osaka, Japan) and diluted into 2.5% DMSO solution with saline (for Injection; Ohtsuka Pharmaceutical Industry Co., Ltd., Tokushima, Japan) at the date of administration. Capecitabine was dissolved by the 40 milli mol/L citrate buffered- 5% Gum arabic solution (pH 6.0; Chugai Pharmaceutical Co., Ltd.).
Determination of antitumor activity
A single cell suspension of HT-29 (1.1 x 107 cells per mouse) was inoculated subcutaneously into the right flank of each mouse. The tumor volume was estimated by using the equation ab2/2, where a and b represent tumor length and width, respectively. Mice bearing tumors of between 172 and 282 mm3 were selected on day 10 after tumor inoculation and were randomly divided into 7 groups, consisting of 6 mice each. Mean tumor volume and body weight of the selected mice were 217 mm3 and 25.0 g respectively. Drug administration was initiated after mice were divided into groups. The compound of formula I-A (3 mg/kg or 4 mg/kg) and its vehicle were administered intravenously. Capecitabine (359 mg/kg or 539 mg/kg) and its vehicle were administered orally. Tumor volume and body weight of each mouse were measured twice a week. The dose was defined as toxic when at least one dead mouse was observed during the administration period, or when half or more mice showed more than 20% continuous body weight loss compared to the initial treatment day during the administration period. Tumor growth inhibition was calculated using the equation (1-ΔT/ΔC) x 100, where ΔT represents the difference in tumor volume from the initial treatment date of the treated group and ΔC represents the difference in tumor volume from the initial treatment date of the vehicle group.

Claims

Claims
1. A pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000029_0001
in combination with capecitabine; trastuzumab; pertuzumab; irinotecan or a pharmaceutically acceptable salt thereof; or cisplatin for simultaneous, sequential or separate administration in the treatment of cancer;
wherein
R1 and R2 are methyl; ethyl; propyl; isopropyl or butyl;
R3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having (C1-C4)- alkylene and wherein the phenyl group optionally maybe substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; alkoxy; phenyl; phenoxy; trifluoromethyl; trifiuoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyi; amino and benzyl.
2. A pharmaceutical composition, comprising at least one compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000029_0002
in combination with, capecitabine; trastuzumab or pertuzumab for simultaneous, sequential or separate administration in the treatment of cancer;
wherein
R1 and R2 are methyl; ethyl; propyl; isopropyl or butyl;
R3 is phenylalkyl-, or phenyldialkylamino or phenylalkyloxy, having ( Q- C4)- alkylene and wherein the phenyl group optionally maybe substituted with one, two or three substituents selected from the group consisting of halogen; alkoxycarbonyl; sulfamoyl; alkylcarbonyloxy; carbamoyloxy; cyano; mono- or di-alkylamino; alkyl; aJkoxy; phenyl; phenoxy; trifluoromethyl; trifluoromethoxy; alkylthio; hydroxy; alkylcarbonylamino; 1,3-dioxolyl; 1,4-dioxolyl; amino and benzyl.
3. The pharmaceutical composition according to claim 1, wherein
R1 and R2 are methyl; and R3 is as defined above.
4. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is the compound of formula (I-A)
Figure imgf000030_0001
5. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is administered in combination with trastuzumab.
6. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is administered in combination with pertuzumab.
7. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is administered in combination with capecitabine.
8. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is administered in combination with cisplatin.
9. The pharmaceutical composition according to claim 1, wherein said compound of formula (I) is administered in combination with irinotecan or a pharmaceutically acceptable salt thereof.
10. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
11. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered simultaneously with capecitabine, trastuzumab or pertuzumab.
12 The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered simultaneously with cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
13. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
14. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered sequentially with capecitabine, trastuzumab or pertuzumab.
15. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab, pertuzumab, cisplatin or irinotecan or a pharmaceutically acceptable salt thereof.
16. The pharmaceutical composition according to claim 1, whereby said compound of formula (I) is administered separately from capecitabine, trastuzumab or pertuzumab.
17. The use of a pharmaceutical composition according to claim 1 for the treatment of cancer.
18. The use of a pharmaceutical composition according to claim 1 for the treatment of solid tumors.
19. The use of a pharmaceutical composition according to claim 1 for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer.
20. The use of a pharmaceutical composition according to claim 1 for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
21. The use of a pharmaceutical composition according to claim 1 for the treatment of non-small cell lung cancer.
22. The use of a pharmaceutical composition according to claim 1 for the production of a medicament for the treatment of cancer.
23. The use of a pharmaceutical composition according to claim 1 for the production of a medicament for the treatment of solid tumors.
24. The use of a pharmaceutical composition according to claim 1 for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer, breast cancer or lung cancer.
25. The use of a pharmaceutical composition according to claim 1 for the production of a medicament for the treatment of colorectal cancer, prostate cancer, pancreatic cancer or breast cancer.
26. The use of a pharmaceutical composition according to claim 1 for the production of a medicament for the treatment of non-small cell lung cancer.
27. The invention as described hereinbefore.
X-X-X-
PCT/EP2005/012992 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative WO2006063707A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2007544793A JP2008523002A (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative
EP05813987A EP1827603A2 (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative
AU2005315912A AU2005315912A1 (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one Dolastatin 10 derivative
BRPI0519023-1A BRPI0519023A2 (en) 2004-12-13 2005-12-05 pharmaceutical composition containing at least one dolastatin derivative 10
MX2007006430A MX2007006430A (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative.
CA002590431A CA2590431A1 (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP04106522 2004-12-13
EP04106522.8 2004-12-13
EP05100866.2 2005-02-08
EP05100866 2005-02-08

Publications (2)

Publication Number Publication Date
WO2006063707A2 true WO2006063707A2 (en) 2006-06-22
WO2006063707A3 WO2006063707A3 (en) 2006-10-26

Family

ID=36113901

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/012992 WO2006063707A2 (en) 2004-12-13 2005-12-05 Novel pharmaceutical composition containing at least one dolastatin 10 derivative

Country Status (15)

Country Link
US (1) US20060292158A1 (en)
EP (1) EP1827603A2 (en)
JP (1) JP2008523002A (en)
KR (1) KR20070086123A (en)
AR (1) AR052046A1 (en)
AU (1) AU2005315912A1 (en)
BR (1) BRPI0519023A2 (en)
CA (1) CA2590431A1 (en)
GT (1) GT200500364A (en)
MX (1) MX2007006430A (en)
PA (1) PA8655401A1 (en)
PE (1) PE20060747A1 (en)
RU (1) RU2007126358A (en)
TW (1) TW200635609A (en)
WO (1) WO2006063707A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012123957A1 (en) * 2011-03-16 2012-09-20 Council Of Scientific & Industrial Research Oligopeptides and process for preparation thereof
WO2015095953A1 (en) 2013-12-27 2015-07-02 The Centre For Drug Research And Development Sulfonamide-containing linkage systems for drug conjugates
WO2023033129A1 (en) 2021-09-03 2023-03-09 東レ株式会社 Pharmaceutical composition for treating and/or preventing cancer

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ712012A (en) 2013-04-16 2020-04-24 Genentech Inc Pertuzumab variants and evaluation thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003008378A1 (en) * 2001-07-19 2003-01-30 F.Hoffmann-La Roche Ag Dolastatin 10 derivatives

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4287523B2 (en) * 1998-12-28 2009-07-01 あすか製薬株式会社 Antitumor agent
EP2266607A3 (en) * 1999-10-01 2011-04-20 Immunogen, Inc. Immunoconjugates for treating cancer
US7968569B2 (en) * 2002-05-17 2011-06-28 Celgene Corporation Methods for treatment of multiple myeloma using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003008378A1 (en) * 2001-07-19 2003-01-30 F.Hoffmann-La Roche Ag Dolastatin 10 derivatives

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HOSHI A; LEESON P; CASTANER J: "TZT-1027. Antineoplastic" DRUGS OF THE FUTURE, vol. 24, no. 4, 1999, pages 404-409, XP008066836 ISSN: 0377-8282 cited in the application *
PONCET J: "THE DOLASTATINS, A FAMILY OF PROMISING ANTINEOPLASTIC AGENTS" CURRENT PHARMACEUTICAL DESIGN, BENTHAM SCIENCE PUBLISHERS, SCHIPHOL, NL, vol. 5, no. 3, 1999, pages 139-162, XP000917665 ISSN: 1381-6128 cited in the application *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012123957A1 (en) * 2011-03-16 2012-09-20 Council Of Scientific & Industrial Research Oligopeptides and process for preparation thereof
US9200034B2 (en) 2011-03-16 2015-12-01 Council Of Scientific & Industrial Research Oligopeptides and process for preparation thereof
WO2015095953A1 (en) 2013-12-27 2015-07-02 The Centre For Drug Research And Development Sulfonamide-containing linkage systems for drug conjugates
WO2023033129A1 (en) 2021-09-03 2023-03-09 東レ株式会社 Pharmaceutical composition for treating and/or preventing cancer

Also Published As

Publication number Publication date
MX2007006430A (en) 2007-07-19
BRPI0519023A2 (en) 2008-12-23
EP1827603A2 (en) 2007-09-05
PA8655401A1 (en) 2006-08-03
GT200500364A (en) 2006-08-02
PE20060747A1 (en) 2006-09-01
KR20070086123A (en) 2007-08-27
AU2005315912A1 (en) 2006-06-22
JP2008523002A (en) 2008-07-03
TW200635609A (en) 2006-10-16
US20060292158A1 (en) 2006-12-28
WO2006063707A3 (en) 2006-10-26
CA2590431A1 (en) 2006-06-22
RU2007126358A (en) 2009-01-20
AR052046A1 (en) 2007-02-28

Similar Documents

Publication Publication Date Title
CN105214086B (en) Anti-VEGF antibodies combine the application for treating breast cancer with chemotherapy
EP2882454B1 (en) Combination therapy for the treatment of glioblastoma
TW201922793A (en) Uses of PD-1 antibody combined with VEGFR inhibitor for treating small cell lung cancer
TW200803892A (en) Antineoplastic combinations with MTOR inhibitor, herceptin, and/or HKI-272
AU2023237217A1 (en) Adjuvant treatment of HER2-positive breast cancer
AU2004251168A1 (en) Treatment with anti-VEGF antibodies
JP2008501652A (en) Treatment with gemcitabine and EGFR inhibitors
JP2008516983A (en) Combination therapy with bortezomib and epidermal growth factor receptor kinase inhibitor
JP7473474B2 (en) Treatment of metastatic brain tumors by administration of antibody-drug conjugates
EP2825558B1 (en) Combination therapy for the treatment of ovarian cancer
CN111132696B (en) Use of PD-1 antibodies in combination with epigenetic modulators for the preparation of a medicament for the treatment of tumors
TW201929901A (en) Methods for treating colorectal and metastatic colorectal cancers
JP2008501651A (en) Treatment with irinotecan (CPT-11) and EGFR inhibitor
AU2021201516A1 (en) Treatment of advanced HER2 expressing cancer
US20210093719A1 (en) Co-Therapy Comprising A Small Molecule CSF-1R Inhibitor And An Agonistic Antibody That Specifically Binds CD40 For The Treatment Of Cancer
JP6425653B2 (en) Antitumor agent and antitumor effect enhancer
CN112043702A (en) Quinolines for the combined treatment of colorectal cancer
WO2020249018A1 (en) Combined pharmaceutical composition for treating driver-gene-positive lung cancer
JP2023080357A (en) Methods for treating lymphoid malignancies
WO2020233602A1 (en) Quinoline derivative used for combination treatment of small cell lung cancer
CN112043831A (en) Quinolines for use in the combined treatment of breast cancer
EP1827603A2 (en) Novel pharmaceutical composition containing at least one dolastatin 10 derivative
JP2022027658A (en) Methods and compositions for inhibition of egf/egfr pathway in combination with tyrosine kinase inhibitors
US20200237779A1 (en) Combination therapy with a bet inhibitor and a bcl-2 inhibitor
CA3167799C (en) Combined use of pertuzumab, trastuzumab, and anthracycline-based chemotherapy for neoadjuvant therapy of early-stage her2-positive breast cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2005813987

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/006430

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2590431

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005315912

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007544793

Country of ref document: JP

Ref document number: 200580042538.8

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2540/CHENP/2007

Country of ref document: IN

Ref document number: 1020077013301

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2005315912

Country of ref document: AU

Date of ref document: 20051205

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005315912

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007126358

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2005813987

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0519023

Country of ref document: BR