WO2014066202A1 - Combination - Google Patents

Combination Download PDF

Info

Publication number
WO2014066202A1
WO2014066202A1 PCT/US2013/065827 US2013065827W WO2014066202A1 WO 2014066202 A1 WO2014066202 A1 WO 2014066202A1 US 2013065827 W US2013065827 W US 2013065827W WO 2014066202 A1 WO2014066202 A1 WO 2014066202A1
Authority
WO
WIPO (PCT)
Prior art keywords
chloro
methyl
amino
ethyl
pharmaceutically acceptable
Prior art date
Application number
PCT/US2013/065827
Other languages
French (fr)
Inventor
Tona M. Gilmer
Rakesh Kumar
Original Assignee
Glaxosmithkline Llc
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 Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Priority to KR1020157013079A priority Critical patent/KR20150074097A/en
Priority to EP13848997.6A priority patent/EP2908815A4/en
Priority to JP2015538098A priority patent/JP2015534986A/en
Priority to CA2889051A priority patent/CA2889051A1/en
Priority to MX2015005113A priority patent/MX2015005113A/en
Priority to RU2015119245A priority patent/RU2015119245A/en
Priority to CN201380067522.7A priority patent/CN104902899A/en
Priority to BR112015008924A priority patent/BR112015008924A2/en
Priority to AU2013334943A priority patent/AU2013334943A1/en
Priority to IN3909DEN2015 priority patent/IN2015DN03909A/en
Priority to US14/437,297 priority patent/US20150272952A1/en
Publication of WO2014066202A1 publication Critical patent/WO2014066202A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • 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

  • the present invention relates to a method of treating cancer in a mammal and to combinations useful in such treatment.
  • the method relates to a novel combination comprising the dual EGF-R/erbB-2 inhibitor: N- ⁇ 3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and the Akt inhibitor: / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1- methyl-1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, pharmaceutical compositions comprising the same, and methods of using such combinations in the treatment of cancer.
  • cancer results from the deregulation of the normal processes that control cell division, differentiation and apoptotic cell death.
  • Apoptosis (programmed cell death) plays essential roles in embryonic development and pathogenesis of various diseases, such as degenerative neuronal diseases, cardiovascular diseases and cancer.
  • One of the most commonly studied pathways, which involves kinase regulation of apoptosis, is cellular signaling from growth factor receptors at the cell surface to the nucleus (Crews and Erikson, Cell, 74:215-17, 1993).
  • PTKs Protein tyrosine kinases
  • PTK protein tyrosine kinase
  • the ErbB family of PTKs which includes ErbB-2, EGFR, ErbB-3 and ErbB-4, is one group of PTKs that has attracted interest as a therapeutic target.
  • ErbB family PTKs is one group of PTKs that has attracted interest as a therapeutic target.
  • Elevated EGFR activity has, for example, been implicated in non-small cell lung, bladder, and head and neck cancers.
  • ErbB-2 activity has been implicated in breast, ovarian, gastric and pancreatic cancers.
  • Overexpression of HRG and/or HER3 has been reported in numerous cancers, including gastric, ovarian, prostate, bladder, and breast tumors and is associated with poor prognosis (B.Tanner,J Clin Oncol. 2006, 24(26):4317-23; M. Hayashi, Clin. Cancer Res. 2008.14(23):7843-9.; H. Kaya, Eur J Gynaecol Oncol. 2008;29(4):350-6;). Consequently, inhibition of ErbB family PTKs should provide a treatment for disorders characterized by aberrant ErbB family PTK activity.
  • Apoptosis plays essential roles in embryonic development and pathogenesis of various diseases, such as degenerative neuronal diseases, cardiovascular diseases and cancer. Recent work has led to the identification of various pro- and anti-apoptotic gene products that are involved in the regulation or execution of programmed cell death. Expression of anti-apoptotic genes, such as Bcl2 or BCI-XL, inhibits apoptotic cell death induced by various stimuli. On the other hand, expression of pro-apoptotic genes, such as Bax or Bad, leads to programmed cell death (Adams et al. Science, 281 :1322-1326 (1998)). The execution of programmed cell death is mediated by caspase -1 related proteinases, including caspase-3, caspase- 7, caspase-8 and caspase-9 etc (Thornberry et al. Science, 281 :1312-1316 (1998)).
  • PI3K phosphatidylinositol 3'-OH kinase
  • Akt/PKB pathway appears important for regulating cell survival/cell death (Kulik et al. Mol.Cell.Biol. 17:1595-1606 (1997); Franke et al, Cell, 88:435-437 (1997); Kauffmann-Zeh et al. Nature 385:544-548 (1997) Hemmings Science, 275:628-630 (1997); Dudek et al., Science, 275:661 -665 (1997)).
  • PI3K phosphatidylinositol 3'-OH kinase
  • PDGF platelet derived growth factor
  • NEF nerve growth factor
  • IGF-I insulin-like growth factor-1
  • Activated PI3K leads to the production of phosphatidylinositol (3,4,5)-triphosphate (Ptdlns (3,4,5)- P3), which in turn binds to, and promotes the activation of, the serine/threonine kinase Akt, which contains a pleckstrin homology (PH)-domain (Franke et al Cell, 81 :727-736 (1995); Hemmings Science, 277:534 (1997); Downward, Curr. Opin. Cell Biol. 10:262-267 (1998), Alessi et al., EMBO J. 15: 6541-6551 (1996)).
  • PH pleckstrin homology
  • PI3K or dominant negative Akt/PKB mutants abolish survival-promoting activities of these growth factors or cytokines. It has been previously disclosed that inhibitors of PI3K (LY294002 or wortmannin) blocked the activation of Akt/PKB by upstream kinases. In addition, introduction of constitutively active PI3K or Akt/PKB mutants promotes cell survival under conditions in which cells normally undergo apoptotic cell death (Kulik et al. 1997, Dudek et al. 1997).
  • Akt2 is overexpressed in a significant number of ovarian (J. Q. Cheung et al. Proc. Natl. Acad. Sci. U.S.A. 89:9267- 9271 (1992)) and pancreatic cancers (J. Q. Cheung et al. Proc. Natl. Acad. Sci. U.S.A. 93:3636-3641 (1996)).
  • Akt3 was found to be overexpressed in breast and prostate cancer cell lines (Nakatani et al. J. Biol.Chem. 274:21528-21532 (1999).
  • Akt-2 was over-expressed in 12% of ovarian carcinomas and that amplification of Akt was especially frequent in 50% of undifferentiated tumors, suggestion that Akt may also be associated with tumor aggressiveness (Bellacosa, et al., Int. J. Cancer, 64, pp. 280-285, 1995). Increased Akt1 kinase activity has been reported in breast, ovarian and prostate cancers (Sun et al. Am. J. Pathol. 159: 431 -7 (2001 )).
  • the tumor suppressor PTEN a protein and lipid phosphatase that specifically removes the 3' phosphate of Ptdlns(3,4,5)-P3, is a negative regulator of the PI3K/Akt pathway (Li et al. Science 275:1943-1947 (1997), Stambolic et al. Cell 95:29-39 (1998), Sun et al. Proc. Nati. Acad. Sci. U.S.A. 96:6199-6204 (1999)).
  • Germline mutations of PTEN are responsible for human cancer syndromes such as Cowden disease (Liaw et al. Nature Genetics 16:64-67 (1997)).
  • PTEN is deleted in a large percentage of human tumors and tumor cell lines without functional PTEN show elevated levels of activated Akt (Li et al. supra, Guldberg et al. Cancer Research 57:3660-3663 (1997), Risinger et al. Cancer Research 57:4736-4738 (1997)).
  • One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2- (methanesulphonyl)ethyl]amino ⁇ methyl)-2-fui l]-4-quinazolinami or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl- 1 /-/-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human.
  • One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2- (methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl- 1 /-/-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
  • the combination is administered within a specified period, and wherein the combination is administered for a duration of time.
  • One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2- (methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
  • Figure 1 depicts representative dose response curves of cell growth inhibition by Compound A, Compound B or a combination of Compound A and Compound B on the growth of ten HER2+ breast tumor lines, UACC893, KPL-4, MDA- MB-361 , HCC202, HCC1419, BT474, SK-BR-3, BT474-J4, SK-BR-3-W13 and JIMT-1.
  • the present invention relates to combinations that exhibit antiproliferative activity.
  • the method relates to methods of treating cancer by the co-administration of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-
  • Compound A is disclosed and claimed, along with pharmaceutically acceptable solvates and salts thereof, as being useful as an inhibitor of EGF-R/erbB-2 activity, particularly in treatment of cancer, in International Application No. PCT/EP99/00048, having an International filing date of January 8, 1999, International Publication Number WO 99/35146 and an International Publication date of July 15, 1999, the entire disclosure of which is hereby incorporated by reference, Compound A is the compound of Example 29. Compound A can be prepared as described in International Application No. PCT/EP99/00048.
  • Compound A is in the form of a ditosylate monohydrate salt.
  • This salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US01/20706, having an International filing date of June 28, 2001 , International Publication Number WO 02/02552 and an International Publication date of January 10, 2002, the entire disclosure of which is hereby incorporated by reference, see particularly Example 10.
  • Suitable pharmaceutical compositions containing Compound A as a single active ingredient are prepared as described in International Application No. PCT/US2006/014447, having an International filing date of April 18, 2006, International Publication Number WO 06/1 13649 and an International Publication date of October 26, 2006, the entire disclosure of which is hereby incorporated by reference, see particularly the formulation in Table 3.
  • Compound A is sold commercially as the ditosylate monohydrate salt and is known by the generic name lapatinib and trade names Tykerb® and Tyverb®.
  • Compound B is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of AKT activity, particularly in treatment of cancer, in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008, the entire disclosure of which is hereby incorporated by reference, Compound B is the compound of example 224. Compound B can be prepared as described in International Application No. PCT/US2008/053269.
  • the administration of a therapeutically effective amount of the combinations of the invention are advantageous over the individual component compounds in that the combinations will provide one or more of the following improved properties when compared to the individual administration of a therapeutically effective amount of a component compound: i) a greater anticancer effect than the most active single agent, ii) synergistic or highly synergistic anticancer activity, iii) a dosing protocol that provides enhanced anticancer activity with reduced side effect profile, iv) a reduction in the toxic effect profile, v) an increase in the therapeutic window, or vi) an increase in the bioavailability of one or both of the component compounds.
  • the compounds of the invention may contain one or more chiral atoms, or may otherwise be capable of existing as two enantiomers.
  • the compounds of this invention include mixtures of enantiomers as well as purified enantiomers or enantiomerically enriched mixtures. Also, it is understood that all tautomers and mixtures of tautomers are included within the scope of Compound A, and pharmaceutically acceptable hydrates and/or salts thereof, and Compound B, and pharmaceutically acceptable salts thereof.
  • the compounds of the invention may form a solvate which is understood to be a complex of variable stoichiometry formed by a solute (in this invention, Compound A or a salt thereof and/or Compound B or a salt thereof) and a solvent.
  • solvents for the purpose of the invention may not interfere with the biological activity of the solute.
  • suitable solvents include, but are not limited to, water, methanol, dimethyl sulfoxide, ethanol and acetic acid.
  • the solvent used is a pharmaceutically acceptable solvent.
  • the solvent used is water.
  • contemplated herein is a method of treating cancer using a combination of the invention where Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and/or Compound B or a pharmaceutically acceptable salt thereof are administered as pro-drugs.
  • Pharmaceutically acceptable pro-drugs of the compounds of the invention are readily prepared by those of skill in the art. When referring to a dosing protocol, the term “day”, “per day” and the like, refer to a time within one calendar day which begins at midnight and ends at the following midnight.
  • treating means: (1 ) to ameliorate or prevent the condition of one or more of the biological manifestations of the condition, (2) to interfere with (a) one or more points in the biological cascade that leads to or is responsible for the condition or (b) one or more of the biological manifestations of the condition, (3) to alleviate one or more of the symptoms, effects or side effects associated with the condition or treatment thereof, or (4) to slow the progression of the condition or one or more of the biological manifestations of the condition.
  • Prophylactic therapy is also contemplated thereby.
  • prevention is not an absolute term.
  • prevention is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof.
  • Prophylactic therapy is appropriate, for example, when a subject is considered at high risk for developing cancer, such as when a subject has a strong family history of cancer or when a subject has been exposed to a carcinogen.
  • the term "effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • terapéuticaally effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • ком ⁇ онент and derivatives thereof, as used herein is meant either, simultaneous administration or any manner of separate sequential administration of a therapeutically effective amount of Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B or a pharmaceutically acceptable salt thereof.
  • the compounds are administered in a close time proximity to each other.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally.
  • both compounds are administered orally.
  • the combination kit as used herein is meant the pharmaceutical position or compositions that are used to administer Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, according to the invention.
  • the combination kit can contain Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions.
  • the combination kit will contain Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in separate pharmaceutical compositions.
  • the combination kit can comprise Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in separate pharmaceutical compositions in a single package or in separate pharmaceutical compositions in separate packages.
  • a combination kit comprising the components: Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, in association with a pharmaceutically acceptable carrier; and
  • Compound B or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier.
  • first container comprising Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, in association with a pharmaceutically acceptable carrier
  • second container comprising Compound B, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier, and a container means for containing said first and second containers.
  • the "combination kit” can also be provided by instruction, such as dosage and administration instructions.
  • dosage and administration instructions can be of the kind that is provided to a doctor, for example by a drug product label, or they can be of the kind that is provided by a doctor, such as instructions to a patient.
  • Compound A means — Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof— .
  • Compound B means — Compound B, or a pharmaceutically acceptable salt thereof— .
  • Compound B is replaced by:
  • the compound 8-[4-(1 -aminocyclobutyl)phenyl]-9-phenyl[1 ,2,4]triazolo[3,4-f]-1 ,6- naphthyridin-3(2H)-one is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of AKT activity, particularly in treatment of cancer, in United States Patent 7,576,209 which issued on August 18, 2009.
  • the combinations of this invention are administered within a "specified period”.
  • the specified period can include simultaneous administration. When both compounds of the invention are administered once a day the specified period refers to timing of the
  • the specified period will be about 24 hours; suitably they will both be administered within about 12 hours of each other - in this case, the specified period will be about 12 hours; suitably they will both be administered within about 1 1 hours of each other - in this case, the specified period will be about 1 1 hours; suitably they will both be administered within about 10 hours of each other - in this case, the specified period will be about 10 hours; suitably they will both be administered within about 9 hours of each other - in this case, the specified period will be about 9 hours; suitably they will both be administered within about 8 hours of each other - in this case, the specified period will be about 8 hours; suitably they will both be administered within about 7 hours of each other - in this case, the specified period will be about 7 hours; suitably they will both be administered within about 6 hours of each other - in this case, the specified period will be about 6 hours; suit
  • Compound A and Compound B in less than about 45 minutes apart is considered simultaneous administration.
  • the compounds when the combination of the invention is administered for a “specified period”, the compounds will be co-administered for a “duration of time".
  • duration of time and derivatives thereof, as used herein is meant that both compounds of the invention are administered within a “specified period” for an indicated number of consecutive days, optionally followed by a number of consecutive days where only one of the component compounds is administered.
  • the "duration of time" and in all dosing protocols described herein do not have to commence with the start of treatment and terminate with the end of treatment, it is only required that the number of consecutive days in which both compounds are administered and the optional number of consecutive days in which only one of the component compounds is administered, or the indicated dosing protocol, occur at some point during the course of treatment.
  • both compounds will be administered within a specified period for at least 1 day - in this case, the duration of time will be at least 1 day; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days - in this case, the duration of time will be at least 2 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days - in this case, the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days - in this case, the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 7 consecutive days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 14 consecutive days - in this case, the duration of time will be at least 14 days; suitably,
  • both compounds are administered within a specified period for over 30 days, the treatment is considered chronic treatment and will continue until an altering event, such as a reassessment in cancer status or a change in the condition of the patient, warrants a modification to the protocol.
  • both compounds will be administered within a specified period for at least 1 day, followed by the administration of Compound
  • the duration of time will be at least 2 days; suitably, during the course of treatment, both compounds will be administered within a
  • the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed by administration of Compound A alone for at least 2 days - in this case, the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at
  • the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed
  • the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed by administration of
  • the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a
  • the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed by administration of Compound A alone for at least 7 days - in this case, the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at
  • the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2
  • the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified
  • the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days, followed by administration of Compound
  • the duration of time will be at least 6 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days, followed by administration of
  • the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days,
  • the duration of time will be at least 9 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive
  • the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days,
  • the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive
  • the duration of time will be at least 6 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of Compound A alone for at least 4 consecutive days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a specified
  • the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of Compound
  • the duration of time will be at least 9 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of
  • the duration of time will be at least 10 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by
  • the duration of time will be at least 5 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days,
  • the duration of time will be at least 6 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4
  • the duration of time will be at least 7 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by administration of Compound
  • the duration of time will be at least 8 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by
  • the duration of time will be at least 1 1 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5
  • the duration of time will be at least 6 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days, followed by administration of Compound A alone for at least 2 consecutive days - in this case, the duration of time will be at least 7 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days, followed by administration of Compound
  • the duration of time will be at least
  • both compounds will be administered within a specified period for at least 5 consecutive days, followed by
  • the duration of time will be at least 9 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5
  • the duration of time will be at least 10 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 7 consecutive days, followed by administration of Compound
  • the duration of time will be at least
  • both compounds will be administered within a specified period for at least 14 consecutive days, followed by
  • the duration of time will be at least 21 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 30
  • both compounds will be administered within a specified period for from 1 to 3 consecutive days, followed by administration of
  • both compounds will be administered within a specified period for 5 consecutive days, followed by administration
  • both compounds will be administered within a specified period for 2 consecutive days,
  • both compounds will be administered within a specified period for from 1 to 3 days over a 7 day period, and during the other days of the
  • Compound A will be administered alone.
  • both compounds will be administered within a specified period for 2 days over
  • Compound B is subsequently administered for 1 or more consecutive days. Unless otherwise defined, the "sequential administration" and in all dosing protocols described herein, do not have to commence with the start of treatment and terminate with the end of
  • Compound B or the indicated dosing protocol, occur at some point during the course of treatment. Also, contemplated herein is a drug holiday utilized between the sequential
  • a drug holiday is a period of days after the
  • the drug holiday will be a period of days selected from: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days and 14 days.
  • one of Compound A and Compound B is administered for from 1 to 30 consecutive days, followed by an optional drug holiday, followed by administration of the
  • one of Compound A and Compound B is administered for from 1 to 21 consecutive days, followed by an optional drug holiday, followed by administration of the other of
  • Compound A and Compound B for from 1 to 21 consecutive days.
  • Compound A and Compound B is administered for from 1 to 14 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of the other of
  • Compound A and Compound B for from 1 to 14 consecutive days.
  • Compound A and Compound B is administered for from 2 to 7 consecutive days, followed by a drug holiday of from 2 to 10 days, followed by administration of the other of
  • Compound B will be administered first in the sequence, followed by an
  • B is administered for from 1 to 21 consecutive days, followed by an optional drug
  • Compound B is administered for from 3 to 21 consecutive days, followed by a
  • Compound B is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by
  • Compound A for from 3 to 21 consecutive days.
  • Compound A for from 3 to 21 consecutive days.
  • B is administered for 21 consecutive days, followed by an optional drug holiday, followed by
  • Compound A for 14 consecutive days.
  • Compound B is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days,
  • Compound B is administered for 7 consecutive days, followed by a drug holiday of from
  • Compound B is administered for 3 consecutive days, followed by a drug
  • Compound B is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound
  • Compound A for 3 consecutive days.
  • Compound A will be administered first in the sequence, followed by an
  • A is administered for from 1 to 21 consecutive days, followed by an optional drug
  • Compound A is administered for from 3 to 21 consecutive days, followed by a
  • Compound A is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by
  • A is administered for 21 consecutive days, followed by an optional drug holiday, followed by
  • Compound B is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days,
  • Compound A is administered for 7 consecutive days, followed by a drug holiday of from
  • Compound A is administered for 3 consecutive days, followed by a drug
  • Compound A is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound
  • Compound A is administered for 7 consecutive days.
  • Compound A is N-(2-aminoethyl)-2 2 days, followed by administration of Compound B for 1 day.
  • Compound A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2 2 2 days, followed by administration of Compound B for 1 day.
  • Compound A is N-(2-aminoethyl)-2 2 days, followed by administration of Compound B for 1 day.
  • Compound B is administered for 1 day, followed by administration of
  • Compound A for 7 consecutive days.
  • Compound B is administered for 1 day,
  • a "specified period” administration and a “sequential” administration can be followed by one or more cycles of repeat dosing or can be followed by an alternate dosing protocol, and a drug holiday may precede the repeat dosing or alternate dosing protocol.
  • the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from about 250mg to about 1 ,500mg; suitably, the amount will be selected from about 500mg to about 1 ,250mg; suitably, the amount will be selected from about 750mg to about 1 ,250mg; suitably, the amount will be selected from about 1 ,000mg to about 1 ,250mg; suitably, the amount will be 250mg, suitably, the amount will be 500mg, suitably, the amount will be 750mg, suitably, the amount will be 1 ,000mg, suitably, the amount will be 1 ,250mg; suitably, the
  • the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from
  • the amount of Compound A administered as part of the combination according to the present invention is suitably selected from 250mg, 500mg, 750mg, 1 ,000mg, 1 ,250mg and 1 ,500mg.
  • the amount of Compound A administered as part of the combination according to the present invention is suitably selected from 250mg, 500mg, 750mg, 1 ,000mg, 1 ,250mg and 1 ,500mg.
  • the amount of Compound A administered as part of the combination according to the present invention is suitably selected from 250mg, 500mg, 750mg, 1 ,000mg, 1 ,250mg and 1 ,500mg.
  • the amount of Compound A administered as part of the combination according to the present invention is suitably selected from 250mg, 500mg, 750mg, 1 ,000mg, 1 ,250mg and 1 ,500mg.
  • the amount of Compound A administered as part of the combination according to the present invention is suitably selected from
  • Compound A is administered from 1 to 4 times a day, in one or more
  • the selected amount of Compound A is administered twice a day, in
  • the selected amount of Compound A is administered once a day, in one or more tablets.
  • the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from about 5mg to about 500mg; suitably, the amount will be selected from about 25mg to about 400mg; suitably, the amount will be selected from about 30mg to about 375mg; suitably, the amount will be selected from about 35mg to about 350mg; suitably, the amount will be selected from about 40mg to about 300mg; suitably, the amount will be selected from about 45mg to about 275mg; suitably, the amount will be selected from about 50mg to about 250mg; suitably, the amount will be selected from about 55mg to about 225mg; suitably, the amount will be selected from about 60mg to about 200mg; suitably, the amount will be selected from about 65mg to about 175mg; suitably, the amount will be selected from about 70mg to about 150mg; suitably, the amount will be selected from about 50mg to about 300mg; suitably, the amount of the amount
  • the amount will be about 100mg. Accordingly, the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from about 5mg to about 500mg. For example, the amount of
  • Compound B administered as part of the combination according to the present invention can be 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 55mg, 60mg, 65mg, 70mg, 75mg, 80mg, 85mg, 90mg, 95mg, 100mg, 105mg, 1 10mg, 1 15mg, 120mg, 125mg, 130mg, 135mg, 140mg, 145mg, 150mg, 175mg, 200mg, 225mg, 250mg, 275mg, 300mg, 325mg, 350mg, 375mg, 400mg, 425mg, 450mg, 475mg or 500mg.
  • the selected amount of Compound B is administered twice a day.
  • the selected amount of Compound B is administered twice a day.
  • compositions While it is possible that, for use in therapy, therapeutically effective amounts of the combinations of the present invention may be administered as the raw chemical, it is preferable to present the combinations as a pharmaceutical composition or compositions.
  • compositions which include
  • Compound A and/or Compound B and one or more pharmaceutically acceptable carriers.
  • the combinations of the present invention are as described above.
  • the carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation, capable of pharmaceutical formulation, and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical formulation including admixing
  • Compound A and/or Compound B with one or more pharmaceutically acceptable carriers may be presented in separate pharmaceutical compositions or formulated together in one pharmaceutical formulation.
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. As is known to those skilled in the art, the amount of active ingredient per dose will depend on the condition being treated, the route of administration and the age, weight and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Furthermore, such pharmaceutical formulations may be prepared by any of the methods well known in the pharmacy art.
  • Compound A and Compound B may be administered by any appropriate route. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal, and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal, and epidural). It will be appreciated that the preferred route may vary with, for example, the condition of the recipient of the combination and the cancer to be treated. It will also be appreciated that each of the agents administered may be administered by the
  • Compound B are administered in separate pharmaceutical compositions.
  • Solid or liquid pharmaceutical carriers are employed.
  • Solid carriers include, starch, lactose, calcium sulfate dihydrate, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid.
  • Liquid carriers include syrup, peanut oil, olive oil, saline, and water.
  • the carrier may include a prolonged release material, such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the amount of solid carrier varies widely but, suitably, may be from about 25 mg to about 1 g per dosage unit.
  • the preparation will suitably be in the form of a syrup, elixir, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampoule, or an aqueous or nonaqueous liquid suspension.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • the formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • Compound A in combination with Compound B are administered to a human.
  • the therapeutically effective amount of the administered agents of the present invention will depend upon a number of factors including, for example, the age and weight of the subject, the precise condition requiring treatment, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the therapeutically effective amount will be at the discretion of the attending physician.
  • the combinations of the invention are tested for efficacy, advantageous and synergistic properties generally according to known procedures.
  • JIMT-1 Human tumor cell lines from breast, BT474, BT474-J4, JIMT-1 , MDA-MB-361 , SK- BR-3, HCC1419, UACC893 and HCC202 in RPMI 1640 containing 10 % FBS media; SK- BR-3-W13 and BT474-J4 in RPMI 1640 containing 10% FBS and 1 ⁇ Compound A (in this assay, Compound A, was used in the form of the ditosylate monohydrate salt) media were kept in a humidified incubator at 37°C in 95% air and 5% C0 2.
  • JIMT-1 is a line derived from a patient clinically resistant to trastuzumab (Herceptin®).
  • SK-BR-3-W13 is a single cell clone isolated by a cloning cylinder after a single treatment of SK-BR-3 cells with 0.5 ⁇ Compound A.
  • BT474-J4 is a single cell clone derived from BT474 cells that were selected to grow in Compound A to a concentration of 3 ⁇ .
  • ATP levels were determined by adding Cell Titer Glo® (Promega) according to the manufacturer's protocol. Briefly, Cell Titer Glo® was added each plate, incubated for 20 minutes then luminescent signal was read on the SpectraMax L plate reader with a 0.5 sec integration time.
  • IC 5 o(a) is the IC 5 o of Compound A
  • IC 5 o(b) is the IC 5 o for Compound B
  • Da is the concentration of Compound A in combination with Compound B that inhibited 50 % of cell growth
  • Db is the concentration of Compound B in combination with Compound A that inhibited 50% of cell growth.
  • a CI value ⁇ 0.9, between 0.9 and 1.1 , or >1 .1 indicates synergy, additivity and antagonism, respectively.
  • the smaller the CI number the greater is the strength of synergy.
  • EOHSA values are defined as increases in improvement
  • a drug combination (at total dose d1 +d2) is said to have a statistically significant EOHSA if the mean response at the combination is significantly better than the mean responses for either drug 1 (alone) at dose d1 or drug 2 (alone) at dose d2.
  • EOHSA is a common approach for evaluating drug combinations, and is an FDA criterion (21 CRF 300.50) for combination drug approval. See Borisy et al. (Borisy AA, et al. Proc Natl Acad Sci;100(13):7977-82, 2003) or Hung et al. (Hung HM, Chi GY, Lipicky RJ.
  • IC 50 for the combination drug corresponds to a total dose of d1 +d2.
  • IC 50 for the combination drug corresponds to a total dose of d1 +d2.
  • d1 +d2 the mean response for the combination (d1 +d2) to drug 1 at d1 and drug 2 at d2 using the respective fitted dose response curves corresponding to the fixed -dose-ratio combination curve and the dose response curves for drugs 1 and 2 alone.
  • UACC893 and KPL-4 HER2+ lines with an H1047R PIK3CA mutation are sensitive to both Compound A and Compound B single agents.
  • the combination of Compound A and Compound B showed synergistic effects as demonstrated by the combination index values (CI, 0.38 and 0.73 respectively) and greater than the most active single agent by EOHSA analysis (29 and 24 ppt respectively).
  • MDA-MB-361 and HCC202 HER2+ lines with an E545K PIK3CA mutation are less sensitive to Compound A or Compound B as single agents.
  • the combination of Compound A and Compound B is beneficial as indicated by the CI of 0.72 in HCC202 and EOHSA values of >12 ppt in both MDA-MB-361 and HCC202 cell lines.
  • Both BT474-J4 and SK-BR-3-W13 lines are HER2+, Compound A acquired resistant clones developed from BT474 and Sk-Br-3 cells respectively.
  • JIMT-1 is a line derived from a patient who was resistant to trastuzumab therapy (Tanner et al, Mol Cancer Ther 2004;3:1585-92).
  • BT474-J4 line is sensitive to cell growth inhibition by Compound B.
  • the combination of Compound A and Compound B is synergistic in BT474-J4 cells.
  • Both SK-BR-3-W13 and JIMT1 are not sensitive to Compound A (IC 50 >5 ⁇ ) or Compound B (IC 50 >1 uM).
  • the combination of Compound A and Compound B showed a greater effect than the most active single agent (EOHSA >10 ppt).
  • the present invention relates to a method for treating or lessening the severity of a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone, thyroid,
  • a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma,
  • Lymphoblastic T cell leukemia Chronic myelogenous leukemia, Chronic lymphocytic leukemia, Hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, Chronic neutrophilic leukemia, Acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma Megakaryoblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia,
  • lymphoma malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma,
  • neuroblastoma bladder cancer, urothelial cancer, lung cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor) and testicular cancer.
  • the present invention relates to a method for treating or lessening the severity of a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma and thyroid.
  • a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma and thyroid.
  • the present invention relates to a method for treating or lessening the severity of a cancer selected from ovarian, breast, pancreatic and prostate.
  • the present invention relates to a method of treating or lessening the severity of a cancer that is either wild type or mutant for Ras/Raf and either wild type or mutant for PIK3CA/PTEN. This includes patients who are wild type for both Ras/Raf and PIK3CA/PTEN, mutant for both Ras/Raf and PIK3CA/PTEN, mutant for Ras/Raf and wild type for PIK3CA/PTEN and wild type for Ras/Raf and mutant for PIK3CA/PTEN.
  • the present invention also relates to a method of treating or lessening the severity of a cancer that has activated AKT, e.g., by mutation or amplification of AKT1 , AKT2 or AKT3 genes.
  • the present invention also relates to a method of treating or lessening the severity of a cancer that has activated EGFR or ErbB-2, e.g., by mutation, amplification of the gene or overexpression of the protein.
  • wild type refers to a polypeptide or polynucleotide sequence that occurs in a native population without genetic modification.
  • a “mutant” includes a polypeptide or polynucleotide sequence having at least one modification to an amino acid or nucleic acid compared to the corresponding amino acid or nucleic acid found in a wild type polypeptide or polynucleotide, respectively. Included in the term mutant is Single Nucleotide Polymorphism (SNP) where a single base pair distinction exists in the sequence of a nucleic acid strand compared to the most prevalently found (wild type) nucleic acid strand.
  • SNP Single Nucleotide Polymorphism
  • Cancers that are either wild type or mutant for Ras/Raf, PIK3CA/PTEN, AKT, EGFR or ErbB-2 or have amplification of PIK3CA, AKT, EGFR or ErbB-2 genes or have overexpression of EGFR or ErbB2 protein are identified by known methods.
  • wild type or mutant Ras/Raf, PIK3CA/PTEN, AKT EGFR or ErbB-2 tumor cells can be identified by DNA amplification and sequencing techniques, DNA and RNA detection techniques, including, but not limited to Northern and Southern blot, respectively, and/or various biochip and array technologies or in-situ hybridization. Wild type and mutant polypeptides can be detected by a variety of techniques including, but not limited to immunodiagnostic techniques such as ELISA, Western blot or immunocytochemistry.
  • This invention provides a combination comprising N- ⁇ 3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof.
  • This invention also provides for a combination comprising N- ⁇ 3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, for use in therapy.
  • This invention also provides for a combination comprising N- ⁇ 3-Chloro-4- [(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and A/- ⁇ (1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, for use in treating cancer.
  • This invention also provides a pharmaceutical composition comprising a combination of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-
  • This invention also provides a combination kit comprising N- ⁇ 3-Chloro-4- [(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof.
  • This invention also provides for the use of a combination comprising N- ⁇ 3- Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2- furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament.
  • This invention also provides for the use of a combination comprising N- ⁇ 3- Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl)ethyl]amino ⁇ methyl)-2- furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament to treat cancer.
  • This invention also provides a method of treating cancer which comprises administering a combination of N- ⁇ 3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl ⁇ -6-[5-( ⁇ [2- (methanesulphonyl)ethyl]amino ⁇ methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and / ⁇ /- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • An oral dosage form for administering a combination of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table I, below.
  • An oral dosage form for administering one of the compounds of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table II, below.
  • An oral dosage form for administering one of the compounds of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table III, below.
  • sucrose, microcrystalline cellulose and the compounds of the invented combination are mixed and granulated in the proportions shown with a 10% gelatin solution.
  • the wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet.
  • sucrose, microcrystalline cellulose and one of the compounds of the invented combination are mixed and granulated in the proportions shown with a 10% gelatin solution.
  • the wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet.
  • sucrose, microcrystalline cellulose and one of the compounds of the invented combination are mixed and granulated in the proportions shown with a 10% gelatin solution.
  • the wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Otolaryngology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)

Abstract

The present invention relates to a method of treating cancer in a human and to pharmaceutical combinations useful in such treatment. In particular, the method relates to a cancer treatment method that includes administering N-{3-Chloro-4-[(3fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and N{(1S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to a human in need thereof.

Description

COMBINATION
FIELD OF THE INVENTION
The present invention relates to a method of treating cancer in a mammal and to combinations useful in such treatment. In particular, the method relates to a novel combination comprising the dual EGF-R/erbB-2 inhibitor: N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and the Akt inhibitor: /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1- methyl-1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, pharmaceutical compositions comprising the same, and methods of using such combinations in the treatment of cancer.
BACKGROUND OF THE INVENTION
Generally, cancer results from the deregulation of the normal processes that control cell division, differentiation and apoptotic cell death. Apoptosis (programmed cell death) plays essential roles in embryonic development and pathogenesis of various diseases, such as degenerative neuronal diseases, cardiovascular diseases and cancer. One of the most commonly studied pathways, which involves kinase regulation of apoptosis, is cellular signaling from growth factor receptors at the cell surface to the nucleus (Crews and Erikson, Cell, 74:215-17, 1993).
Protein tyrosine kinases (PTKs) catalyze the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth and differentiation. (A.F. Wilks, Progress in Growth Factor Research, 1990, 2, 97-1 1 1 ; S.A. Courtneidge, Dev. Supp.l, 1993, 57-64; J.A. Cooper, Semin. Cell Biol., 1994, 5(6), 377-387; R.F. Paulson, Semin. Immunol., 1995, 7(4), 267-277; A.C. Chan, Curr. Opin. Immunol., 1996, 8(3), 394-401 ). Inappropriate or uncontrolled activation of many PTKs, i.e. aberrant PTK activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growth.
Aberrant protein tyrosine kinase (PTK) activity has been implicated in a variety of disorders including psoriasis, rheumatoid arthritis, bronchitis, as well as cancer. Development of effective treatments for such disorders is a constant and ongoing enterprise in the medical field. The ErbB family of PTKs, which includes ErbB-2, EGFR, ErbB-3 and ErbB-4, is one group of PTKs that has attracted interest as a therapeutic target. Currently, of special interest, is the role of ErbB family PTKs in hyperproliferative disorders, particularly human malignancies. Elevated EGFR activity has, for example, been implicated in non-small cell lung, bladder, and head and neck cancers. Furthermore, increased ErbB-2 activity has been implicated in breast, ovarian, gastric and pancreatic cancers. Overexpression of HRG and/or HER3 has been reported in numerous cancers, including gastric, ovarian, prostate, bladder, and breast tumors and is associated with poor prognosis (B.Tanner,J Clin Oncol. 2006, 24(26):4317-23; M. Hayashi, Clin. Cancer Res. 2008.14(23):7843-9.; H. Kaya, Eur J Gynaecol Oncol. 2008;29(4):350-6;). Consequently, inhibition of ErbB family PTKs should provide a treatment for disorders characterized by aberrant ErbB family PTK activity. The biological role of ErbB family PTKs and their implication in various disease states is discussed, for instance in U.S. patent 5,773,476; International Patent Application WO 99/35146; M.C. Hung et al, Seminars in Oncology, 26: 4, Suppl. 12 (August) 1999, 51 -59; Ullrich et al, Cell, 61 : 203-212, April 20, 1990; Modjtahedi et al, Int'l. J. of Oncology, 13: 335-342, 1998; and J.R. Woodburn, Pharmacol. Ther., 82: 2-3, 241 -250, 1999, it is generally accepted that inhibitors of ErbB family kinases will be useful for the treatment of such cancers or other condition associated with ErbB family kinases.
Apoptosis (programmed cell death) plays essential roles in embryonic development and pathogenesis of various diseases, such as degenerative neuronal diseases, cardiovascular diseases and cancer. Recent work has led to the identification of various pro- and anti-apoptotic gene products that are involved in the regulation or execution of programmed cell death. Expression of anti-apoptotic genes, such as Bcl2 or BCI-XL, inhibits apoptotic cell death induced by various stimuli. On the other hand, expression of pro-apoptotic genes, such as Bax or Bad, leads to programmed cell death (Adams et al. Science, 281 :1322-1326 (1998)). The execution of programmed cell death is mediated by caspase -1 related proteinases, including caspase-3, caspase- 7, caspase-8 and caspase-9 etc (Thornberry et al. Science, 281 :1312-1316 (1998)).
The phosphatidylinositol 3'-OH kinase (PI3K)/Akt/PKB pathway appears important for regulating cell survival/cell death (Kulik et al. Mol.Cell.Biol. 17:1595-1606 (1997); Franke et al, Cell, 88:435-437 (1997); Kauffmann-Zeh et al. Nature 385:544-548 (1997) Hemmings Science, 275:628-630 (1997); Dudek et al., Science, 275:661 -665 (1997)). Survival factors, such as platelet derived growth factor (PDGF), nerve growth factor (NGF) and insulin-like growth factor-1 (IGF-I), promote cell survival under various conditions by inducing the activity of PI3K (Kulik et al. 1997, Hemmings 1997). Activated PI3K leads to the production of phosphatidylinositol (3,4,5)-triphosphate (Ptdlns (3,4,5)- P3), which in turn binds to, and promotes the activation of, the serine/threonine kinase Akt, which contains a pleckstrin homology (PH)-domain (Franke et al Cell, 81 :727-736 (1995); Hemmings Science, 277:534 (1997); Downward, Curr. Opin. Cell Biol. 10:262-267 (1998), Alessi et al., EMBO J. 15: 6541-6551 (1996)). Specific inhibitors of PI3K or dominant negative Akt/PKB mutants abolish survival-promoting activities of these growth factors or cytokines. It has been previously disclosed that inhibitors of PI3K (LY294002 or wortmannin) blocked the activation of Akt/PKB by upstream kinases. In addition, introduction of constitutively active PI3K or Akt/PKB mutants promotes cell survival under conditions in which cells normally undergo apoptotic cell death (Kulik et al. 1997, Dudek et al. 1997).
Analysis of Akt levels in human tumors showed that Akt2 is overexpressed in a significant number of ovarian (J. Q. Cheung et al. Proc. Natl. Acad. Sci. U.S.A. 89:9267- 9271 (1992)) and pancreatic cancers (J. Q. Cheung et al. Proc. Natl. Acad. Sci. U.S.A. 93:3636-3641 (1996)). Similarly, Akt3 was found to be overexpressed in breast and prostate cancer cell lines (Nakatani et al. J. Biol.Chem. 274:21528-21532 (1999). It was demonstrated that Akt-2 was over-expressed in 12% of ovarian carcinomas and that amplification of Akt was especially frequent in 50% of undifferentiated tumors, suggestion that Akt may also be associated with tumor aggressiveness (Bellacosa, et al., Int. J. Cancer, 64, pp. 280-285, 1995). Increased Akt1 kinase activity has been reported in breast, ovarian and prostate cancers (Sun et al. Am. J. Pathol. 159: 431 -7 (2001 )).
The tumor suppressor PTEN, a protein and lipid phosphatase that specifically removes the 3' phosphate of Ptdlns(3,4,5)-P3, is a negative regulator of the PI3K/Akt pathway (Li et al. Science 275:1943-1947 (1997), Stambolic et al. Cell 95:29-39 (1998), Sun et al. Proc. Nati. Acad. Sci. U.S.A. 96:6199-6204 (1999)). Germline mutations of PTEN are responsible for human cancer syndromes such as Cowden disease (Liaw et al. Nature Genetics 16:64-67 (1997)). PTEN is deleted in a large percentage of human tumors and tumor cell lines without functional PTEN show elevated levels of activated Akt (Li et al. supra, Guldberg et al. Cancer Research 57:3660-3663 (1997), Risinger et al. Cancer Research 57:4736-4738 (1997)).
These observations demonstrate that the PI3K/Akt pathway plays important roles for regulating cell survival or apoptosis in tumorigenesis and/or cancer.
It would be useful to provide a novel therapy which provides more effective and/or enhanced treatment of an individual suffering the effects of cancer.
SUMMARY OF THE INVENTION
One embodiment of this invention provides a combination comprising:
(i) a compound of Structure (I):
Figure imgf000006_0001
or a pharmaceutically acceptable hydrate and/or salt thereof; and (ii) a compound of Structure (II):
Figure imgf000006_0002
or a pharmaceutically acceptable salt thereof.
One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-fui l]-4-quinazolinami or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 /-/-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human.
One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 /-/-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the combination is administered within a specified period, and wherein the combination is administered for a duration of time.
One embodiment of this invention provides a method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the compounds of the combination are administered sequentially.
BRIEF DESCRIPTION OF THE DRAWINGS Figure - 1 Figure 1 depicts representative dose response curves of cell growth inhibition by Compound A, Compound B or a combination of Compound A and Compound B on the growth of ten HER2+ breast tumor lines, UACC893, KPL-4, MDA- MB-361 , HCC202, HCC1419, BT474, SK-BR-3, BT474-J4, SK-BR-3-W13 and JIMT-1. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to combinations that exhibit antiproliferative activity. Suitably, the method relates to methods of treating cancer by the co-administration of N- {3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, (hereinafter Compound A, or a pharmaceutically acceptable hydrate and/or salt, suitably the ditosylate monohydrate salt, thereof,
which compound is represented by Structure I:
Figure imgf000008_0001
and N-{(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1- methyl-1 H-pyrazol-5-yl)-2-furancarboxamide or a pharmaceutically acceptable salt thereof, (hereinafter Compound B or a pharmaceutically acceptable salt thereof,
which compound is represented by Structure II:
Figure imgf000008_0002
Compound A is disclosed and claimed, along with pharmaceutically acceptable solvates and salts thereof, as being useful as an inhibitor of EGF-R/erbB-2 activity, particularly in treatment of cancer, in International Application No. PCT/EP99/00048, having an International filing date of January 8, 1999, International Publication Number WO 99/35146 and an International Publication date of July 15, 1999, the entire disclosure of which is hereby incorporated by reference, Compound A is the compound of Example 29. Compound A can be prepared as described in International Application No. PCT/EP99/00048.
Suitably, Compound A is in the form of a ditosylate monohydrate salt. This salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US01/20706, having an International filing date of June 28, 2001 , International Publication Number WO 02/02552 and an International Publication date of January 10, 2002, the entire disclosure of which is hereby incorporated by reference, see particularly Example 10.
Suitable pharmaceutical compositions containing Compound A as a single active ingredient are prepared as described in International Application No. PCT/US2006/014447, having an International filing date of April 18, 2006, International Publication Number WO 06/1 13649 and an International Publication date of October 26, 2006, the entire disclosure of which is hereby incorporated by reference, see particularly the formulation in Table 3.
Compound A is sold commercially as the ditosylate monohydrate salt and is known by the generic name lapatinib and trade names Tykerb® and Tyverb®.
Compound B is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of AKT activity, particularly in treatment of cancer, in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008, the entire disclosure of which is hereby incorporated by reference, Compound B is the compound of example 224. Compound B can be prepared as described in International Application No. PCT/US2008/053269. The administration of a therapeutically effective amount of the combinations of the invention are advantageous over the individual component compounds in that the combinations will provide one or more of the following improved properties when compared to the individual administration of a therapeutically effective amount of a component compound: i) a greater anticancer effect than the most active single agent, ii) synergistic or highly synergistic anticancer activity, iii) a dosing protocol that provides enhanced anticancer activity with reduced side effect profile, iv) a reduction in the toxic effect profile, v) an increase in the therapeutic window, or vi) an increase in the bioavailability of one or both of the component compounds. The compounds of the invention may contain one or more chiral atoms, or may otherwise be capable of existing as two enantiomers. Accordingly, the compounds of this invention include mixtures of enantiomers as well as purified enantiomers or enantiomerically enriched mixtures. Also, it is understood that all tautomers and mixtures of tautomers are included within the scope of Compound A, and pharmaceutically acceptable hydrates and/or salts thereof, and Compound B, and pharmaceutically acceptable salts thereof.
The compounds of the invention may form a solvate which is understood to be a complex of variable stoichiometry formed by a solute (in this invention, Compound A or a salt thereof and/or Compound B or a salt thereof) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. Examples of suitable solvents include, but are not limited to, water, methanol, dimethyl sulfoxide, ethanol and acetic acid. Suitably the solvent used is a pharmaceutically acceptable solvent. Suitably the solvent used is water.
The pharmaceutically acceptable salts of the compounds of the invention are readily prepared by those of skill in the art.
Also, contemplated herein is a method of treating cancer using a combination of the invention where Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and/or Compound B or a pharmaceutically acceptable salt thereof are administered as pro-drugs. Pharmaceutically acceptable pro-drugs of the compounds of the invention are readily prepared by those of skill in the art. When referring to a dosing protocol, the term "day", "per day" and the like, refer to a time within one calendar day which begins at midnight and ends at the following midnight.
By the term "treating" and derivatives thereof as used herein, is meant therapeutic therapy. In reference to a particular condition, treating means: (1 ) to ameliorate or prevent the condition of one or more of the biological manifestations of the condition, (2) to interfere with (a) one or more points in the biological cascade that leads to or is responsible for the condition or (b) one or more of the biological manifestations of the condition, (3) to alleviate one or more of the symptoms, effects or side effects associated with the condition or treatment thereof, or (4) to slow the progression of the condition or one or more of the biological manifestations of the condition. Prophylactic therapy is also contemplated thereby. The skilled artisan will appreciate that "prevention" is not an absolute term. In medicine, "prevention" is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof. Prophylactic therapy is appropriate, for example, when a subject is considered at high risk for developing cancer, such as when a subject has a strong family history of cancer or when a subject has been exposed to a carcinogen. As used herein, the term "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.
By the term "combination" and derivatives thereof, as used herein is meant either, simultaneous administration or any manner of separate sequential administration of a therapeutically effective amount of Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B or a pharmaceutically acceptable salt thereof. Preferably, if the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and the other compound may be administered orally. Suitably, both compounds are administered orally.
By the term "combination kit" as used herein is meant the pharmaceutical position or compositions that are used to administer Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, according to the invention. When both compounds are administered simultaneously, the combination kit can contain Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the compounds are not administered simultaneously, the combination kit will contain Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in separate pharmaceutical compositions. The combination kit can comprise Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, in separate pharmaceutical compositions in a single package or in separate pharmaceutical compositions in separate packages.
In one aspect there is provided a combination kit comprising the components: Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, in association with a pharmaceutically acceptable carrier; and
Compound B, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier.
In one embodiment of the invention the combination kit comprises the following components:
Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, in association with a pharmaceutically acceptable carrier; and
Compound B, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier,
wherein the components are provided in a form which is suitable for sequential, separate and/or simultaneous administration.
In one embodiment the combination kit comprises:
a first container comprising Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof, in association with a pharmaceutically acceptable carrier; and a second container comprising Compound B, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier, and a container means for containing said first and second containers.
The "combination kit" can also be provided by instruction, such as dosage and administration instructions. Such dosage and administration instructions can be of the kind that is provided to a doctor, for example by a drug product label, or they can be of the kind that is provided by a doctor, such as instructions to a patient.
2
As used herein the term "Compound A " means — Compound A, or a pharmaceutically acceptable hydrate and/or salt thereof— .
2
As used herein the term "Compound B " means — Compound B, or a pharmaceutically acceptable salt thereof— . In one embodiment of the present invention Compound B is replaced by:
8-[4-(1-aminocyclobutyl)phenyl]-9-phenyl[1 ,2,4]triazolo[3,4-f]-1 ,6-naphthyridin- 3(2H)-one; which has the following structure (depicted as the chloride salt):
Figure imgf000013_0001
provided that when said compound is administered on a day 1 , day 3, and day 5 of a dosing protocol the compound is not administered at a dose selected from: 30mg, 45mg or 60mg.
In one embodiment of the present invention Compound B is replaced by the compound:
8-[4-(1-aminocyclobutyl)phenyl]-9-phenyl[1 ,2,4]triazolo[3,4-f]-1 ,6-naphthyridin- 3(2H)-one or a pharmaceutically acceptable salt thereof;
provided that when said compound is administered on a day 1 , day 3, and day 5 of a dosing protocol the compound is not administered at a dose selected from: 30mg, 45mg or 60mg. The compound 8-[4-(1 -aminocyclobutyl)phenyl]-9-phenyl[1 ,2,4]triazolo[3,4-f]-1 ,6- naphthyridin-3(2H)-one is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of AKT activity, particularly in treatment of cancer, in United States Patent 7,576,209 which issued on August 18, 2009. 8-[4-(1- aminocyclobutyl)phenyl]-9-phenyl[1 ,2,4]triazolo[3,4-f]-1 ,6-naphthyridin-3(2H)-one can be prepared as described in United States Patent 7,576,209.
Suitably the combinations of this invention are administered within a "specified period".
By the term "specified period" and derivatives thereof, as used herein is meant the
2 2 interval of time between the administration of one of Compound A and Compound B
2 2
and the other of Compound A and Compound B . Unless otherwise defined, the specified period can include simultaneous administration. When both compounds of the invention are administered once a day the specified period refers to timing of the
2 2
administration of Compound A and Compound B during a single day. When one or both compounds of the invention are administered more than once a day, the specified period is calculated based on the first administration of each compound on a specific day. All administrations of a compound of the invention that are subsequent to the first during a specific day are not considered when calculating the specific period.
Suitably, if the compounds are administered within a "specified period" and not administered simultaneously, they are both administered within about 24 hours of each other - in this case, the specified period will be about 24 hours; suitably they will both be administered within about 12 hours of each other - in this case, the specified period will be about 12 hours; suitably they will both be administered within about 1 1 hours of each other - in this case, the specified period will be about 1 1 hours; suitably they will both be administered within about 10 hours of each other - in this case, the specified period will be about 10 hours; suitably they will both be administered within about 9 hours of each other - in this case, the specified period will be about 9 hours; suitably they will both be administered within about 8 hours of each other - in this case, the specified period will be about 8 hours; suitably they will both be administered within about 7 hours of each other - in this case, the specified period will be about 7 hours; suitably they will both be administered within about 6 hours of each other - in this case, the specified period will be about 6 hours; suitably they will both be administered within about 5 hours of each other - in this case, the specified period will be about 5 hours; suitably they will both be administered within about 4 hours of each other - in this case, the specified period will be about 4 hours; suitably they will both be administered within about 3 hours of each other - in this case, the specified period will be about 3 hours; suitably they will be administered within about 2 hours of each other - in this case, the specified period will be about 2 hours; suitably they will both be administered within about 1 hour of each other - in this case, the specified period will be about 1 hour. As used herein, the administration of
2 2
Compound A and Compound B in less than about 45 minutes apart is considered simultaneous administration.
Suitably, when the combination of the invention is administered for a "specified period", the compounds will be co-administered for a "duration of time". By the term "duration of time" and derivatives thereof, as used herein is meant that both compounds of the invention are administered within a "specified period" for an indicated number of consecutive days, optionally followed by a number of consecutive days where only one of the component compounds is administered. Unless otherwise defined, the "duration of time" and in all dosing protocols described herein, do not have to commence with the start of treatment and terminate with the end of treatment, it is only required that the number of consecutive days in which both compounds are administered and the optional number of consecutive days in which only one of the component compounds is administered, or the indicated dosing protocol, occur at some point during the course of treatment.
Regarding "specified period" administration:
Suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day - in this case, the duration of time will be at least 1 day; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days - in this case, the duration of time will be at least 2 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days - in this case, the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days - in this case, the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 7 consecutive days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 14 consecutive days - in this case, the duration of time will be at least 14 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 30 consecutive days - in this case, the duration of time will be at least 30 days. When, during the course of treatment, both compounds are administered within a specified period for over 30 days, the treatment is considered chronic treatment and will continue until an altering event, such as a reassessment in cancer status or a change in the condition of the patient, warrants a modification to the protocol.
Further regarding "specified period" administration:
Suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed by the administration of Compound
2
A alone for at least 1 day - in this case, the duration of time will be at least 2 days; suitably, during the course of treatment, both compounds will be administered within a
2
specified period for at least 1 day, followed by administration of Compound A alone for at least 2 days - in this case, the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at
2
least 1 day, followed by administration of Compound A alone for at least 3 days - in this case, the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed
2
by administration of Compound A alone for at least 4 days - in this case, the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 1 day, followed by administration of
2
Compound A alone for at least 5 days - in this case, the duration of time will be at least
6 days; suitably, during the course of treatment, both compounds will be administered
2 within a specified period for at least 1 day, followed by administration of Compound A alone for at least 6 days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a
2
specified period for at least 1 day, followed by administration of Compound A alone for at least 7 days - in this case, the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at
2
least 2 consecutive days, followed by administration of Compound A alone for at least 1 day - in this case, the duration of time will be at least 3 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2
2
consecutive days, followed by administration of Compound A alone for at least 2 consecutive days - in this case, the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified
2 period for at least 2 consecutive days, followed by administration of Compound A alone for at least 3 consecutive days - in this case, the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days, followed by administration of Compound
2
A alone for at least 4 consecutive days - in this case, the duration of time will be at least 6 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days, followed by administration of
2
Compound A alone for at least 5 consecutive days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days, followed by
2
administration of Compound A alone for at least 6 consecutive days - in this case, the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 2 consecutive days,
2
followed by administration of Compound A alone for at least 7 consecutive days - in this case, the duration of time will be at least 9 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive
2
days, followed by administration of Compound A alone for at least 1 day - in this case, the duration of time will be at least 4 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days,
2
followed by administration of Compound A alone for at least 2 consecutive days - in this case, the duration of time will be at least 5 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive
2
days, followed by administration of Compound A alone for at least 3 consecutive days - in this case, the duration of time will be at least 6 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of Compound A alone for at least 4 consecutive days - in this case, the duration of time will be at least 7 days; suitably, during the course of treatment, both compounds will be administered within a specified
2 period for at least 3 consecutive days, followed by administration of Compound A alone for at least 5 consecutive days - in this case, the duration of time will be at least 8 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of Compound
2
A alone for at least 6 consecutive days - in this case, the duration of time will be at least 9 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 3 consecutive days, followed by administration of
2
Compound A alone for at least 7 consecutive days - in this case, the duration of time will be at least 10 days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by
2
administration of Compound A alone for at least 1 day - in this case, the duration of time will be at least 5 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days,
2
followed by administration of Compound A alone for at least 2 consecutive days - in this case, the duration of time will be at least 6 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4
2
consecutive days, followed by administration of Compound A alone for at least 3 consecutive days - in this case, the duration of time will be at least 7 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by administration of Compound
2
A alone for at least 4 consecutive days - in this case, the duration of time will be at least 8 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 4 consecutive days, followed by
2
administration of Compound A alone for at least 7 consecutive days - in this case, the duration of time will be at least 1 1 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5
2
consecutive days, followed by administration of Compound A alone for at least 1 day - in this case, the duration of time will be at least 6 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days, followed by administration of Compound A alone for at least 2 consecutive days - in this case, the duration of time will be at least 7 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days, followed by administration of Compound
2
A alone for at least 3 consecutive days - in this case, the duration of time will be at least
8 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5 consecutive days, followed by
2
administration of Compound A alone for at least 4 consecutive days - in this case, the duration of time will be at least 9 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 5
2
consecutive days, followed by administration of Compound A alone for at least 5 consecutive days - in this case, the duration of time will be at least 10 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 7 consecutive days, followed by administration of Compound
2
A alone for at least 2 consecutive days - in this case, the duration of time will be at least
9 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 14 consecutive days, followed by
2
administration of Compound A alone for at least 7 consecutive days - in this case, the duration of time will be at least 21 consecutive days; suitably, during the course of treatment, both compounds will be administered within a specified period for at least 30
2
consecutive days, followed by administration of Compound A alone for at least 7 consecutive days - in this case, the duration of time will be at least 37 consecutive days. Suitably, during the course of treatment, both compounds will be administered within a specified period for from 1 to 3 consecutive days, followed by administration of
2
Compound A alone for from 3 to 7 consecutive days. Suitably, during the course of treatment, both compounds will be administered within a specified period for from 3 to 6
2
consecutive days, followed by administration of Compound A alone for from 1 to 4 consecutive days. Suitably, during the course of treatment, both compounds will be administered within a specified period for 5 consecutive days, followed by administration
2
of Compound A alone for 2 consecutive days. Suitably, during the course of treatment, both compounds will be administered within a specified period for 2 consecutive days,
2
followed by administration of Compound A alone for from 3 to 7 consecutive days. Suitably, during the course of treatment, both compounds will be administered within a specified period for from 1 to 3 days over a 7 day period, and during the other days of the
2
7 day period Compound A will be administered alone. Suitably, during the course of treatment, both compounds will be administered within a specified period for 2 days over
2 a 7 day period, and during the other days of the 7 day period Compound A will be administered alone.
Suitably, if the compounds are not administered during a "specified period", they are administered sequentially. By the term "sequential administration", and derivates
2 2 thereof, as used herein is meant that one of Compound A and Compound B is
2 administered for 1 or more consecutive days and the other of Compound A and
2
Compound B is subsequently administered for 1 or more consecutive days. Unless otherwise defined, the "sequential administration" and in all dosing protocols described herein, do not have to commence with the start of treatment and terminate with the end of
2 treatment, it is only required that the administration of one of Compound A and
2 2 Compound B followed by the administration of the other of Compound A and
2
Compound B , or the indicated dosing protocol, occur at some point during the course of treatment. Also, contemplated herein is a drug holiday utilized between the sequential
2 2 2 administration of one of Compound A and Compound B and the other of Compound A
2
and Compound B . As used herein, a drug holiday is a period of days after the
2 2
sequential administration of one of Compound A and Compound B and before the
2 2
administration of the other of Compound A and Compound B where neither Compound
2 2
A nor Compound B is administered. Suitably the drug holiday will be a period of days selected from: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days and 14 days.
Regarding sequential administration:
2 2
Suitably, one of Compound A and Compound B is administered for from 1 to 30 consecutive days, followed by an optional drug holiday, followed by administration of the
2 2
other of Compound A and Compound B for from 1 to 30 consecutive days. Suitably,
2 2
one of Compound A and Compound B is administered for from 1 to 21 consecutive days, followed by an optional drug holiday, followed by administration of the other of
2 2
Compound A and Compound B for from 1 to 21 consecutive days. Suitably, one of
2 2
Compound A and Compound B is administered for from 1 to 14 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of the other of
2 2
Compound A and Compound B for from 1 to 14 consecutive days. Suitably, one of
2 2
Compound A and Compound B is administered for from 2 to 7 consecutive days, followed by a drug holiday of from 2 to 10 days, followed by administration of the other of
2 2
Compound A and Compound B for from 2 to 7 consecutive days.
2
Suitably, Compound B will be administered first in the sequence, followed by an
2
optional drug holiday, followed by administration of Compound A . Suitably, Compound
2
B is administered for from 1 to 21 consecutive days, followed by an optional drug
2
holiday, followed by administration of Compound A for from 1 to 21 consecutive days.
2
Suitably, Compound B is administered for from 3 to 21 consecutive days, followed by a
2 drug holiday of from 1 to 14 days, followed by administration of Compound A for from 3
2
to 21 consecutive days. Suitably, Compound B is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by
2
administration of Compound A for from 3 to 21 consecutive days. Suitably, Compound
2
B is administered for 21 consecutive days, followed by an optional drug holiday, followed
2 2 by administration of Compound A for 14 consecutive days. Suitably, Compound B is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days,
2
followed by administration of Compound A for 14 consecutive days. Suitably,
2
Compound B is administered for 7 consecutive days, followed by a drug holiday of from
2
3 to 10 days, followed by administration of Compound A for 7 consecutive days.
2
Suitably, Compound B is administered for 3 consecutive days, followed by a drug
2 holiday of from 3 to 14 days, followed by administration of Compound A for 7
2
consecutive days. Suitably, Compound B is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound
2
A for 3 consecutive days. Suitably, Compound A will be administered first in the sequence, followed by an
2
optional drug holiday, followed by administration of Compound B . Suitably, Compound
2
A is administered for from 1 to 21 consecutive days, followed by an optional drug
2
holiday, followed by administration of Compound B for from 1 to 21 consecutive days.
2
Suitably, Compound A is administered for from 3 to 21 consecutive days, followed by a
2 drug holiday of from 1 to 14 days, followed by administration of Compound B for from 3
2
to 21 consecutive days. Suitably, Compound A is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by
2
administration of Compound B for from 3 to 21 consecutive days. Suitably, Compound
2
A is administered for 21 consecutive days, followed by an optional drug holiday, followed
2 2 by administration of Compound B for 14 consecutive days. Suitably, Compound A is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days,
2
followed by administration of Compound B for 14 consecutive days. Suitably,
2
Compound A is administered for 7 consecutive days, followed by a drug holiday of from
2
3 to 10 days, followed by administration of Compound B for 7 consecutive days.
2
Suitably, Compound A is administered for 3 consecutive days, followed by a drug
2 holiday of from 3 to 14 days, followed by administration of Compound B for 7
2
consecutive days. Suitably, Compound A is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound
2 2
B for 3 consecutive days. Suitably, Compound A is administered for 7 consecutive
2 2 days, followed by administration of Compound B for 1 day. Suitably, Compound A is
2 administered for 6 consecutive days, followed by administration of Compound B for 1
2
day. Suitably, Compound B is administered for 1 day, followed by administration of
2 2
Compound A for 7 consecutive days. Suitably, Compound B is administered for 1 day,
2
followed by administration of Compound A for 6 consecutive days.
It is understood that a "specified period" administration and a "sequential" administration can be followed by one or more cycles of repeat dosing or can be followed by an alternate dosing protocol, and a drug holiday may precede the repeat dosing or alternate dosing protocol.
2
Suitably, the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from about 250mg to about 1 ,500mg; suitably, the amount will be selected from about 500mg to about 1 ,250mg; suitably, the amount will be selected from about 750mg to about 1 ,250mg; suitably, the amount will be selected from about 1 ,000mg to about 1 ,250mg; suitably, the amount will be 250mg, suitably, the amount will be 500mg, suitably, the amount will be 750mg, suitably, the amount will be 1 ,000mg, suitably, the amount will be 1 ,250mg; suitably, the
2
amount will be 1 ,500mg. Accordingly, the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from
2 about 250mg to about 1 ,500 mg. For example, the amount of Compound A administered as part of the combination according to the present invention is suitably selected from 250mg, 500mg, 750mg, 1 ,000mg, 1 ,250mg and 1 ,500mg. Suitably, the
2
selected amount of Compound A is administered from 1 to 4 times a day, in one or more
2
tablets. Suitably, the selected amount of Compound A is administered twice a day, in
2
one or more tablets. Suitably, the selected amount of Compound A is administered once a day, in one or more tablets.
2
Suitably, the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from about 5mg to about 500mg; suitably, the amount will be selected from about 25mg to about 400mg; suitably, the amount will be selected from about 30mg to about 375mg; suitably, the amount will be selected from about 35mg to about 350mg; suitably, the amount will be selected from about 40mg to about 300mg; suitably, the amount will be selected from about 45mg to about 275mg; suitably, the amount will be selected from about 50mg to about 250mg; suitably, the amount will be selected from about 55mg to about 225mg; suitably, the amount will be selected from about 60mg to about 200mg; suitably, the amount will be selected from about 65mg to about 175mg; suitably, the amount will be selected from about 70mg to about 150mg; suitably, the amount will be selected from about 50mg to about 300mg; suitably, the amount will be selected from about 75mg to about 150mg;
2 suitably, the amount will be about 100mg. Accordingly, the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from about 5mg to about 500mg. For example, the amount of
2
Compound B administered as part of the combination according to the present invention can be 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 55mg, 60mg, 65mg, 70mg, 75mg, 80mg, 85mg, 90mg, 95mg, 100mg, 105mg, 1 10mg, 1 15mg, 120mg, 125mg, 130mg, 135mg, 140mg, 145mg, 150mg, 175mg, 200mg, 225mg, 250mg, 275mg, 300mg, 325mg, 350mg, 375mg, 400mg, 425mg, 450mg, 475mg or 500mg. Suitably, the
2
selected amount of Compound B is administered twice a day. Suitably, the selected
2
amount of Compound B is administered once a day.
2 2
As used herein, all amounts specified for Compound A and Compound B are indicated as the administered amount of free or unsalted and unsolvated compound per dose. The method of the present invention may also be employed with other therapeutic methods of cancer treatment.
While it is possible that, for use in therapy, therapeutically effective amounts of the combinations of the present invention may be administered as the raw chemical, it is preferable to present the combinations as a pharmaceutical composition or compositions.
Accordingly, the invention further provides pharmaceutical compositions, which include
2 2
Compound A and/or Compound B , and one or more pharmaceutically acceptable carriers. The combinations of the present invention are as described above. The carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation, capable of pharmaceutical formulation, and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical formulation including admixing
2 2
Compound A and/or Compound B with one or more pharmaceutically acceptable carriers. As indicated above, such elements of the pharmaceutical combination utilized may be presented in separate pharmaceutical compositions or formulated together in one pharmaceutical formulation.
Pharmaceutical formulations may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. As is known to those skilled in the art, the amount of active ingredient per dose will depend on the condition being treated, the route of administration and the age, weight and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Furthermore, such pharmaceutical formulations may be prepared by any of the methods well known in the pharmacy art.
2 2
Compound A and Compound B may be administered by any appropriate route. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal, and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal, and epidural). It will be appreciated that the preferred route may vary with, for example, the condition of the recipient of the combination and the cancer to be treated. It will also be appreciated that each of the agents administered may be administered by the
2 2
same or different routes and that Compound A and Compound B may be compounded
2 together in a pharmaceutical composition/formulation. Suitably, Compound A and
2
Compound B are administered in separate pharmaceutical compositions.
The compounds or combinations of the current invention are incorporated into convenient dosage forms such as capsules, tablets, or injectable preparations. Solid or liquid pharmaceutical carriers are employed. Solid carriers include, starch, lactose, calcium sulfate dihydrate, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Liquid carriers include syrup, peanut oil, olive oil, saline, and water. Similarly, the carrier may include a prolonged release material, such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies widely but, suitably, may be from about 25 mg to about 1 g per dosage unit. When a liquid carrier is used, the preparation will suitably be in the form of a syrup, elixir, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampoule, or an aqueous or nonaqueous liquid suspension.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present. It should be understood that in addition to the ingredients mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
As indicated, therapeutically effective amounts of the combinations of the
2 2
invention (Compound A in combination with Compound B ) are administered to a human. Typically, the therapeutically effective amount of the administered agents of the present invention will depend upon a number of factors including, for example, the age and weight of the subject, the precise condition requiring treatment, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the therapeutically effective amount will be at the discretion of the attending physician. The combinations of the invention are tested for efficacy, advantageous and synergistic properties generally according to known procedures.
Methods: Cell lines and growth conditions
Human tumor cell lines from breast, BT474, BT474-J4, JIMT-1 , MDA-MB-361 , SK- BR-3, HCC1419, UACC893 and HCC202 in RPMI 1640 containing 10 % FBS media; SK- BR-3-W13 and BT474-J4 in RPMI 1640 containing 10% FBS and 1 μΜ Compound A (in this assay, Compound A, was used in the form of the ditosylate monohydrate salt) media were kept in a humidified incubator at 37°C in 95% air and 5% C02. JIMT-1 is a line derived from a patient clinically resistant to trastuzumab (Herceptin®). SK-BR-3-W13 is a single cell clone isolated by a cloning cylinder after a single treatment of SK-BR-3 cells with 0.5 μΜ Compound A. BT474-J4 is a single cell clone derived from BT474 cells that were selected to grow in Compound A to a concentration of 3 μΜ.
Cell growth inhibition assay and combination data analysis.
All cells were cultured without Compound A for a minimum of 72 hours prior to cell plating. Cells were assayed in a 96-well tissue culture plate (NUNC 136102) of RPMI containing 10% FBS at 2,000 cells/well. Approximately 24 hours after plating, cells were exposed to ten, two-fold or three-fold serial dilutions of Compound A or the combination of the two agents at a constant molar to molar ratio of 10:1 Compound A to Compound B (in this assay, Compound B refers to /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide and was used as the free or unsalted compound) in RPMI media containing 10% FBS. Cells were incubated in the presence of compounds for 3 days. ATP levels were determined by adding Cell Titer Glo® (Promega) according to the manufacturer's protocol. Briefly, Cell Titer Glo® was added each plate, incubated for 20 minutes then luminescent signal was read on the SpectraMax L plate reader with a 0.5 sec integration time.
Inhibition of cell growth was estimated after treatment with Compound A or the combination of Compound A and Compound B for three days and comparing the signal to cells treated with vehicle (DMSO). Cell growth was calculated relative to vehicle (DMSO) treated control wells. Concentration of compound that inhibits 50% of control cell growth (IC50) was interpolated using nonlinear regression with the equation, y=(A+(B- A)/(1 +(C/x)AD)))), where A is the minimum response (ymin)> B is the maximum response (ymax), C is the inflection point of the curve (EC50) and D is the Hill coefficient.
Combination effects on potency were evaluated using Combination Index (CI) which was calculated with the back-interpolated IC50 values and the mutually nonexclusive equation derived by Chou and Talalay (Chou TC, Talalay P. Adv Enzyme Regul; 22:27-55, 1984): CI = Da/IC50(a) + Db/IC50(b) + (Da x Db)/(IC50(a) x IC50(b))
where IC5o(a) is the IC5o of Compound A; IC5o(b) is the IC5o for Compound B; Da is the concentration of Compound A in combination with Compound B that inhibited 50 % of cell growth; and Db is the concentration of Compound B in combination with Compound A that inhibited 50% of cell growth. In general, a CI value <0.9, between 0.9 and 1.1 , or >1 .1 indicates synergy, additivity and antagonism, respectively. In general, the smaller the CI number, the greater is the strength of synergy.
The combination effects on the response scale were quantified by Excess Over
Highest Single Agent (EOHSA). EOHSA values are defined as increases in improvement
(here, in 'percentage points' (ppts) difference) produced by the combination over the best single drug at its component dose level. More specifically, suppose we have a combination composed of drug 1 at dose d1 and drug 2 at dose d2. If the effect of the combination of drugs 1 and 2 at doses d1 and d2 is better than either drug 1 (alone) at dose d1 or drug 2 (alone) at dose d2, then the combination is said to have a positive EOHSA and beneficial for that combination. For a combination drug experiment (involving drug 1 at dose d1 and drug 2 at dose d2), a drug combination (at total dose d1 +d2) is said to have a statistically significant EOHSA if the mean response at the combination is significantly better than the mean responses for either drug 1 (alone) at dose d1 or drug 2 (alone) at dose d2. EOHSA is a common approach for evaluating drug combinations, and is an FDA criterion (21 CRF 300.50) for combination drug approval. See Borisy et al. (Borisy AA, et al. Proc Natl Acad Sci;100(13):7977-82, 2003) or Hung et al. (Hung HM, Chi GY, Lipicky RJ. Biometrics 49(1 ):85-94, 1993) for examples and discussion. The EOHSA analysis was conducted as follows. Since dose response curves were fit to the experimental data (for both of the single drug regimens and also for the combination drug at a fixed-dose-ratio ray), comparisons needed for making EOHSA statistical inferences could be done by interpolation using the fitted regression models. At specified total dose levels of IC5o along the dose response curve of a fixed-dose-ratio ray, the dose combination (corresponding to IC50) was determined for making EOHSA statistical inferences. Here, the mean response at a given combination, IC50 for example, was compared to the mean response at the component dose levels for drugs 1 and 2 on their dose response curves. More specifically, suppose that the IC50 for the combination drug (along the fixed-dose-ratio ray) corresponds to a total dose of d1 +d2. We then compare the mean response for the combination (d1 +d2) to drug 1 at d1 and drug 2 at d2 using the respective fitted dose response curves corresponding to the fixed -dose-ratio combination curve and the dose response curves for drugs 1 and 2 alone.
Cell growth inhibition by Compound A, Compound B and the combination of Compound A with Compound B.
The effects of cell growth inhibition by Compound A, Compound B and their combination were determined in ten HER2+ breast tumor lines, UACC893, KPL-4, MDA- MB-361 , HCC202, HCC1419, BT474, SK-BR-3, BT474-J4, SK-BR-3-W13 and JIMT-1 . The mean IC50s (at least two independent experiments) and the combination effects at IC50s are summarized in Table 1 . Representative dose response curves are provided in Figure 1. HCC1419, BT474 and SK-BR-3 HER2+ lines are highly sensitive to Compound A with IC50 values of less than 0.2 μΜ, and less sensitive to Compound B with IC50 > 0.5 μΜ. The combination of Compound A and Compound B showed additive or similar to the most active single agent effects in these cells.
UACC893 and KPL-4 HER2+ lines with an H1047R PIK3CA mutation are sensitive to both Compound A and Compound B single agents. The combination of Compound A and Compound B showed synergistic effects as demonstrated by the combination index values (CI, 0.38 and 0.73 respectively) and greater than the most active single agent by EOHSA analysis (29 and 24 ppt respectively). MDA-MB-361 and HCC202 HER2+ lines with an E545K PIK3CA mutation are less sensitive to Compound A or Compound B as single agents. The combination of Compound A and Compound B is beneficial as indicated by the CI of 0.72 in HCC202 and EOHSA values of >12 ppt in both MDA-MB-361 and HCC202 cell lines.
Both BT474-J4 and SK-BR-3-W13 lines are HER2+, Compound A acquired resistant clones developed from BT474 and Sk-Br-3 cells respectively. JIMT-1 is a line derived from a patient who was resistant to trastuzumab therapy (Tanner et al, Mol Cancer Ther 2004;3:1585-92). BT474-J4 line is sensitive to cell growth inhibition by Compound B. The combination of Compound A and Compound B is synergistic in BT474-J4 cells. Both SK-BR-3-W13 and JIMT1 are not sensitive to Compound A (IC50>5 μΜ) or Compound B (IC50>1 uM). The combination of Compound A and Compound B showed a greater effect than the most active single agent (EOHSA >10 ppt).
Table 1. Cell growth inhibition by Compound A, Compound B and their combination in tumor cell lines.
Figure imgf000030_0001
*lines resistant to Compound A and trastuzumab; ND: not determined; NA: not applicable; Combination index:CI.
Because the combinations of the present invention are active in the above assays they exhibit advantageous therapeutic utility in treating cancer.
Because the combinations of the present invention are active in the above assays they exhibit advantageous therapeutic utility in treating cancer.
Suitably, the present invention relates to a method for treating or lessening the severity of a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone, thyroid,
Lymphoblastic T cell leukemia, Chronic myelogenous leukemia, Chronic lymphocytic leukemia, Hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, Chronic neutrophilic leukemia, Acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma Megakaryoblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia,
malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma,
neuroblastoma, bladder cancer, urothelial cancer, lung cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor) and testicular cancer.
Suitably, the present invention relates to a method for treating or lessening the severity of a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma and thyroid.
Suitably, the present invention relates to a method for treating or lessening the severity of a cancer selected from ovarian, breast, pancreatic and prostate. Suitably, the present invention relates to a method of treating or lessening the severity of a cancer that is either wild type or mutant for Ras/Raf and either wild type or mutant for PIK3CA/PTEN. This includes patients who are wild type for both Ras/Raf and PIK3CA/PTEN, mutant for both Ras/Raf and PIK3CA/PTEN, mutant for Ras/Raf and wild type for PIK3CA/PTEN and wild type for Ras/Raf and mutant for PIK3CA/PTEN. The present invention also relates to a method of treating or lessening the severity of a cancer that has activated AKT, e.g., by mutation or amplification of AKT1 , AKT2 or AKT3 genes. The present invention also relates to a method of treating or lessening the severity of a cancer that has activated EGFR or ErbB-2, e.g., by mutation, amplification of the gene or overexpression of the protein.
The term "wild type" as is understood in the art refers to a polypeptide or polynucleotide sequence that occurs in a native population without genetic modification. As is also understood in the art, a "mutant" includes a polypeptide or polynucleotide sequence having at least one modification to an amino acid or nucleic acid compared to the corresponding amino acid or nucleic acid found in a wild type polypeptide or polynucleotide, respectively. Included in the term mutant is Single Nucleotide Polymorphism (SNP) where a single base pair distinction exists in the sequence of a nucleic acid strand compared to the most prevalently found (wild type) nucleic acid strand.
Cancers that are either wild type or mutant for Ras/Raf, PIK3CA/PTEN, AKT, EGFR or ErbB-2 or have amplification of PIK3CA, AKT, EGFR or ErbB-2 genes or have overexpression of EGFR or ErbB2 protein are identified by known methods. For example, wild type or mutant Ras/Raf, PIK3CA/PTEN, AKT EGFR or ErbB-2 tumor cells can be identified by DNA amplification and sequencing techniques, DNA and RNA detection techniques, including, but not limited to Northern and Southern blot, respectively, and/or various biochip and array technologies or in-situ hybridization. Wild type and mutant polypeptides can be detected by a variety of techniques including, but not limited to immunodiagnostic techniques such as ELISA, Western blot or immunocytochemistry.
This invention provides a combination comprising N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt thereof, and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof. This invention also provides for a combination comprising N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, for use in therapy.
This invention also provides for a combination comprising N-{3-Chloro-4- [(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and A/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, for use in treating cancer. This invention also provides a pharmaceutical composition comprising a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2 -furancarboxamide, or a pharmaceutically acceptable salt thereof.
This invention also provides a combination kit comprising N-{3-Chloro-4- [(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof.
This invention also provides for the use of a combination comprising N-{3- Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2- furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament.
This invention also provides for the use of a combination comprising N-{3- Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2- furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament to treat cancer. This invention also provides a method of treating cancer which comprises administering a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, suitably the ditosylate monohydrate salt, thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
The following examples are intended for illustration only and are not intended to limit the scope of the invention in any way.
Experimental Details
Example 1 - Capsule Composition
An oral dosage form for administering a combination of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table I, below.
Table I
INGREDIENTS AMOUNTS
N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- 250mg
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate (the ditosylate
monohydrate salt of Compound A)
Λ/-{(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5- 75mg
chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide (Compound B)
Mannitol 250 mg
Talc 125 mg
Magnesium Stearate 8 mg Example 2 - Capsule Composition
An oral dosage form for administering one of the compounds of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table II, below.
Table II
INGREDIENTS
N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate (the ditosylate
monohydrate salt of Compound A)
Mannitol
Talc
Magnesium Stearate
Example 3 - Capsule Composition
An oral dosage form for administering one of the compounds of the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table III, below.
Table III
INGREDIENTS AMOUNTS
Λ/-{(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5 75mg
chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide (Compound B)
Mannitol 250mg
Talc 125mg
Magnesium Stearate 8mg
Example 4 - Tablet Composition
The sucrose, microcrystalline cellulose and the compounds of the invented combination, as shown in Table IV below, are mixed and granulated in the proportions shown with a 10% gelatin solution. The wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet. Table IV
INGREDIENTS
N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate (the ditosylate
monohydrate salt of Compound A)
Λ/-{(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5- chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide (Compound B)
Microcrystalline cellulose
sucrose
starch
talc
stearic acid
Example 5 - Tablet Composition
The sucrose, microcrystalline cellulose and one of the compounds of the invented combination, as shown in Table V below, are mixed and granulated in the proportions shown with a 10% gelatin solution. The wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet.
Table V
INGREDIENTS AMOUNTS
N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- 250mg
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate (the ditosylate
monohydrate salt of Compound A)
Microcrystalline cellulose 30mg
sucrose 4mg
starch 2mg
talc 1 mg
stearic acid 0.5mg Example 6 - Tablet Composition
The sucrose, microcrystalline cellulose and one of the compounds of the invented combination, as shown in Table VI below, are mixed and granulated in the proportions shown with a 10% gelatin solution. The wet granules are screened, dried, mixed with the starch, talc and stearic acid, then screened and compressed into a tablet.
Table VI
INGREDIENTS
Λ/-{(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5- chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide (Compound B)
Microcrystalline cellulose
sucrose
starch
talc
stearic acid
While the preferred embodiments of the invention are illustrated by the above, it is to be understood that the invention is not limited to the precise instructions herein disclosed and that the right to all modifications coming within the scope of the following claims is reserved.

Claims

We claim:
1. A combination comprising: (i) a compound of Structure (I):
Figure imgf000038_0001
or a pharmaceutically acceptable hydrate and/or salt thereof; and (ii) a compound of Structure (II):
Figure imgf000038_0002
or a pharmaceutically acceptable salt thereof.
2. A combination according to claim 1 where the compound of Structure (I) in the form of a ditosylate monohydrate salt.
3. A combination kit comprising a combination according to claim 1 or claim together with a pharmaceutically acceptable carrier or carriers.
4. A combination according to any one of claims 1 to 3 where the amount of the compound of Structure (I) is an amount selected from 750mg to 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of the compound of Structure (II) is an amount selected from 50mg to 300mg, and that amount is administered once per day.
5. Use of a combination according to any of claims 1 to 4 in the manufacture of a medicament or medicaments for the treatment of cancer.
6. A method of treating cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3- Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2- furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl- 1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the combination is administered within a specified period, and wherein the combination is administered for a duration of time.
7. A method according to claim 6 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 50mg to about 300mg, and that amount is administered once per day.
8. A method according to claim 7 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 60mg to about 300mg, and that amount is administered once per day.
9. A method according to claim 8 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for from 1 to 3 consecutive days followed by administration of N- {3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate for from 3 to 7 consecutive days, optionally followed by one or more cycles of repeat dosing.
10. A method treating a cancer selected from: brain (gliomas), glioblastomas,
Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone, thyroid,
Lymphoblastic T cell leukemia, Chronic myelogenous leukemia, Chronic lymphocytic leukemia, Hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, Chronic neutrophilic leukemia, Acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma Megakaryoblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia,
malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma,
neuroblastoma, bladder cancer, urothelial cancer, lung cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor) and testicular cancer;
in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and N- {(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H- pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the combination is administered within a specified period, and wherein the combination is administered for a duration of time.
1 1 . A method according to claim 10 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 50mg to about 300mg, and that amount is administered once per day.
12. A method according to claim 1 1 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 60mg to about 300mg, and that amount is administered once per day.
13. A method according to claim 12 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for from 1 to 3 consecutive days followed by administration of N- {3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate for from 3 to 7 consecutive days, optionally followed by one or more cycles of repeat dosing.
14. A method according to claim 10 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
15. A method according to claim 1 1 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
16. A method according to claim 12 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
17. A method according to claim 13 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
18. A method of treating a cancer that is either wild type or mutant for Ras/Raf, PIK3CA/PTEN, AKT, EGFR or ErbB-2 or have amplification of PIK3CA, AKT, EGFR or ErbB-2 genes or have overexpression of EGFR or ErbB2 protein, in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human, wherein the combination is administered within a specified period, and wherein the combination is administered for a duration of time.
19. A method according to claim 18 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 50mg to about 300mg, and that amount is administered once per day.
20. A method according to claim 19 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 60mg to about 300mg, and that amount is administered once per day.
21 . A method according to claim 20 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for from 1 to 3 consecutive days followed by administration of N- {3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate for from 3 to 7 consecutive days, optionally followed by one or more cycles of repeat dosing.
22. A method according to claim 18 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
23. A method according to claim 19 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
24. A method according to claim 20 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
25. A method according to claim 21 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
26. A method treating a cancer selected from: brain (gliomas), glioblastomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon, head and neck, kidney, lung, liver, melanoma, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone, thyroid,
Lymphoblastic T cell leukemia, Chronic myelogenous leukemia, Chronic lymphocytic leukemia, Hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, Chronic neutrophilic leukemia, Acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma Megakaryoblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia,
malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma,
neuroblastoma, bladder cancer, urothelial cancer, lung cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor) and testicular cancer; in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and N- {(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H- pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the compounds of the combination are administered sequentially.
27. A method according to claim 26 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 50mg to about 300mg, and that amount is administered once per day.
28. A method according to claim 27 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 60mg to about 300mg, and that amount is administered once per day.
29. A method according to claim 28 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate is administered for from 1 to 30 consecutive days, followed by an optional drug holiday of from 1 to 14 days, followed by administration of /\/-{(1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4- chloro-1-methyl-1 H-pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, for from 1 to 30 days, optionally followed by one or more cycles of repeat dosing.
30. A method according to claim 26 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
31 . A method according to claim 27 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
31 . A method according to claim 28 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
33. A method according to claim 29 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
34. A method of treating a cancer that is either wild type or mutant for Ras/Raf, PIK3CA/PTEN, AKT, EGFR or ErbB-2 or have amplification of PIK3CA, AKT, EGFR or ErbB-2 genes or have overexpression of EGFR or ErbB2 protein, in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, to such human,
wherein the compounds of the combination are administered sequentially.
35. A method according to claim 34 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 50mg to about 300mg, and that amount is administered once per day.
36. A method according to claim 35 wherein the amount of N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine, or a pharmaceutically acceptable hydrate and/or salt thereof, is selected from about 750mg to about 1 ,250mg, and that amount is administered once per day in one or more tablets, and the amount of /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is selected from about 60mg to about 300mg, and that amount is administered once per day.
37. A method according to claim 36 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate is administered for from 1 to 7 consecutive days, followed by an optional drug holiday of from 1 to 14 days, followed by administration of N- {(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H- pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, for 1 day, optionally followed by one or more cycles of repeat dosing.
38. A method according to claim 34 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
39. A method according to claim 35 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
40. A method according to claim 36 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
41 . A method according to claim 37 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
42. A method according to claim 36 wherein /\/-{(1 S)-2-amino-1 -[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, is administered for from 1 to 3 consecutive days, followed by an optional drug holiday, followed by administration of N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2- (methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate, for from 3 to 7 days, optionally followed by one or more cycles of repeat dosing.
43. A method according to claim 42 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
44. A method according to claim 29 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate is administered for from 1 to 21 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of N- {(1 S)-2-amino-1 -[(3,4-difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1 -methyl-1 H- pyrazol-5-yl)-2-furancarboxamide, or a pharmaceutically acceptable salt thereof, from 1 to 21 days, optionally followed by one or more cycles of repeat dosing.
45. A method according to claim 44 wherein the cancer selected from ovarian, breast, pancreatic and prostate.
46. A method according to claim 9 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for from 4 to 6 consecutive days, optionally followed by one or more cycles of repeat dosing.
47. A method according to claim 8 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 days over a 7 day period, and during the other days of the 7 day period N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate is administered alone, optionally followed by one or more cycles of repeat dosing.
48. A method according to claim 13 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for from 4 to 6 consecutive days, optionally followed by one or more cycles of repeat dosing.
49. A method according to claim 12 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 days over a 7 day period, and during the other days of the 7 day period N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate is administered alone, optionally followed by one or more cycles of repeat dosing.
50. A method according to claim 21 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for from 4 to 6 consecutive days, optionally followed by one or more cycles of repeat dosing.
51 . A method according to claim 20 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 2 days over a 7 day period, and during the other days of the 7 day period N-{3-Chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-
(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine ditosylate monohydrate is administered alone, optionally followed by one or more cycles of repeat dosing.
52. A method according to claim 8 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 5 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for 2 consecutive days, optionally followed by one or more cycles of repeat dosing.
53. A method according to claim 12 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 5 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for 2 consecutive days, optionally followed by one or more cycles of repeat dosing.
54. A method according to claim 20 wherein N-{3-Chloro-4-[(3- fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate and /\/-{(1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl}-5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- furancarboxamide, or a pharmaceutically acceptable salt thereof, are administered within 12 hours of each other for 5 consecutive days followed by administration of N-{3-Chloro- 4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methanesulphonyl)ethyl]amino}methyl)-2-furyl]-4- quinazolinamine ditosylate monohydrate for 2 consecutive days, optionally followed by one or more cycles of repeat dosing.
55. A combination according to claim 4 wherein the compound of Structure (I) and the compound of Structure (II) are administered within 12 hours of each other for at least 5 consecutive days.
56. A combination according to claim 4 wherein the compound of Structure (I) and the compound of Structure (II) are administered within 12 hours of each other for at least 7 consecutive days.
57. A combination according to claim 4 wherein the compound of Structure (I) and the compound of Structure (II) are administered within 12 hours of each other for at least 14 consecutive days.
PCT/US2013/065827 2012-10-22 2013-10-21 Combination WO2014066202A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
KR1020157013079A KR20150074097A (en) 2012-10-22 2013-10-21 Combination
EP13848997.6A EP2908815A4 (en) 2012-10-22 2013-10-21 Combination
JP2015538098A JP2015534986A (en) 2012-10-22 2013-10-21 combination
CA2889051A CA2889051A1 (en) 2012-10-22 2013-10-21 Combination
MX2015005113A MX2015005113A (en) 2012-10-22 2013-10-21 Combination.
RU2015119245A RU2015119245A (en) 2012-10-22 2013-10-21 COMBINATION
CN201380067522.7A CN104902899A (en) 2012-10-22 2013-10-21 Combination
BR112015008924A BR112015008924A2 (en) 2012-10-22 2013-10-21 combination
AU2013334943A AU2013334943A1 (en) 2012-10-22 2013-10-21 Combination
IN3909DEN2015 IN2015DN03909A (en) 2012-10-22 2013-10-21
US14/437,297 US20150272952A1 (en) 2012-10-22 2013-10-21 Combination

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261716780P 2012-10-22 2012-10-22
US61/716,780 2012-10-22

Publications (1)

Publication Number Publication Date
WO2014066202A1 true WO2014066202A1 (en) 2014-05-01

Family

ID=50545138

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/065827 WO2014066202A1 (en) 2012-10-22 2013-10-21 Combination

Country Status (12)

Country Link
US (1) US20150272952A1 (en)
EP (1) EP2908815A4 (en)
JP (1) JP2015534986A (en)
KR (1) KR20150074097A (en)
CN (1) CN104902899A (en)
AU (1) AU2013334943A1 (en)
BR (1) BR112015008924A2 (en)
CA (1) CA2889051A1 (en)
IN (1) IN2015DN03909A (en)
MX (1) MX2015005113A (en)
RU (1) RU2015119245A (en)
WO (1) WO2014066202A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011044414A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
WO2012095505A1 (en) * 2011-01-12 2012-07-19 Boehringer Ingelheim International Gmbh Anticancer therapy with dual aurora kinase / mek inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UY30892A1 (en) * 2007-02-07 2008-09-02 Smithkline Beckman Corp AKT ACTIVITY INHIBITORS
WO2011060380A1 (en) * 2009-11-14 2011-05-19 The Regents Of The University Of California Pik3ca mutation status and sash1 expression predicts synergy between lapatinib and an akt inhibitor in her2 positive breast cancer
US20130072507A1 (en) * 2010-05-21 2013-03-21 Glaxosmithkline Llc Combination

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011044414A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
WO2012095505A1 (en) * 2011-01-12 2012-07-19 Boehringer Ingelheim International Gmbh Anticancer therapy with dual aurora kinase / mek inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
OPDAM, FL ET AL.: "Lapatinib for Advanced or Metastatic Breast Cancer.", THE ONCOLOGIST, vol. 17, 3 April 2012 (2012-04-03), pages 536 - 542, XP055249889, Retrieved from the Internet <URL:https://theoncologist.alphamedpress.org/content/17/4/536.full> [retrieved on 20141222] *
See also references of EP2908815A4 *

Also Published As

Publication number Publication date
JP2015534986A (en) 2015-12-07
BR112015008924A2 (en) 2017-07-04
EP2908815A1 (en) 2015-08-26
CA2889051A1 (en) 2014-05-01
RU2015119245A (en) 2016-12-10
CN104902899A (en) 2015-09-09
EP2908815A4 (en) 2016-06-22
IN2015DN03909A (en) 2015-10-02
KR20150074097A (en) 2015-07-01
AU2013334943A1 (en) 2015-05-14
US20150272952A1 (en) 2015-10-01
MX2015005113A (en) 2015-10-29

Similar Documents

Publication Publication Date Title
US20160213672A1 (en) Combination
AU2010303363B2 (en) Combination
EP2571358B1 (en) Combination therapy for treating cancer
EP2485594B1 (en) Combination
US20130137701A1 (en) Combination
US20150272952A1 (en) Combination
US20150094283A1 (en) Combination
WO2011038085A1 (en) Combination
US20150272950A1 (en) Combination

Legal Events

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

Ref document number: 13848997

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2889051

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015538098

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14437297

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/005113

Country of ref document: MX

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015008924

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2013848997

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2013848997

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013334943

Country of ref document: AU

Date of ref document: 20131021

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20157013079

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015119245

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015008924

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150420