US20130288984A1 - Combination - Google Patents

Combination Download PDF

Info

Publication number
US20130288984A1
US20130288984A1 US13/978,765 US201213978765A US2013288984A1 US 20130288984 A1 US20130288984 A1 US 20130288984A1 US 201213978765 A US201213978765 A US 201213978765A US 2013288984 A1 US2013288984 A1 US 2013288984A1
Authority
US
United States
Prior art keywords
cancer
compound
administered
chloro
methyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US13/978,765
Other languages
English (en)
Inventor
Rakesh Kumar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Novartis Pharma AG
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 US13/978,765 priority Critical patent/US20130288984A1/en
Assigned to GLAXOSMITHKLINE LLC reassignment GLAXOSMITHKLINE LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KUMAR, RAKESH
Publication of US20130288984A1 publication Critical patent/US20130288984A1/en
Assigned to NOVARTIS PHARMA AG reassignment NOVARTIS PHARMA AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GLAXOSMITHKLINE LLC
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMA AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/005Enzyme inhibitors
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

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 a proteasome inhibiting compound, and the Akt inhibitor: N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt thereof, and optional additional antineoplastic agents; 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).
  • 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 Bcl-x L , 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)).
  • 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, suggesting 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. Natl. 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)).
  • proteasome inhibitors are drugs that block the action of proteasomes, cellular complexes that break down proteins, like the p53 protein. Several proteasome inhibitors are marketed or are being studied in the treatment of cancer.
  • 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 a proteasome inhibiting compound, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents, 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 a proteasome inhibiting compound, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride 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 a proteasome inhibiting compound, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to such human,
  • FIG. 1 shows the combination of bortezomib and Compound C in Multiple Myeloma Cell Lines.
  • FIG. 2 shows the combination of bortezomib and Compound C in NCI-H929 Multiple Myeloma cell xenografts.
  • FIG. 3 shows the Combination of bortezomib and Compound C in RPMI-8226 Multiple Myeloma cell xenografts.
  • the present invention relates to combinations that exhibit antiproliferative activity.
  • the method relates to methods of treating cancer by the co-administration of a proteasome inhibiting compound, suitably bortezomib (Velcade®), (hereinafter Compound A, or a pharmaceutically acceptable salt thereof,
  • Compound A is sold commercially for the treatment of cancer.
  • Compound A is known by the generic name bortezomib and the trade name Velcade®.
  • 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 Feb. 7, 2008; International Publication Number WO 2008/098104 and an International Publication date of Aug. 14, 2008, the entire disclosure of which is hereby incorporated by reference, Compound B is the compound of example 96. Compound B can be prepared as described in International Application No. PCT/US2008/053269.
  • Compound B is in the form of a hydrochloride salt.
  • the salt form can be prepared by one of skill in the art from the description in United States Patent Application Publication: US 2010/0197754 A1, filed 28 Jan. 2010, having a publication date of Aug. 5, 2010.
  • Compound C means N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide hydrochloride.
  • the administration of a therapeutically effective amount of the combinations of the invention are advantageous over the individual component compounds in that the combinations 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 proteasome inhibiting compounds, suitably Compound A, and pharmaceutically acceptable salts thereof, and Compound B, and pharmaceutically acceptable salts thereof and optional additional antineoplastic agents.
  • 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 optional additional antineoplastic agents) 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, 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 salt thereof, and/or Compound B or a pharmaceutically acceptable salt thereof or optional additional antineoplastic agents 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.
  • day refers 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 or combination 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.
  • 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.
  • ком ⁇ онент and derivatives thereof, unless otherwise defined, is meant either, simultaneous administration or any manner of separate sequential administration of a therapeutically effective amount of Compound A, or a pharmaceutically acceptable salt thereof, and Compound B or a pharmaceutically acceptable salt thereof and optional additional antineoplastic agents.
  • 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.
  • Compound A is administered intravenously and Compound B is administered orally.
  • the combination kit as used herein is meant the pharmaceutical composition or compositions that are used to administer Compound A, or a pharmaceutically acceptable salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof and optional additional antineoplastic agents, according to the invention.
  • the combination kit can contain Compound A, or a pharmaceutically acceptable 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 salt thereof, and Compound B, or a pharmaceutically acceptable salt thereof, and optional additional antineoplastic agents, in separate pharmaceutical compositions.
  • the combination kit can comprise Compound A, or a pharmaceutically acceptable 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.
  • Compound B or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier.
  • a first container comprising Compound A, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier;
  • 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.
  • 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 2 means—Compound A, or a pharmaceutically acceptable salt thereof or another proteasome inhibitor as defined herein, suitably Compound A—.
  • Compound B 2 means—Compound B, or a pharmaceutically acceptable salt thereof—.
  • Compound B 2 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 U.S. Pat. No. 7,576,209 which issued on Aug. 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 U.S. Pat. No. 7,576,209.
  • proteasome inhibiting compound By the term “proteasome inhibiting compound”, “proteasome inhibitors” and derivatives thereof, as used herein, unless otherwise defined, is meant the class of compounds that block the action of proteasomes, cellular complexes that break down proteins, like the p53 protein.
  • proteasome inhibitors are marketed or are being studied in the treatment of cancer. Suitable proteasome inhibitors for use herein include:
  • Bortezomib has the following chemical structure and name.
  • Epigallocatechin gallate has the following chemical structure and name.
  • Salinosporamide A has the following chemical structure and name.
  • Carfilzomib has the following chemical structure and name.
  • additional antineoplastic agent(s) and derivatives thereof, as used herein, unless otherwise defined, is meant further compounds known to treat cancer or to be useful when administered with a compound or compounds that are known to treat cancer.
  • additional antineoplastic agents for use herein include: gemcitabine, carboplatin, cisplatin, cytarabine, thalidomide, Revlimid® (lenalidomide), decitabine, melphalan (Alkeran), cyclophosphamide (Cytoxan), vincristine (Oncovin), doxorubicin (Adriamycin), liposomal doxorubicin (Doxil), prednisone, dexamethasone (Decadron) and eltrombopag (Promacta®).
  • the additional antineoplastic agent is an agent useful in the treatment of multiple myeloma.
  • the specified period is meant the interval of time between the administration of one of Compound A 2 and Compound B 2 and the other of Compound A 2 and Compound B 2 .
  • the specified period can include simultaneous administration.
  • the specified period refers to timing of the administration of Compound A 2 and Compound B 2 during a single 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.
  • 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 11 hours of each other—in this case, the specified period will be about 11 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
  • 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, 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.
  • both compounds will be administered within a specified period for at least 1 day, followed by the administration of Compound A 2 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 specified period for at least 1 day, followed by administration of Compound A 2 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 least 1 day, followed by administration of Compound A 2 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 by administration of Compound A 2 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 Compound A 2
  • both compounds will be administered within a specified period for from 1 to 3 consecutive days, followed by administration of Compound A 2 alone for from 3 to 7 consecutive days.
  • both compounds will be administered within a specified period for from 3 to 6 consecutive days, followed by administration of Compound A 2 alone for from 1 to 4 consecutive days.
  • both compounds will be administered within a specified period for 5 consecutive days, followed by administration of Compound A 2 alone for 2 consecutive days.
  • both compounds will be administered within a specified period for 2 consecutive days, followed by administration of Compound A 2 alone for from 3 to 7 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 7 day period Compound A 2 will be administered alone.
  • both compounds will be administered within a specified period for 2 days over a 7 day period, and during the other days of the 7 day period Compound A 2 will be administered alone.
  • both compounds will be administered within a specified period for 2 days over a 7 day period, and during the other days of the 7 day period Compound B 2 will be administered alone.
  • both compounds will be administered within a specified period for 1 day over a 7 day period, and during the other days of the 7 day period Compound B 2 will be administered alone.
  • compositions when Compound A 2 and Compound B 2 are administered for a specified period, optional additional antineoplastic agents can be administered at any time during the course of treatment or during the duration of time as indicated herein.
  • the compounds are not administered during a “specified period”, they are administered sequentially.
  • sequential administration and derivates thereof, as used herein is meant that one of Compound A 2 and Compound B 2 is administered for one or more consecutive days and the other of Compound A 2 and Compound B 2 is subsequently administered for one or more consecutive days.
  • a drug holiday utilized between the sequential administration of one of Compound A 2 and Compound B 2 and the other of Compound A 2 and Compound B 2 .
  • a drug holiday is a period of days after the sequential administration of one of Compound A 2 and Compound B 2 and before the administration of the other of Compound A 2 and Compound B 2 where neither Compound A 2 nor Compound B 2 is administered.
  • 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, 11 days, 12 days, 13 days and 14 days.
  • one of Compound A 2 and Compound B 2 is administered for from 1 to 30 consecutive days, followed by an optional drug holiday, followed by administration of the other of Compound A 2 and Compound B 2 for from 1 to 30 consecutive days.
  • one of Compound A 2 and Compound B 2 is administered for from 1 to 21 consecutive days, followed by an optional drug holiday, followed by administration of the other of Compound A 2 and Compound B 2 for from 1 to 21 consecutive days.
  • one of Compound A 2 and Compound B 2 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 2 and Compound B 2 for from 1 to 14 consecutive days.
  • one of Compound A 2 and Compound B 2 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 A 2 and Compound B 2 for from 2 to 7 consecutive days.
  • Compound B 2 will be administered first in the sequence, followed by an optional drug holiday, followed by administration of Compound A 2 .
  • Compound B 2 is administered for from 1 to 21 consecutive days, followed by an optional drug holiday, followed by administration of Compound A 2 for from 1 to 21 consecutive days.
  • Compound B 2 is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of Compound A 2 for from 3 to 21 consecutive days.
  • Compound B 2 is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by administration of Compound A 2 for from 3 to 21 consecutive days.
  • Compound B 2 is administered for 21 consecutive days, followed by an optional drug holiday, followed by administration of Compound A 2 for 14 consecutive days.
  • Compound B 2 is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of Compound A 2 for 14 consecutive days.
  • Compound B 2 is administered for 7 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound A 2 for 7 consecutive days.
  • Compound B 2 is administered for 3 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by administration of Compound A 2 for 7 consecutive days.
  • Compound B 2 is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound A 2 for 3 consecutive days.
  • Compound A 2 will be administered first in the sequence, followed by an optional drug holiday, followed by administration of Compound B 2 .
  • Compound A 2 is administered for from 1 to 21 consecutive days, followed by an optional drug holiday, followed by administration of Compound B 2 for from 1 to 21 consecutive days.
  • Compound A 2 is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of Compound B 2 for from 3 to 21 consecutive days.
  • Compound A 2 is administered for from 3 to 21 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by administration of Compound B 2 for from 3 to 21 consecutive days.
  • Compound A 2 is administered for 21 consecutive days, followed by an optional drug holiday, followed by administration of Compound B 2 for 14 consecutive days.
  • Compound A 2 is administered for 14 consecutive days, followed by a drug holiday of from 1 to 14 days, followed by administration of Compound B 2 for 14 consecutive days.
  • Compound A 2 is administered for 7 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound B 2 for 7 consecutive days.
  • Compound A 2 is administered for 3 consecutive days, followed by a drug holiday of from 3 to 14 days, followed by administration of Compound B 2 for 7 consecutive days.
  • Compound A 2 is administered for 3 consecutive days, followed by a drug holiday of from 3 to 10 days, followed by administration of Compound B 2 for 3 consecutive days.
  • Compound A 2 is administered for 7 consecutive days, followed by administration of Compound B 2 for 1 day.
  • Compound A 2 is administered for 6 consecutive days, followed by administration of Compound B 2 for 1 day.
  • Compound B 2 is administered for 1 day, followed by administration of Compound A 2 for 7 consecutive days.
  • Compound B 2 is administered for 1 day, followed by administration of Compound A 2 for 6 consecutive days.
  • a “specified period” administration and a “sequential” administration can be followed by 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 2 administered as part of the combination according to the present invention will be an efficacious, non toxic amount administered from 1 to 4 times a day.
  • the selected amount of Compound A 2 is administered twice a day.
  • the selected amount of Compound A 2 is administered once a day.
  • the selected amount of Compound A 2 is administered once a week intravenously.
  • the selected amount of Compound A 2 is administered twice a week intravenously.
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week.
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount selected from 0.5 to 1.3 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 1 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 1.3 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 0.6 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 0.7 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 0.8 mg/m 2 .
  • Compound A 2 is Velcade® and is administered intravenously once or twice a week in an amount of 0.9 mg/m 2 .
  • the amount of Compound B 2 administered as part of the combination according to the present invention will be an amount selected from about 5 mg to about 500 mg; suitably, the amount will be selected from about 25 mg to about 400 mg; suitably, the amount will be selected from about 30 mg to about 375 mg; suitably, the amount will be selected from about 35 mg to about 350 mg; suitably, the amount will be selected from about 40 mg to about 300 mg; suitably, the amount will be selected from about 45 mg to about 275 mg; suitably, the amount will be selected from about 50 mg to about 250 mg; suitably, the amount will be selected from about 55 mg to about 225 mg; suitably, the amount will be selected from about 60 mg to about 200 mg; suitably, the amount will be selected from about 65 mg to about 175 mg; suitably, the amount will be selected from about 70 mg to about 150 mg; suitably, the amount will be selected from about 50 mg to about 300 mg; suitably, the amount will be selected from about 75 mg to about 150 mg; suitably, the amount will be about 100 mg.
  • the amount of Compound B 2 administered as part of the combination according to the present invention will be an amount selected from about 5 mg to about 500 mg.
  • the amount of Compound B 2 administered as part of the combination according to the present invention can be 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, 125 mg, 130 mg, 135 mg, 140 mg, 145 mg, 150 mg, 175 mg, 200 mg, 225 mg, 250 mg, 275 mg, 300 mg, 325 mg, 350 mg, 375 mg, 400 mg, 425 mg, 450 mg, 475 mg or 500 mg.
  • the selected amount of Compound B 2 is administered twice a day.
  • the selected amount of Compound B 2 is administered once a day.
  • the method of the present invention may also be employed with other therapeutic methods of cancer treatment.
  • the invention further provides pharmaceutical compositions, which include Compound A 2 and/or Compound B 2 , 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 2 and/or Compound B 2 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.
  • 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 2 and Compound B 2 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 same or different routes and that Compound A 2 and Compound B 2 may be compounded together in a pharmaceutical composition/formulation. Suitably, Compound A 2 and Compound B 2 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.
  • 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.
  • therapeutically effective amounts of the combinations of the invention 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.
  • AKT inhibitor Compound B
  • SID once daily
  • BID twice daily
  • Compound A 2 is administered at 0.5 or 1.0 mg/kg, intraperitoneally at twice/week for the duration of study either alone or in combination with Compound B.
  • Optional additional antineoplastic agents may also be added.
  • the combinations of the invention are tested for efficacy, advantageous and synergistic properties generally according to the following combination cell proliferation assays.
  • Cells are plated in 96 or 384-well plates at 500-5000 cells/well in culture media appropriate for each cell type, supplemented with 10% FBS and 1% penicillin/streptomycin, and incubated overnight at 37° C., 5% CO 2 .
  • Cells are treated in a grid manner with dilution of Compound A 2 (8 dilutions, including no compound, of 3-fold dilutions starting from 0.1-30 ⁇ M depending on combination) from left to right on 96-well plate and also treated with Compound B 2 (8 dilutions, including no compound, of 3-fold dilutions starting from 1-30 ⁇ M depending on combination) from top to bottom on 96-well plate and incubated as above for a further 72 hours.
  • Optional additional antineoplastic agents may also be added. In some instances compounds are added in a staggered manner and incubation time can be extended up to 7 days.
  • Cell growth is measured using CellTiter-Glo® reagent according to the manufacturer's protocol and signals are read on a PerkinElmer EnVisionTM reader set for luminescence mode with a 0.5-second read. Data are analyzed as described below.
  • the cellular response is determined for each compound and/or compound combination using a 4- or 6-parameter curve fit of cell viability against concentration using the IDBS XLfit plug-in for Microsoft Excel software and determining the concentration required for 50% inhibition of cell growth (gIC 50 ). Background correction is made by subtraction of values from wells containing no cells.
  • CI Combination Index
  • EHSA Excess Over Highest Single Agent
  • EOBliss Excess Over Bliss
  • 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 for treating or lessening the severity of multiple myeloma.
  • This invention provides a combination comprising a proteasome inhibiting compound, suitably bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents.
  • a proteasome inhibiting compound suitably bortezomib
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents.
  • This invention also provides for a combination comprising bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof.
  • This invention also provides for a combination comprising bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, for use in treating cancer.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a combination of bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof.
  • This invention also provides a combination kit comprising a proteasome inhibiting compound, suitably bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents.
  • a proteasome inhibiting compound suitably bortezomib
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents.
  • This invention also provides for the use of a combination comprising a proteasome inhibiting compound, suitably bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, in the manufacture of a medicament.
  • a proteasome inhibiting compound suitably bortezomib
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, in the manufacture of a medicament.
  • This invention also provides for the use of a combination comprising a proteasome inhibiting compound, suitably bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, in the manufacture of a medicament to treat cancer.
  • a proteasome inhibiting compound suitably bortezomib
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide or a pharmaceutically acceptable salt, suitably the hydrochloride 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 a proteasome inhibiting compound, suitably bortezomib, and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, and optional additional antineoplastic agents to a subject in need thereof.
  • a proteasome inhibiting compound suitably bortezomib
  • An oral dosage form for administering a combination of the present invention is produced by filling 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.
  • An injectable form for administering a presently invented combinations is produced by stirring 1.5% by weight of (Compound A 2 ) and N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide hydrochloride (the hydrochloride salt of Compound B) in 10% by volume propylene glycol in water.
  • An injectable form for administering a compound of the presently invented combinations is produced by stirring 1.5% by weight of (Compound A 2 ) in 10% by volume propylene glycol in water.
  • An injectable form for administering a compound of the presently invented combinations is produced by stirring 1.5% by weight of N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide hydrochloride (the hydrochloride salt of Compound B) in 10% by volume propylene glycol in water.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US13/978,765 2011-01-11 2012-01-11 Combination Abandoned US20130288984A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/978,765 US20130288984A1 (en) 2011-01-11 2012-01-11 Combination

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161431508P 2011-01-11 2011-01-11
US13/978,765 US20130288984A1 (en) 2011-01-11 2012-01-11 Combination
PCT/US2012/020863 WO2012097021A1 (en) 2011-01-11 2012-01-11 Combination

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/020863 A-371-Of-International WO2012097021A1 (en) 2011-01-11 2012-01-11 Combination

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/831,261 Continuation US20160022641A1 (en) 2011-01-11 2015-08-20 Combination

Publications (1)

Publication Number Publication Date
US20130288984A1 true US20130288984A1 (en) 2013-10-31

Family

ID=46507420

Family Applications (6)

Application Number Title Priority Date Filing Date
US13/978,765 Abandoned US20130288984A1 (en) 2011-01-11 2012-01-11 Combination
US13/368,434 Abandoned US20120258933A1 (en) 2011-01-11 2012-02-08 Combination
US14/590,143 Abandoned US20150119337A1 (en) 2011-01-11 2015-01-06 Combination
US14/831,261 Abandoned US20160022641A1 (en) 2011-01-11 2015-08-20 Combination
US15/259,694 Abandoned US20160375086A1 (en) 2011-01-11 2016-09-08 Combination
US15/385,205 Active US10449226B2 (en) 2011-01-11 2016-12-20 Combination

Family Applications After (5)

Application Number Title Priority Date Filing Date
US13/368,434 Abandoned US20120258933A1 (en) 2011-01-11 2012-02-08 Combination
US14/590,143 Abandoned US20150119337A1 (en) 2011-01-11 2015-01-06 Combination
US14/831,261 Abandoned US20160022641A1 (en) 2011-01-11 2015-08-20 Combination
US15/259,694 Abandoned US20160375086A1 (en) 2011-01-11 2016-09-08 Combination
US15/385,205 Active US10449226B2 (en) 2011-01-11 2016-12-20 Combination

Country Status (15)

Country Link
US (6) US20130288984A1 (ko)
EP (1) EP2663189B1 (ko)
JP (1) JP6100700B2 (ko)
KR (1) KR20140053836A (ko)
CN (1) CN103298345B (ko)
AU (1) AU2012205601B2 (ko)
BR (1) BR112013017722A2 (ko)
CA (1) CA2824201A1 (ko)
EA (1) EA029000B1 (ko)
ES (1) ES2689760T3 (ko)
IL (1) IL227097B (ko)
MX (1) MX356704B (ko)
SG (2) SG191724A1 (ko)
WO (1) WO2012097021A1 (ko)
ZA (1) ZA201304139B (ko)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160228456A1 (en) * 2013-10-01 2016-08-11 Novartis Ag Combination
KR20210063475A (ko) * 2013-11-07 2021-06-01 데시페라 파마슈티칼스, 엘엘씨. 암치료에 유용한 tie2 키나아제의 억제 방법
CA3089566A1 (en) 2018-01-31 2019-08-08 Deciphera Pharmaceuticals, Llc Combination therapy for the treatment of gastrointestinal stromal tumors
JP2022544234A (ja) 2019-08-12 2022-10-17 デシフェラ・ファーマシューティカルズ,エルエルシー 胃腸間質腫瘍を治療するためのリプレチニブ
WO2021030405A1 (en) 2019-08-12 2021-02-18 Deciphera Pharmaceuticals, Llc Ripretinib for treating gastrointestinal stromal tumors
KR20220123058A (ko) 2019-12-30 2022-09-05 데시페라 파마슈티칼스, 엘엘씨. 1-(4-브로모-5-(1-에틸-7-(메틸아미노)-2-옥소-1,2-디히드로-1,6-나프티리딘-3-일)-2-플루오로페닐)-3-페닐우레아의 조성물
EP4084778B1 (en) 2019-12-30 2023-11-01 Deciphera Pharmaceuticals, LLC Amorphous kinase inhibitor formulations and methods of use thereof
US11779572B1 (en) 2022-09-02 2023-10-10 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5604213A (en) 1992-03-31 1997-02-18 British Technology Group Limited 17-substituted steroids useful in cancer treatment
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
GB9716557D0 (en) 1997-08-06 1997-10-08 Glaxo Group Ltd Benzylidene-1,3-dihydro-indol-2-one derivatives having anti-cancer activity
US6328590B1 (en) 2000-07-20 2001-12-11 Emc Corporation Methods and apparatus for controlling attachment of an electronic module with a circuit board connector
JP5136929B2 (ja) * 2004-03-24 2013-02-06 アボット・ラボラトリーズ 三環式ピラゾール系キナーゼ阻害薬
EP1802589B1 (en) 2004-10-13 2014-04-09 Pharmacia & Upjohn Company LLC Crystalline forms of 3-[5-chloro-4-[(2,4-difluorobenzyl) oxy]-6-oxopyrimidin-1(6h)-yl]-n-(2-hydroxyethyl)-4-methylbenzamide
US7709517B2 (en) 2005-05-13 2010-05-04 The Regents Of The University Of California Diarylhydantoin compounds
CA2620333A1 (en) * 2005-08-26 2007-03-01 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP1840114A1 (en) 2006-03-30 2007-10-03 LTB4 Sweden AB Crystalline Leukotriene B4
BRPI0710291A2 (pt) 2006-04-05 2011-08-09 Novartis Ag combinações de agente terapêuticos para tratamento de cáncer
WO2007122686A1 (ja) 2006-04-14 2007-11-01 Eisai R & D Management Co., Ltd. ベンズイミダゾール化合物
US20080051380A1 (en) 2006-08-25 2008-02-28 Auerbach Alan H Methods and compositions for treating cancer
DK2061561T3 (da) 2006-08-25 2013-10-07 Janssen Oncology Inc Sammensætninger til behandling af cancer
JP4299327B2 (ja) 2006-08-31 2009-07-22 トヨタ自動車株式会社 可変バルブタイミング装置
US8828451B2 (en) * 2006-10-04 2014-09-09 University Of South Florida Akt sensitization of cancer cells
PE20080859A1 (es) 2006-10-13 2008-09-04 Basf Ag Hidratos de 2-cloro-5-[3,6-dihidro-3-metil-2,6-dioxo-4-(trifluorometil)-1-(2h)-pirimidinil]-4-fluor-n-[[metil(1-metiletil)amino]sulfonil]benzamida
AR064010A1 (es) 2006-12-06 2009-03-04 Merck & Co Inc Inhibidores de la actividad de la akt
UY30892A1 (es) 2007-02-07 2008-09-02 Smithkline Beckman Corp Inhibidores de la actividad akt
EP2201012B1 (en) 2007-10-11 2014-06-25 AstraZeneca AB Pyrrolo[2,3-d]pyrimidin derivatives as protein kinase b inhibitors
US20090131367A1 (en) * 2007-11-19 2009-05-21 The Regents Of The University Of Colorado Combinations of HDAC Inhibitors and Proteasome Inhibitors
DE102008044901A1 (de) 2008-08-29 2010-03-04 Siemens Aktiengesellschaft Verfahren und Vorrichtung zur Auswahl eines Bestrahlungsplans sowie Bestrahlungsanlage
EP2400964A4 (en) 2009-01-30 2012-08-01 Glaxosmithkline Llc CRYSTALLINE FORM OF N - {(1-5) -2-AMINO-1 - [(3-FLUOROPHENYL) METHYL] ETHYL} -5-CHLORO-4- (4-CHLORO-1-METHYL-1H-PYRAZOL) CHLOROHYDRATE 5-YL) -2-THIOPHENE CARBOXAMIDE
UA105794C2 (uk) 2009-06-26 2014-06-25 Новартіс Аг 1,3-ДИЗАМІЩЕНІ ПОХІДНІ ІМІДАЗОЛІДИН-2-ОНУ ЯК ІНГІБІТОРИ Cyp17
BR112012008274A2 (pt) 2009-10-08 2015-09-22 Glaxosmithkline Llc combinação
CN104586861A (zh) 2011-04-01 2015-05-06 基因泰克公司 Akt抑制剂化合物和阿比特龙的组合及使用方法
CN103813788A (zh) 2011-05-03 2014-05-21 罗切斯特大学 治疗***癌的方法
EA201490265A1 (ru) 2011-07-13 2014-12-30 Фармасайкликс, Инк. Ингибиторы тирозинкиназы брутона
SI2766381T1 (sl) 2011-10-10 2016-11-30 Zach System, Z.I. Postopek za pripravo 17-substituiranih steroidov
WO2013079964A1 (en) 2011-11-30 2013-06-06 Astrazeneca Ab Combination treatment of cancer
BR112014030424A8 (pt) 2012-06-04 2017-07-11 Pharmacyclics Inc Formas cristalinas de um inibidor de quinase de tirosina de bruton
KR101797415B1 (ko) 2013-10-01 2017-11-13 노파르티스 아게 암의 치료를 위한 아푸레세르팁과 조합된 엔잘루타미드

Also Published As

Publication number Publication date
US10449226B2 (en) 2019-10-22
US20160375086A1 (en) 2016-12-29
MX2013008074A (es) 2013-08-09
US20170100449A1 (en) 2017-04-13
KR20140053836A (ko) 2014-05-08
EP2663189A4 (en) 2014-06-11
EA201391027A1 (ru) 2014-01-30
AU2012205601A1 (en) 2013-08-01
ZA201304139B (en) 2014-02-26
CA2824201A1 (en) 2012-07-19
JP2014507412A (ja) 2014-03-27
WO2012097021A1 (en) 2012-07-19
JP6100700B2 (ja) 2017-03-22
SG191724A1 (en) 2013-08-30
BR112013017722A2 (pt) 2016-07-12
US20120258933A1 (en) 2012-10-11
MX356704B (es) 2018-06-11
CN103298345A (zh) 2013-09-11
CN103298345B (zh) 2016-12-14
EP2663189B1 (en) 2018-07-04
US20160022641A1 (en) 2016-01-28
EP2663189A1 (en) 2013-11-20
ES2689760T3 (es) 2018-11-15
US20150119337A1 (en) 2015-04-30
SG10201600077RA (en) 2016-02-26
AU2012205601B2 (en) 2016-03-24
EA029000B1 (ru) 2018-01-31
IL227097B (en) 2018-02-28

Similar Documents

Publication Publication Date Title
US10449226B2 (en) Combination
US20160213672A1 (en) Combination
EP2485593B1 (en) Combination
EP2485594B1 (en) Combination
US20150094283A1 (en) Combination
US20150272950A1 (en) Combination

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE LLC, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KUMAR, RAKESH;REEL/FRAME:027607/0355

Effective date: 20120112

AS Assignment

Owner name: NOVARTIS PHARMA AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GLAXOSMITHKLINE LLC;REEL/FRAME:035806/0473

Effective date: 20150302

AS Assignment

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:035812/0424

Effective date: 20150302

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION