WO2012106302A1 - Combination - Google Patents

Combination Download PDF

Info

Publication number
WO2012106302A1
WO2012106302A1 PCT/US2012/023261 US2012023261W WO2012106302A1 WO 2012106302 A1 WO2012106302 A1 WO 2012106302A1 US 2012023261 W US2012023261 W US 2012023261W WO 2012106302 A1 WO2012106302 A1 WO 2012106302A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
amount
administered
Prior art date
Application number
PCT/US2012/023261
Other languages
French (fr)
Inventor
Rakesh Kumar
Robert S. Kerbel
Original Assignee
Glaxo Wellcome Manufacturing Pte Ltd
Sunnybrook Research Institute
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 Glaxo Wellcome Manufacturing Pte Ltd, Sunnybrook Research Institute filed Critical Glaxo Wellcome Manufacturing Pte Ltd
Priority to US13/980,904 priority Critical patent/US20130303560A1/en
Priority to EP12742225.1A priority patent/EP2672969A4/en
Priority to JP2013552574A priority patent/JP2014504638A/en
Publication of WO2012106302A1 publication Critical patent/WO2012106302A1/en
Priority to US15/291,463 priority patent/US20170027938A1/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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not 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
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles

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 VEGFR inhibitor and a topoisomerase inhibitor, 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).
  • Angiogenesis is the development of new blood vessels from the pre-existing vasculature.
  • Angiogenesis is defined herein as involving: (i) activation of endothelial cells; (ii) increased vascular permeability; (iii) subsequent dissolution of the basement membrane and extravasation of plasma components leading to formation of a provisional fibrin gel extracellular matrix; (iv) proliferation and mobilization of endothelial cells; (v) reorganization of mobilized endothelial cells to form functional capillaries; (vi) capillary loop formation; and (vi) deposition of basement membrane and recruitment of perivascular cells to newly formed vessels.
  • Normal angiogenesis is active during tissue growth from embryonic development through maturity and then enters a period of relative quiescence during adulthood. Normal angiogenesis is also activated during wound healing, and at certain stages of the female reproductive cycle. Inappropriate or pathological angiogenesis has been associated with several disease states including various retinopathies, ischemic disease, atherosclerosis, chronic inflammatory disorders, and cancer. The role of angiogenesis in disease states is discussed, for instance, in Fan et al., Trends in Pharmacol Sci. 16:54-66; Shawver et al., DDT Vol. 2, No.2 February 1997; Folkmann, 1995, Nature Medicine 1 :27-31.
  • VEGF vascular endothelial growth factor
  • VEGFRs vascular endothelial growth factor receptor(s)
  • VEGF is a polypeptide, which has been linked to inappropriate or pathological angiogenesis (Pinedo, H. M. et al. The Oncologist, Vol.5, No. 90001 , 1 -2, Apr. 2000).
  • VEGFR(s) are protein tyrosine kinases (PTKs) that catalyze the phosphorylation of specific tyrosine residues in proteins that are involved in the regulation of cell growth, differentiation, and survival.
  • PTKs protein tyrosine kinases
  • VEGFRI Flt-I
  • VEGFR2 Flk-I and KDR
  • VEGFR3 Flt-4
  • VEGFR2 is a transmembrane receptor PTK expressed primarily in endothelial cells.
  • VEGF vascular endothelial growth factor-2
  • VEGF expression may be constitutive to tumor cells and can also be upregulated in response to certain stimuli.
  • One such stimulus is hypoxia, where VEGF expression is upregulated in both tumor and associated host tissues.
  • the VEGF ligand activates VEGFR2 by binding to its extracellular VEGF binding site. This leads to receptor dimerization of VEGFRs and autophosphorylation of tyrosine residues at the intracellular kinase domain of VEGFR2.
  • the kinase domain operates to transfer a phosphate from ATP to the tyrosine residues, thus providing binding sites for signaling proteins downstream of VEGFR-2 leading ultimately to angiogenesis.
  • antagonism of the VEGFR2 kinase domain would block phosphorylation of tyrosine residues and serve to disrupt initiation of angiogenesis.
  • inhibition at the ATP binding site of the VEGFR2 kinase domain would prevent binding of ATP and prevent phosphorylation of tyrosine residues.
  • Such disruption of the proangiogenesis signal transduction pathway associated with VEGFR2 should therefore inhibit tumor angiogenesis and thereby provide a potent treatment for cancer or other disorders associated with inappropriate angiogenesis.
  • Votrient is a multi-tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR)-1 , VEGFR-2, VEGFR-3, platelet-derived growth factor receptor (PDGFR)-a and - ⁇ , fibroblast growth factor receptor (FGFR) -1 and -3, cytokine receptor (Kit), interleukin-2 receptor inducible T-cell kinase (Itk), leukocyte-specific protein tyrosine kinase (Lck), and transmembrane glycoprotein receptor tyrosine kinase (c-Fms) and is approved in the US for the treatment of patients with advanced renal cell carcinoma.
  • VEGFR vascular endothelial growth factor receptor
  • VEGFR-2 VEGFR-2
  • VEGFR-3 platelet-derived growth factor receptor
  • FGFR fibroblast growth factor receptor
  • FGFR fibroblast growth factor receptor
  • c-Fms transmembrane glycoprotein receptor t
  • pazopanib hydrochloride is 5-[[4-[(2,3-dimethyl-2H- indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide
  • the structure of the DNA helix within eukaryotic cells imposes certain topological problems that the cellular apparatus must solve in order to use its genetic material as a template.
  • the separation of the DNA strands is fundamental to cellular processes such as DNA replication and transcription. Since eukaryotic DNA is organized into chromatin by chromosomal proteins, the ends are constrained and the strands cannot unwind without the aid of enzymes that alter topology. It has long been recognized that the advancement of the transcription or replication complex along the DNA helix would be facilitated by a swivel point which would relieve the torsional strain generated during these processes.
  • Topoisomerases are enzymes that are capable of altering DNA topology in eukaryotic cells. They are critical for important cellular functions and cell proliferation.
  • Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single strand break, unwinds the double helix (or allows it to unwind), and subsequently reseals the break before dissociating from the DNA strand.
  • Topoisomerase II consists of two identical subunits of molecular weight 170,000. Topoisomerase II transiently breaks both strands of the helix and passes another double-strand segment through the break.
  • Camptothecin is a water- insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to
  • Camptothecin and a few close congeners thereof are the only class of compounds known to inhibit topoisomerase I. Inhibition of topoisomerase II is the major target of important commercial oncolytic agents (e.g., etoposide, doxorubicin and mitoxantrone) as well as other oncolytic agents still undergoing development. Camptothecin (and its known congeners) have no effect on topoisomerase II and none of the known topoisomerase II inhibitors has any significant effect on topoisomerase I.
  • Hycamtin® topotecan hydrochloride
  • campotothecin that exhibits topoisomerase l-inhibitory activity.
  • Hycamtin® is approved in the US for the treatment of relapsed small cell lung cancer, ovarian cancer and cervical cancer.
  • the chemical name for topotecan hydrochloride is (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 /-/- pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H,12/-/)-dione monohydrochloride.
  • One embodiment of this invention provides a method of treating breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonam or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to such human.
  • One embodiment of this invention provides a method of treating breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride 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 breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to such human, wherein the compounds of the combination are administered sequentially.
  • Figure 1 illustrates percent survival after an indicated number of days when treatment of Advanced Metastatic Breast Cancer (MDA-MB-231/LM2-4) was initiated at day 39, 19 days post primary tumor resection, for the control group, the low dose metronomic oral topotecan alone group, the maximum tolerated group, the pazopanib alone group, the low dose oral topotecan + pazopanib group, and the maximum tolerated dose topotecan + pazopanib group.
  • MDA-MB-231/LM2-4 Advanced Metastatic Breast Cancer
  • the present invention relates to combinations that exhibit antitumor activity.
  • the method relates to methods of treating breast cancer by the co- administration of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, (hereinafter Compound A, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof), which compound is represented by Structure I:
  • Compound A is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of VEGFR activity, particularly in treatment of cancer, in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001 , International Publication Number WO02/0591 10 and an International Publication date of August 1 , 2002, the entire disclosure of which is hereby incorporated by reference, Compound A is the compound of Example 69.
  • Compound A can be prepared as described in International Application No. PCT/US01/49367.
  • Compound A is in the form of a monohydrochloride salt. This salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001.
  • Compound A is sold commercially as the monohydrochloride salt.
  • Compound B is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of topoisomerase I, particularly in treatment of cancer, in US Patent No. 5,004,758, having a filing date of November 2, 1988, the entire disclosure of which is hereby incorporated by reference, Compound B is compound 1 S (as the acetate salt). Compound B can be prepared as described in US Patent No. 5,734,056.
  • 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 US Patent No. 5,004,758 and/or by methods that will be readily apparent to those skilled in the art.
  • Compound B is sold commercially as the monohydrochloride salt.
  • 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 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, ethanol and acetic acid.
  • the solvent used is a pharmaceutically acceptable solvent.
  • the solvent used is water.
  • contemplated herein is a method of treating breast 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 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 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 breast cancer, such as when a subject has a strong family history of breast 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.
  • 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, 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 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.
  • Compound A means — Compound A, or a pharmaceutically acceptable salt thereof— .
  • Compound B means — Compound B, or a pharmaceutically acceptable salt thereof— .
  • 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 can be various time periods.
  • Compound A 2 and Compound B 2 can be administered within about 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 hours of each other, in which case the specified period will be about 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9,
  • Compound A and Compound B in less than about 45 minutes apart is considered simultaneous administration.
  • 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.
  • the duration of time can be various time periods.
  • Compound A 2 and Compound B 2 can both be administered within a specified period for at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 consecutive days during the course of treatment, in which case the duration of time will be 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30, respectively.
  • both compounds are administered within a specified period for over 30 consecutive days, the treatment is considered chronic treatment and will continue until an altering event, such as a reassessment in breast cancer status or a change in the condition of the patient, warrants a modification to the protocol.
  • Compound A 2 and B 2 can be co-administered within a specified period for at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 ,
  • the duration of time will be at least the number of consecutive days that Compound A 2 and Compound B 2 are both administered plus the number of consecutive days of administration of Compound A 2 alone (e.g., if Compound A 2 and Compound B 2 are both administered for 6 consecutive days followed by administration of Compound A 2 alone for 8 consecutive days, the duration of time will be at least 14 consecutive days).
  • Compound A 2 and Compound B 2 are both administered within a specified period for a number of consecutive days during a certain time period, and compound A 2 is administered during the other days of the certain time period.
  • Compound A 2 and Compound B 2 can be administered within a specified time period for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 or 13 consecutive days over a certain time period of 14 days, during which Compound A 2 is administered for the other 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 days, respectively.
  • the consecutive days during which Compound A 2 and Compound B 2 are both administered within a specified time period can occur any time during the certain time period. Accordingly, in the foregoing example, Compound A 2 could be administered alone for 4 consecutive days follow by administration of both Compound A 2 and Compound B 2 for 5 consecutive days, followed by administering Compound A 2 alone for 5 consecutive days to complete the 14 day certain time period.
  • treatment protocols have been described with respect to administration of both Compound A 2 and Compound B 2 within a specified period in conjunction with administration of Compound A 2 alone, embodiments of the present invention also include similar treatment protocols in which Compound A 2 and Compound B 2 are both administered within a specified period in conjunction with administration of Compound B 2 alone.
  • compositions of the present invention include administration of both Compound A 2 and Compound B 2 within a specified period in conjunction with administration of Compound A 2 alone and administration of Compound B 2 alone.
  • Compound A 2 and Compound B 2 are both administered within a specified period for a number of consecutive days during a certain time period, Compound A 2 is administered alone during a number of days during the certain time period, and Compound B 2 is administered alone during the other days during the certain time period.
  • the days of administration of Compound B 2 is n - m - p, where n - m - p is at least 1.
  • Compound A 2 and Compound B 2 can both be administered within a specified time period for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 consecutive days over a certain time period of 14 days, during which Compound A 2 is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 days, and Compound B 2 is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 days.
  • the consecutive days during which Compound A 2 and Compound B 2 are both administered within a specified time period can occur any time during the certain time period. Accordingly, in the foregoing example, Compound A 2 could be administered alone for 4 consecutive days follow by administration of both Compound A 2 and Compound B 2 for 5 consecutive days, followed by administering Compound B 2 alone for 5 consecutive days to complete the 14 day certain time period. Administration of Compound A 2 alone and administration of Compound B 2 alone do not have to occur on consecutive days.
  • Compound A 2 could be administered for 2 consecutive days, followed by administration of Compound B 2 for 1 day followed by administration of both Compound A 2 and Compound B 2 for 5 consecutive days, followed by administration of Compound A 2 for 1 day, followed by administration of Compound B 2 for 5 consecutive days.
  • Compound B is subsequently administered for one or more consecutive days. Also, contemplated herein is a drug holiday utilized between the sequential administration of
  • a drug holiday is a period of one or more days after the
  • the drug holiday can be a various number of days. In some embodiments, the drug holiday is 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13 or 14 days.
  • one of Compound A and Compound B is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 consecutive days, followed by an optional drug holiday of 1 , 2, 3, 4,
  • 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.
  • treatment protocols and regimens described herein can comprise the entire treatment protocol for a given patient or, alternatively, can comprise only a portion of the entire treatment protocol for the patient.
  • the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from a lower limit of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295 or 300 mg to an upper limit of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215,
  • Compound A is administered from 1 , 2, 3, 4, 5, or 6 times a day.
  • the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from a lower limit of about 0.05, 0.10, 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1.0, 1.05, 1.10, 1 .15, 1 .20, 1 .25, 1.30, 1.35, 1 .40, 1 .45, 1.50, 1.55, 1.60, 1 .65, 1 .70, 1 .75, 1 .80, 1.85, 1.90, 1.95 or 2.0 mg to an upper limit of about 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1 .0, 1.05, 1 .10, 1 .15, 1.20, 1.25, 1 .30, 0.35,
  • the selected amount of Compound B is administered 1 , 2, 3, 4, 5 or 6 times a day.
  • the method of the present invention may also be employed with other therapeutic methods of breast cancer treatment.
  • 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,
  • compositions which include Compound A and/or Compound B , and one or more pharmaceutically acceptable carriers for the treatment of breast cancer.
  • 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 for the treatment of breast cancer 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 precise nature of the breast 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 and Compound B may be compounded together in a pharmaceutical
  • Compound A and Compound B are administered in separate pharmaceutical compositions. In other embodiments,
  • Compound A and Compound B are administered in fixed-dose pharmaceutical compositions that include both Compound A 2 and Compound B 2 .
  • 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 0.05 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.
  • Compound A in combination with Compound B are administered to a human.
  • the human is a female.
  • 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.
  • This invention also provides for a combination comprising 5-[[4-[(2,3-dimethyl- 2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfona or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- i)]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, for use in the treatment of breast cancer.
  • This invention also provides for the use of a combination comprising 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9- dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, in the manufacture of a medicament for the treatment of breast cancer.
  • This invention also provides a method of treating breast cancer which comprises administering a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)-10-[(dimethylamino)methyl]-4- ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H,12H)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to a subject in need thereof.
  • Topotecan hydrochloride (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy- 1 /-/-yrano[3',4':6,7]indolizino[1 ,2-6]quinoline-3, 14(4/-/, 12/-/)-dione monohydrochloride and pazopanib monohydrochloride, (5-[[4-[(2,3-Dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzolsulfonamide are available from GlaxoSmithkline. Topotecan and pazopanib were obtained from GlaxoSmithKline. Topotecan used for MTD schedule was manufactured by Sandoz Canada Inc, QC, Canada, and purchased from the institutional pharmacy. All drugs were prepared according to manufacturers' instructions.
  • the original cell line, MDA-MB 231 was a gift from Dr. Jeffrey F. Lemontt in Sept, 1989, together with other variants of this cell line.
  • Cell line 231/LM2-4 human breast cancer cells used in the study was derived by one of the inventors as described in Munoz R, Man S, Shaked Y, et al. Highly efficacious non-toxic treatment for advanced metastatic breast cancer using combination UFT-cyclophosphamide metronomic chemotherapy, Cancer Res 2006; 66:3386-91 .
  • mice Two million 231/LM2-4 human breast cancer cells were implanted into the mammary fat pad of 6 -8 week old female CB-17 SCID mice purchased from Charles River Canada. Twenty days later when tumor volumes were approximately 400mm 3 , the primary tumors were surgically resected. Therapy was initiated 19 days after resection when visceral metastases were established in sites such as lung and liver. Mice were randomized into groups of 4 and treated as follows: 1 ) Control - vehicle; 2) Low dose metronomic oral topotecan; 3) pazopanib; 4) Low dose metronomic oral topotecan + pazopanib; 5) Maximum tolerated dose topotecan; and 6) maximum tolerated dose topotecan + pazopanib.
  • the schedule and doses of drugs used were metronomic oral topotecan 1 mg/kg/d by gavage, maximum tolerated dose topotecan 1 .5mg/kg by gavage for 5 consecutive days followed by 16 days break ip, pazopanib 150mg/kg/d by gavage, and control mice were given the topotecan vehicle and/or pazopanib vehicle.
  • the oral topotecan and pazopanib preparations were mixed just prior to administration to avoid possible drug interaction. Institutional guidelines for survival endpoint were followed. Results:

Landscapes

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

Abstract

The present invention relates to a method of treating breast cancer in a human and to pharmaceutical combinations useful in such treatment. In particular, the method relates to a breast cancer treatment method that includes administering 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1H-pyrano[3',4':6,7] indolizino [1,2-b]quinoline-3,14-(4H,12H)-dione, 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 a VEGFR inhibitor and a topoisomerase inhibitor, 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).
The process of angiogenesis is the development of new blood vessels from the pre-existing vasculature. Angiogenesis is defined herein as involving: (i) activation of endothelial cells; (ii) increased vascular permeability; (iii) subsequent dissolution of the basement membrane and extravasation of plasma components leading to formation of a provisional fibrin gel extracellular matrix; (iv) proliferation and mobilization of endothelial cells; (v) reorganization of mobilized endothelial cells to form functional capillaries; (vi) capillary loop formation; and (vi) deposition of basement membrane and recruitment of perivascular cells to newly formed vessels. Normal angiogenesis is active during tissue growth from embryonic development through maturity and then enters a period of relative quiescence during adulthood. Normal angiogenesis is also activated during wound healing, and at certain stages of the female reproductive cycle. Inappropriate or pathological angiogenesis has been associated with several disease states including various retinopathies, ischemic disease, atherosclerosis, chronic inflammatory disorders, and cancer. The role of angiogenesis in disease states is discussed, for instance, in Fan et al., Trends in Pharmacol Sci. 16:54-66; Shawver et al., DDT Vol. 2, No.2 February 1997; Folkmann, 1995, Nature Medicine 1 :27-31.
In cancer the growth of solid tumors has been shown to be dependent on angiogenesis. The progression of leukemias as well as the accumulation of fluid associated with malignant ascites and pleural effusions also involve pro-angiogenic factors. (See Folkmann, J., J. Nat'l. Cancer Inst, 1990, 82, 4-6).
Central to the process of angiogenesis are vascular endothelial growth factor (VEGF) and its receptors, termed vascular endothelial growth factor receptor(s) (VEGFRs). The roles VEGF and VEGFRs play in the vascularization of solid tumors, progression of hematopoietic cancers and modulation of vascular permeability have drawn great interest in the scientific community. VEGF is a polypeptide, which has been linked to inappropriate or pathological angiogenesis (Pinedo, H. M. et al. The Oncologist, Vol.5, No. 90001 , 1 -2, Apr. 2000). VEGFR(s) are protein tyrosine kinases (PTKs) that catalyze the phosphorylation of specific tyrosine residues in proteins that are involved in the regulation of cell growth, differentiation, and survival. (A. F. Wilks, Progress in Growth Factor Research, 1990, 2, 97-1 1 1 ; S. A. Courtneidge, Dev. Supp.1 , 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 ).
Three PTK receptors for VEGF have been identified: VEGFRI (Flt-I); VEGFR2 (Flk-I and KDR) and VEGFR3 (Flt-4). These receptors are involved in angiogenesis and participate in signal transduction. (Mustonen, T. et al. J. Cell. Biol. 1995: 129:895-898; Ferrara and Davis-Smyth, Endocrine Reviews, 18(1 ):4-25, 1997; McMahon, G., The Oncologist, Vol. 5, No 90001 , 3-10, Apr. 2000).
Of particular interest is VEGFR2, which is a transmembrane receptor PTK expressed primarily in endothelial cells. Activation of VEGFR-2 by VEGF is a critical step in the signal transduction pathway that initiates tumor angiogenesis. VEGF expression may be constitutive to tumor cells and can also be upregulated in response to certain stimuli. One such stimulus is hypoxia, where VEGF expression is upregulated in both tumor and associated host tissues. The VEGF ligand activates VEGFR2 by binding to its extracellular VEGF binding site. This leads to receptor dimerization of VEGFRs and autophosphorylation of tyrosine residues at the intracellular kinase domain of VEGFR2. The kinase domain operates to transfer a phosphate from ATP to the tyrosine residues, thus providing binding sites for signaling proteins downstream of VEGFR-2 leading ultimately to angiogenesis. (Ferrara and Davis-Smyth, Endocrine Reviews, 18(1 ):4-25, 1997; McMahon, G. The Oncologist, Vol. 5, No.9000l, 3-10, Apr. 2000.)
Consequently, antagonism of the VEGFR2 kinase domain would block phosphorylation of tyrosine residues and serve to disrupt initiation of angiogenesis. Specifically, inhibition at the ATP binding site of the VEGFR2 kinase domain would prevent binding of ATP and prevent phosphorylation of tyrosine residues. Such disruption of the proangiogenesis signal transduction pathway associated with VEGFR2 should therefore inhibit tumor angiogenesis and thereby provide a potent treatment for cancer or other disorders associated with inappropriate angiogenesis. Votrient (pazopanib hydrochloride) is a multi-tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR)-1 , VEGFR-2, VEGFR-3, platelet-derived growth factor receptor (PDGFR)-a and -β, fibroblast growth factor receptor (FGFR) -1 and -3, cytokine receptor (Kit), interleukin-2 receptor inducible T-cell kinase (Itk), leukocyte-specific protein tyrosine kinase (Lck), and transmembrane glycoprotein receptor tyrosine kinase (c-Fms) and is approved in the US for the treatment of patients with advanced renal cell carcinoma. The chemical name of pazopanib hydrochloride is 5-[[4-[(2,3-dimethyl-2H- indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide
monohydrochloride.
The structure of the DNA helix within eukaryotic cells imposes certain topological problems that the cellular apparatus must solve in order to use its genetic material as a template. The separation of the DNA strands is fundamental to cellular processes such as DNA replication and transcription. Since eukaryotic DNA is organized into chromatin by chromosomal proteins, the ends are constrained and the strands cannot unwind without the aid of enzymes that alter topology. It has long been recognized that the advancement of the transcription or replication complex along the DNA helix would be facilitated by a swivel point which would relieve the torsional strain generated during these processes. Topoisomerases are enzymes that are capable of altering DNA topology in eukaryotic cells. They are critical for important cellular functions and cell proliferation.
There are two classes of topoisomerases in eukaryotic cells, type I and type II. Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single strand break, unwinds the double helix (or allows it to unwind), and subsequently reseals the break before dissociating from the DNA strand. Topoisomerase II consists of two identical subunits of molecular weight 170,000. Topoisomerase II transiently breaks both strands of the helix and passes another double-strand segment through the break. Camptothecin is a water- insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to
India. Camptothecin and a few close congeners thereof are the only class of compounds known to inhibit topoisomerase I. Inhibition of topoisomerase II is the major target of important commercial oncolytic agents (e.g., etoposide, doxorubicin and mitoxantrone) as well as other oncolytic agents still undergoing development. Camptothecin (and its known congeners) have no effect on topoisomerase II and none of the known topoisomerase II inhibitors has any significant effect on topoisomerase I. Hycamtin® (topotecan hydrochloride) is a semi-synthetic derivative of campotothecin that exhibits topoisomerase l-inhibitory activity. Hycamtin® is approved in the US for the treatment of relapsed small cell lung cancer, ovarian cancer and cervical cancer. The chemical name for topotecan hydrochloride is (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 /-/- pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H,12/-/)-dione monohydrochloride.
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 that includes:
(i) a compound of Structure (I):
Figure imgf000006_0001
or a pharmaceutically acceptable 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 breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonam or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to such human.
One embodiment of this invention provides a method of treating breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride 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 breast cancer in a human in need thereof which comprises the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- 6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, to such human, wherein the compounds of the combination are administered sequentially.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 illustrates percent survival after an indicated number of days when treatment of Advanced Metastatic Breast Cancer (MDA-MB-231/LM2-4) was initiated at day 39, 19 days post primary tumor resection, for the control group, the low dose metronomic oral topotecan alone group, the maximum tolerated group, the pazopanib alone group, the low dose oral topotecan + pazopanib group, and the maximum tolerated dose topotecan + pazopanib group.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to combinations that exhibit antitumor activity.
Suitably, the method relates to methods of treating breast cancer by the co- administration of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof, (hereinafter Compound A, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof), which compound is represented by Structure I:
Figure imgf000008_0001
and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4/-/, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, (hereinafter Compound B or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof), which compound is represented by Structure II:
Figure imgf000008_0002
Compound A is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of VEGFR activity, particularly in treatment of cancer, in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001 , International Publication Number WO02/0591 10 and an International Publication date of August 1 , 2002, the entire disclosure of which is hereby incorporated by reference, Compound A is the compound of Example 69. Compound A can be prepared as described in International Application No. PCT/US01/49367. Suitably, Compound A is in the form of a monohydrochloride salt. This salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001.
Compound A is sold commercially as the monohydrochloride salt. Compound A
®
is known by the generic name pazopanib and the trade name Votrient .
Compound B is disclosed and claimed, along with pharmaceutically acceptable salts thereof, as being useful as an inhibitor of topoisomerase I, particularly in treatment of cancer, in US Patent No. 5,004,758, having a filing date of November 2, 1988, the entire disclosure of which is hereby incorporated by reference, Compound B is compound 1 S (as the acetate salt). Compound B can be prepared as described in US Patent No. 5,734,056.
Suitably, 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 US Patent No. 5,004,758 and/or by methods that will be readily apparent to those skilled in the art.
Compound B is sold commercially as the monohydrochloride salt. Compound B
®
is known by the generic name topotecan and the trade name Hycamtin .
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 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, 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 breast 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 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 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 breast cancer, such as when a subject has a strong family history of breast 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 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.
2
As used herein the term "Compound A " means — Compound A, or a pharmaceutically acceptable salt thereof— .
2
As used herein the term "Compound B " means — Compound B, or a pharmaceutically acceptable salt thereof— .
In some embodiments according to the present invention, the combinations of this invention are administered within a "specified period".
By the term "specified period" and derivatives thereof, as used herein is meant
2
the interval of time between the administration of one of Compound A and Compound
2 2 2
B 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.
The specified period can be various time periods. For example, Compound A2 and Compound B2 can be administered within about 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 hours of each other, in which case the specified period will be about 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9,
8, 7, 6, 5, 4, 3, 2 or 1 hours, respectively. 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.
The duration of time can be various time periods. For example, Compound A2 and Compound B2 can both be administered within a specified period for at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 consecutive days during the course of treatment, in which case the duration of time will be 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30, respectively. When, during the course of treatment, both compounds are administered within a specified period for over 30 consecutive days, the treatment is considered chronic treatment and will continue until an altering event, such as a reassessment in breast cancer status or a change in the condition of the patient, warrants a modification to the protocol.
Various treatment protocols are contemplated in embodiments of the present invention. For example, Compound A2 and B2 can be co-administered within a specified period for at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 ,
22, 23, 24, 25, 26, 27, 28, 29 or 30 days, followed by the administration of Compound A2 alone for at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22,
23, 24, 25, 26, 27, 28, 29 or 30 days, in which case the duration of time will be at least the number of consecutive days that Compound A2 and Compound B2 are both administered plus the number of consecutive days of administration of Compound A2 alone (e.g., if Compound A2 and Compound B2 are both administered for 6 consecutive days followed by administration of Compound A2 alone for 8 consecutive days, the duration of time will be at least 14 consecutive days).
In other embodiments, Compound A2 and Compound B2 are both administered within a specified period for a number of consecutive days during a certain time period, and compound A2 is administered during the other days of the certain time period. In some embodiments, the certain time period is n = 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 days, the days of consecutive administration of Compound A2 and Compound B2 within a specified time period is m = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28 or 29, and the days of administration of Compound A2 is n - m, where n - m is at least 1. For example, Compound A2 and Compound B2 can be administered within a specified time period for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 or 13 consecutive days over a certain time period of 14 days, during which Compound A2 is administered for the other 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 days, respectively. In this example, n = 14, m = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 or 13, and n - m = 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 , respectively. The consecutive days during which Compound A2 and Compound B2 are both administered within a specified time period can occur any time during the certain time period. Accordingly, in the foregoing example, Compound A2 could be administered alone for 4 consecutive days follow by administration of both Compound A2 and Compound B2 for 5 consecutive days, followed by administering Compound A2 alone for 5 consecutive days to complete the 14 day certain time period.
While treatment protocols have been described with respect to administration of both Compound A2 and Compound B2 within a specified period in conjunction with administration of Compound A2 alone, embodiments of the present invention also include similar treatment protocols in which Compound A2 and Compound B2 are both administered within a specified period in conjunction with administration of Compound B2 alone.
Other embodiments of the present invention include administration of both Compound A2 and Compound B2 within a specified period in conjunction with administration of Compound A2 alone and administration of Compound B2 alone. For example, in some embodiments Compound A2 and Compound B2 are both administered within a specified period for a number of consecutive days during a certain time period, Compound A2 is administered alone during a number of days during the certain time period, and Compound B2 is administered alone during the other days during the certain time period. In some embodiments, the certain time period is n = 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 days, the days of consecutive administration of Compound A2 and Compound B2 within a specified time period is m = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27 or 28, the days of administration of Compound A2 during the certain time period is p = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27 or 28, and the days of administration of Compound B2 is n - m - p, where n - m - p is at least 1. For example, Compound A2 and Compound B2 can both be administered within a specified time period for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 consecutive days over a certain time period of 14 days, during which Compound A2 is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 days, and Compound B2 is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 days. In this example, n = 14, m = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12, p = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12, and n - m - p = 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12. The consecutive days during which Compound A2 and Compound B2 are both administered within a specified time period can occur any time during the certain time period. Accordingly, in the foregoing example, Compound A2 could be administered alone for 4 consecutive days follow by administration of both Compound A2 and Compound B2 for 5 consecutive days, followed by administering Compound B2 alone for 5 consecutive days to complete the 14 day certain time period. Administration of Compound A2 alone and administration of Compound B2 alone do not have to occur on consecutive days. Accordingly, in the foregoing example, Compound A2 could be administered for 2 consecutive days, followed by administration of Compound B2 for 1 day followed by administration of both Compound A2 and Compound B2 for 5 consecutive days, followed by administration of Compound A2 for 1 day, followed by administration of Compound B2 for 5 consecutive days.
If the compounds are not administered during a "specified period", they are administered sequentially. By the term "sequential administration", and derivatives
2 2 thereof, as used herein is meant that one of Compound A and Compound B is
2 administered for one or more consecutive days and the other of Compound A and
2
Compound B is subsequently administered for one or more consecutive days. Also, contemplated herein is a drug holiday utilized between the sequential administration of
2 2 2
one of Compound A and Compound B and the other of Compound A and Compound
2
B . As used herein, a drug holiday is a period of one or more days after the
2 2
administration of one of Compound A and Compound B and before the sequential
2 2
administration of the other of Compound A and Compound B where neither Compound
2 2
A nor Compound B is administered. The drug holiday can be a various number of days. In some embodiments, the drug holiday is 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13 or 14 days.
2 2
In some embodiments, one of Compound A and Compound B is administered for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 consecutive days, followed by an optional drug holiday of 1 , 2, 3, 4,
5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29
2 2 or 30 days, followed by administration of the other of Compound A and Compound B for 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 consecutive days.
It is understood that 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.
It is to be understood that the treatment protocols and regimens described herein can comprise the entire treatment protocol for a given patient or, alternatively, can comprise only a portion of the entire treatment protocol for the patient.
2
Suitably, the amount of Compound A administered as part of the combination according to the present invention will be an amount selected from a lower limit of about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295 or 300 mg to an upper limit of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 1 10, 1 15, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 305, 310, 315, 320, 325, 330, 335, 340, 345, 350, 355, 360, 365, 370, 375, 380, 385, 390, 395, 400, 405, 410, 415, 420, 425, 430, 435, 440, 445, 450, 455, 460, 465, 470, 475, 480, 485, 490, 495, 500, 505, 510, 515, 520, 525, 530, 535, 540, 545, 550, 555, 560, 565, 570, 575, 580, 585, 590, 595, 600, 605, 610, 615, 620, 625, 630, 635, 640, 645, 650, 655, 660, 665, 670, 675, 680, 685, 690,
695, 700, 705, 710, 715, 720, 725, 730, 735, 740, 745, 750, 755, 760, 765, 770, 775,
780, 785, 790, 795, or 800 mg. It is to be understood that embodiments of the present invention include any number in the ranges listed above. In some embodiments, the
2
selected amount of Compound A is administered from 1 , 2, 3, 4, 5, or 6 times a day.
2
Suitably, the amount of Compound B administered as part of the combination according to the present invention will be an amount selected from a lower limit of about 0.05, 0.10, 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1.0, 1.05, 1.10, 1 .15, 1 .20, 1 .25, 1.30, 1.35, 1 .40, 1 .45, 1.50, 1.55, 1.60, 1 .65, 1 .70, 1 .75, 1 .80, 1.85, 1.90, 1.95 or 2.0 mg to an upper limit of about 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1 .0, 1.05, 1 .10, 1 .15, 1.20, 1.25, 1 .30, 1 .35, 1 .40, 1.45, 1.50, 1 .55, 1 .60, 1.65, 1.70, 1.75, 1 .80, 1 .85, 1.90, 1.95, 2.0, 2.05, 2.10, 2.15, 2.20, 2.25, 2.30, 2.35, 2.40, 2.45, 2.50, 2.55, 2.60, 2.65, 2.70, 2.75, 2.80, 2.85, 2.90, 2.95, 3.0, 3.05, 3.10, 3.15, 3.20, 3.25, 3.30,
3.35, 3.40, 3.45, 3.50, 3.55, 3.60, 3.65, 3.70, 3.75, 3.80, 3.85, 3.90, 3.95 or 4.0 mg. In
2
some embodiments, the selected amount of Compound B is administered 1 , 2, 3, 4, 5 or 6 times 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 compound per dose.
The method of the present invention may also be employed with other therapeutic methods of breast 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,
2 2
which include Compound A and/or Compound B , and one or more pharmaceutically acceptable carriers for the treatment of breast cancer. 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 for the treatment of breast cancer 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 precise nature of the breast 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
2 2
A and Compound B may be compounded together in a pharmaceutical
2 2 composition/formulation. In some embodiments, Compound A and Compound B are administered in separate pharmaceutical compositions. In other embodiments,
2 2
Compound A and Compound B are administered in fixed-dose pharmaceutical compositions that include both Compound A2 and Compound B2.
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 0.05 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. In some embodiments, the human is a female. 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.
This invention also provides for a combination comprising 5-[[4-[(2,3-dimethyl- 2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfona or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- i)]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, for use in the treatment of breast cancer.
This invention also provides for the use of a combination comprising 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9- dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride salt, thereof, in the manufacture of a medicament for the treatment of breast cancer.
This invention also provides a method of treating breast cancer which comprises administering a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt, thereof and (S)-10-[(dimethylamino)methyl]-4- ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H,12H)-dione, or a pharmaceutically acceptable salt, suitably the hydrochloride 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
Materials and Methods:
Drugs and reagents:
Topotecan hydrochloride, (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy- 1 /-/-yrano[3',4':6,7]indolizino[1 ,2-6]quinoline-3, 14(4/-/, 12/-/)-dione monohydrochloride and pazopanib monohydrochloride, (5-[[4-[(2,3-Dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzolsulfonamide are available from GlaxoSmithkline. Topotecan and pazopanib were obtained from GlaxoSmithKline. Topotecan used for MTD schedule was manufactured by Sandoz Canada Inc, QC, Canada, and purchased from the institutional pharmacy. All drugs were prepared according to manufacturers' instructions.
Cell lines:
The original cell line, MDA-MB 231 , was a gift from Dr. Jeffrey F. Lemontt in Sept, 1989, together with other variants of this cell line. Cell line 231/LM2-4 (human breast cancer cells) used in the study was derived by one of the inventors as described in Munoz R, Man S, Shaked Y, et al. Highly efficacious non-toxic treatment for advanced metastatic breast cancer using combination UFT-cyclophosphamide metronomic chemotherapy, Cancer Res 2006; 66:3386-91 .
Metastatic model:
Two million 231/LM2-4 human breast cancer cells were implanted into the mammary fat pad of 6 -8 week old female CB-17 SCID mice purchased from Charles River Canada. Twenty days later when tumor volumes were approximately 400mm3, the primary tumors were surgically resected. Therapy was initiated 19 days after resection when visceral metastases were established in sites such as lung and liver. Mice were randomized into groups of 4 and treated as follows: 1 ) Control - vehicle; 2) Low dose metronomic oral topotecan; 3) pazopanib; 4) Low dose metronomic oral topotecan + pazopanib; 5) Maximum tolerated dose topotecan; and 6) maximum tolerated dose topotecan + pazopanib. The schedule and doses of drugs used were metronomic oral topotecan 1 mg/kg/d by gavage, maximum tolerated dose topotecan 1 .5mg/kg by gavage for 5 consecutive days followed by 16 days break ip, pazopanib 150mg/kg/d by gavage, and control mice were given the topotecan vehicle and/or pazopanib vehicle. The oral topotecan and pazopanib preparations were mixed just prior to administration to avoid possible drug interaction. Institutional guidelines for survival endpoint were followed. Results:
As shown in Figure 1 , neither low dose metronomic (LDM) oral topotecan alone nor pazopanib alone had any survival benefit when compared to the control untreated mice. One mouse in the LDM oral topotecan + pazopanib treated group was euthanized early in the experiment due to bowel obstruction and this was taken out of the results. Median survival for control was 66 days, for LDM oral topotecan alone was 56 days, for pazopanib alone was 66 days, for maximum tolerated dose (MTD) topotecan was 63 days and for MTD topotecan + pazopanib was 80 days, whereas more than 50% of mice treated with LDM oral topotecan in combination with pazopanib were still alive at 150 days.
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

CLAIMS We claim:
1. A combination comprising: (i) a compound of Structure (I):
Figure imgf000021_0001
or a pharmaceutically acceptable salt thereof; and
(ii) a compound of Structure (II):
Figure imgf000021_0002
or a pharmaceutically acceptable salt thereof.
2. A combination according to claim 1 where the compounds of Structure (I) and Structure (II) are each in the form of a monohydrochloride salt.
3. A combination according to claim 1 or 2 where the amount of the compound of Structure (I) is an amount from 5 mg to 800 mg, 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 from 0.05 mg to 1 mg, and that amount is administered once per day.
4. Use of a combination according to any of claims 1 to 3 in the manufacture of a medicament or medicaments for the treatment of breast cancer.
5. A method of treating breast cancer in a human in need thereof, comprising the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 /-/- pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12/-/)-dione, 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.
6. A method according to claim 5 wherein the amount of 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, is from about 5 mg to about 800 mg, and that amount is administered once per day.
7. A method according to claim 5 or 6, wherein the amount of (S)-10- [(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- i)]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt thereof, is from about 0.05 mg to about 1 mg, and that amount is administered once per day.
8. A method according to any one of claims 5 to 7, wherein the specified period is within about 1 to about 12 hours.
9. A method according to any one of claims 5 to 8, wherein the duration of time is for from 1 to 30 consecutive days.
10. A method according to any one of claims 5 to 9, wherein the human is female.
1 1 . A method according to any one of claims 5 to 10, wherein 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9- dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3,14-(4H, 12/-/)-dione are each in the form of a monohydrochloride salt.
12. A method treating breast cancer in a human in need thereof, comprising the in vivo administration of a therapeutically effective amount of a combination of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2- pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H- pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt thereof, to such human, wherein the compounds of the combination are administered sequentially.
13. A method according to claim 12, wherein the amount of 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, is from about 5 mg to about 800 mg, and that amount is administered once per day.
14. A method according to claim 12 or 13, wherein the amount of (S)-10- [(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- i)]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt thereof, is from about 0.05 mg to about 1 mg, and that amount is administered once per day.
15. A method according to any one of claims 12 to 14, wherein 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, 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 (S)-10-[(dimethylamino)methyl]-4- ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12H)- dione, or a pharmaceutically acceptable salt thereof for from 1 to 30 days.
16. A method according to any one of claims 12 to 15, wherein the human is female.
17. A method according to any one of claims 12 to 16, wherein 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9- dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12/-/)-dione are each in the form of a monohydrochloride salt.
18. A combination comprising 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, and (S)-10-[(dimethylamino)methyl]-4-ethyl- 4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12H)-dione, or a pharmaceutically acceptable salt thereof for use in the treatment of breast cancer.
19. A combination according to claim 18, wherein the amount of 5-[[4- [(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, is from about 5 mg to about 800 mg.
20. A combination according to claim 18 or 19, wherein the amount of (S)- 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2- i)]quinoline-3, 14-(4H, 12/-/)-dione, or a pharmaceutically acceptable salt thereof, is from about 0.05 mg to about 1 mg.
21 . A combination according to any one of claims 18 to 20, wherein 5-[[4- [(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide and (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9- dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3,14-(4H, 12/-/)-dione are each in the form of a monohydrochloride salt.
22. The use of a combination comprising 5-[[4-[(2,3-dimethyl-2H-indazol- 6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt thereof, and (S)-10-[(dimethylamino)methyl]-4-ethyl- 4,9-dihydroxy-1 H-pyrano[3',4':6,7] indolizino [1 ,2-6]quinoline-3, 14-(4H, 12H)-dione, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of breast cancer.
PCT/US2012/023261 2011-02-01 2012-01-31 Combination WO2012106302A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US13/980,904 US20130303560A1 (en) 2011-02-01 2012-01-31 Combination
EP12742225.1A EP2672969A4 (en) 2011-02-01 2012-01-31 Combination
JP2013552574A JP2014504638A (en) 2011-02-01 2012-01-31 combination
US15/291,463 US20170027938A1 (en) 2011-02-01 2016-10-12 Combination

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161438426P 2011-02-01 2011-02-01
US61/438,426 2011-02-01

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/980,904 A-371-Of-International US20130303560A1 (en) 2011-02-01 2012-01-31 Combination
US15/291,463 Continuation US20170027938A1 (en) 2011-02-01 2016-10-12 Combination

Publications (1)

Publication Number Publication Date
WO2012106302A1 true WO2012106302A1 (en) 2012-08-09

Family

ID=46603053

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/023261 WO2012106302A1 (en) 2011-02-01 2012-01-31 Combination

Country Status (4)

Country Link
US (2) US20130303560A1 (en)
EP (1) EP2672969A4 (en)
JP (1) JP2014504638A (en)
WO (1) WO2012106302A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8709420B2 (en) 2010-12-17 2014-04-29 Glaxosmithkline Intellectual Property Limited Pharmaceutical combination of pazopanib and topotecan to treat neuroblastoma, osteosarcoma, and rhabdomyosarcoma in a human

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5004758A (en) 1987-12-01 1991-04-02 Smithkline Beecham Corporation Water soluble camptothecin analogs useful for inhibiting the growth of animal tumor cells
US5734056A (en) 1990-09-28 1998-03-31 Smithkline Beecham Corporation Process for the preparation of certain 9-substituted camptothecins
WO2002059110A1 (en) 2000-12-21 2002-08-01 Glaxo Group Limited Pyrimidineamines as angiogenesis modulators
US20060216288A1 (en) * 2005-03-22 2006-09-28 Amgen Inc Combinations for the treatment of cancer
US20100249127A1 (en) * 2009-03-31 2010-09-30 Arqule, Inc. Substituted indolo-pyridinone compounds

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6833373B1 (en) * 1998-12-23 2004-12-21 G.D. Searle & Co. Method of using an integrin antagonist and one or more antineoplastic agents as a combination therapy in the treatment of neoplasia
EP1301334A1 (en) * 2000-06-16 2003-04-16 Avery Dennison Corporation A process and apparatus for precise embossing
MX2007014049A (en) * 2005-05-12 2008-02-11 Abbott Lab Apoptosis promoters.
WO2007143483A2 (en) * 2006-06-01 2007-12-13 Smithkline Beecham Corporation Combination of pazopanib and lapatinib for treating cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5004758A (en) 1987-12-01 1991-04-02 Smithkline Beecham Corporation Water soluble camptothecin analogs useful for inhibiting the growth of animal tumor cells
US5734056A (en) 1990-09-28 1998-03-31 Smithkline Beecham Corporation Process for the preparation of certain 9-substituted camptothecins
WO2002059110A1 (en) 2000-12-21 2002-08-01 Glaxo Group Limited Pyrimidineamines as angiogenesis modulators
US20060216288A1 (en) * 2005-03-22 2006-09-28 Amgen Inc Combinations for the treatment of cancer
US20100249127A1 (en) * 2009-03-31 2010-09-30 Arqule, Inc. Substituted indolo-pyridinone compounds

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
A. C. CHAN, CURR. OPIN. IMMUNOL., vol. 8, no. 3, 1996, pages 394 - 401
A. F. WILKS, PROGRESS IN GROWTH FACTOR RESEARCH, vol. 2, 1990, pages 97 - 111
CREWS; ERIKSON, CELL, vol. 74, 1993, pages 215 - 17
FAN ET AL., TRENDS IN PHARMACOL SCI., vol. 16, pages 54 - 66
FERRARA; DAVIS-SMYTH, ENDOCRINE REVIEWS, vol. 18, no. 1, 1997, pages 4 - 25
FOLKMANN, J., J. NAT'L. CANCER INST, vol. 82, 1990, pages 4 - 6
FOLKMANN, NATURE MEDICINE, vol. 1, 1995, pages 27 - 31
J. A. COOPER, SEMIN. CELL BIOL., vol. 5, no. 6, 1994, pages 377 - 387
J. R. MOLINA ET AL., CLINICAL CANCER RESEARCH, vol. 14, no. 23, 1 December 2008 (2008-12-01), pages 7900 - 7908
K. HASHIMOTO ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 9, no. 4, 1 April 2010 (2010-04-01), pages 996 - 1006
MCMAHON, G., THE ONCOLOGIST, vol. 5, no. 90001, April 2000 (2000-04-01), pages 3 - 10
MUNOZ R; MAN S; SHAKED Y ET AL.: "Highly efficacious non-toxic treatment for advanced metastatic breast cancer using combination UFT-cyclophosphamide metronomic chemotherapy", CANCER RES, vol. 66, 2006, pages 3386 - 91
MUSTONEN, T. ET AL., J. CELL. BIOL., vol. 129, 1995, pages 895 - 898
PINEDO, H. M. ET AL., THE ONCOLOGIST, vol. 5, no. 90001, April 2000 (2000-04-01), pages 1 - 2
R. F. PAULSON, SEMIN. IMMUNOL., vol. 7, no. 4, 1995, pages 267 - 277
S. A. COURTNEIDGE, DEV. SUPP., vol. 1, 1993, pages 57 - 64
See also references of EP2672969A4 *
SHAWVER ET AL., DDT, vol. 2, no. 2, February 1997 (1997-02-01)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8709420B2 (en) 2010-12-17 2014-04-29 Glaxosmithkline Intellectual Property Limited Pharmaceutical combination of pazopanib and topotecan to treat neuroblastoma, osteosarcoma, and rhabdomyosarcoma in a human

Also Published As

Publication number Publication date
JP2014504638A (en) 2014-02-24
US20130303560A1 (en) 2013-11-14
EP2672969A4 (en) 2014-07-16
EP2672969A1 (en) 2013-12-18
US20170027938A1 (en) 2017-02-02

Similar Documents

Publication Publication Date Title
JP6508544B2 (en) Pharmaceutical composition for treating a tumor
TW201138764A (en) Anticancer combinations of artemisinin-based drugs and other chemotherapeutic agents
JP2019513812A (en) Chemotherapy improvement
US20160067248A1 (en) Combinations
US20170239255A1 (en) Combination
JP6373252B2 (en) Methods of treating cancer using aurora kinase inhibitors
US20170027938A1 (en) Combination
JP2006504721A (en) Use of Imatinib (Gleevec, STI-571) to inhibit breast cancer resistance protein (BCRP) -mediated resistance to therapeutic agents
TWI430801B (en) Use of berberine-containing compound for preparing drug for inhibiting cancer stem cells growth or metastasis
WO2021018310A1 (en) Aminopyridine derivatives for treatment of non-small cell lung cancer
CA2888094A1 (en) Combinations
KR20180125456A (en) Methods involving fixed intermittent dosing of cediranib
US8709420B2 (en) Pharmaceutical combination of pazopanib and topotecan to treat neuroblastoma, osteosarcoma, and rhabdomyosarcoma in a human
KR20180112066A (en) A pharmaceutical composition comprising a pyrrolo-fused 6-membered heterocyclic compound
CN111700893A (en) Use of pyrimidine derivatives for cancer therapy
JP2013526613A (en) combination
JP2023520383A (en) Pharmaceutical composition containing protein kinase inhibitor and chemotherapeutic agent and use thereof
JP2014034534A (en) COMBINATION OF HSP90 INHIBITOR AND mTOR INHIBITOR
JP2015534987A (en) combination
JP2014034535A (en) Combination of hsp90 inhibitor, and vegfr tyrosine kinase and pdgf tyrosine kinase inhibitor

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: 12742225

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 13980904

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2013552574

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2012742225

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012742225

Country of ref document: EP