US20140057943A1 - Method of Treating Cancer - Google Patents

Method of Treating Cancer Download PDF

Info

Publication number
US20140057943A1
US20140057943A1 US13/876,290 US201113876290A US2014057943A1 US 20140057943 A1 US20140057943 A1 US 20140057943A1 US 201113876290 A US201113876290 A US 201113876290A US 2014057943 A1 US2014057943 A1 US 2014057943A1
Authority
US
United States
Prior art keywords
compound
formula
bone
pharmaceutically acceptable
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/876,290
Other languages
English (en)
Inventor
David C. Smith
Maha Hussain
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Michigan
Exelixis Inc
Original Assignee
Exelixis Inc
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=44741730&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140057943(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Exelixis Inc filed Critical Exelixis Inc
Priority to US13/876,290 priority Critical patent/US20140057943A1/en
Publication of US20140057943A1 publication Critical patent/US20140057943A1/en
Assigned to THE REGENTS OF THE UNIVERSITY OF MICHIGAN reassignment THE REGENTS OF THE UNIVERSITY OF MICHIGAN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUSSAIN, MAHA, SMITH, DAVID
Assigned to EXELIXIS, INC. reassignment EXELIXIS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THE REGENTS OF THE UNIVERSITY OF MICHIGAN
Assigned to REGENTS OF THE UNIVERSITY OF MICHIGAN reassignment REGENTS OF THE UNIVERSITY OF MICHIGAN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUSSAIN, MAHA, SMITH, DAVID
Assigned to EXELIXIS, INC. reassignment EXELIXIS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REGENTS OF THE UNIVERSITY OF MICHIGAN
Assigned to REGENTS OF THE UNIVERSITY OF MICHIGAN reassignment REGENTS OF THE UNIVERSITY OF MICHIGAN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUSSAIN, MAHA, SMITH, DAVID
Assigned to EXELIXIS, INC. reassignment EXELIXIS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REGENTS OF THE UNIVERSITY OF MICHIGAN
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/435Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C55/00Saturated compounds having more than one carboxyl group bound to acyclic carbon atoms
    • C07C55/02Dicarboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4

Definitions

  • This invention is directed to the treatment of cancer, particularly castration-resistant prostate cancer and osteoblastic bone metastases, with a dual inhibitor of MET and VEGF.
  • Castration-Resistant Prostate Cancer is a leading cause of cancer-related death in men.
  • CRPC Castration-Resistant Prostate Cancer
  • improvements in survival are modest, and virtually all patients succumb to this disease within about 2 years.
  • the primary cause of morbidity and mortality in CRPC is metastasis to the bone, which occurs in about 90% of cases.
  • Metastasis to the bone is a complex process that involves interactions between cancer cells and components of the bone microenvironment including osteoblasts, osteoclasts, and endothelial cells.
  • Bone metastases cause local disruption of normal bone remodeling, and lesions generally show a propensity for either osteoblastic (bone-forming) or osteolytic (bone-resorbing) activity.
  • osteoblastic bone-forming
  • osteolytic bone-resorbing
  • prostate cancer bone metastases are often osteoblastic, with abnormal deposition of unstructured bone accompanied by increased skeletal fractures, spinal cord compression, and severe bone pain.
  • the receptor tyrosine kinase MET plays important roles in cell motility, proliferation, and survival, and it has been shown to be a key factor in tumor angiogenesis, invasiveness, and metastasis. Prominent expression of MET has been observed in primary and metastatic prostate carcinomas, with evidence for higher levels of expression in bone metastases compared to lymph node metastases or primary tumors.
  • VEGF Vascular endothelial growth factor
  • endothelial cells are widely accepted as key mediators in the process of tumor angiogenesis.
  • elevated VEGF in either plasma or urine is associated with shorter overall survival.
  • VEGF may also play a role in activating the MET pathway in tumor cells by binding to neuropilin-1, which is frequently unregulated in prostate cancer and appears to activate MET in a co-receptor complex.
  • Agents targeting the VEGF signaling pathway have demonstrated some activity in patients with CRPC.
  • the present invention is directed to a method for treating bone cancer, prostate cancer, or bone cancer associated with prostate cancer.
  • the method comprises administering a therapeutically effective amount of a compound that modulates both MET and VEGF to a patient in need of such treatment.
  • the bone cancer is osteoblastic bone metastases.
  • the prostate cancer is CRPC.
  • the bone cancer is osteoblastic bone metastases associated with CRPC.
  • the present invention is directed to a method for treating osteoblastic bone metastases, CRPC, or osteoblastic bone metastases associated with CRPC, comprising administering a therapeutically effective amount of a compound that dually modulates MET and VEGF to a patient in need of such treatment.
  • the dual acting MET/VEGF inhibitor is a compound of Formula I
  • R 1 is halo
  • R 2 is halo
  • R 3 is (C 1 -C 6 )alkyl
  • R 4 is (C 1 -C 6 )alkyl
  • Q is CH or N.
  • the compound of Formula I is a compound of Formula Ia
  • R 1 is halo
  • R 2 is halo
  • Q is CH or N.
  • the compound of Formula I is compound 1:
  • Compound 1 is known as N-(4- ⁇ [6,7-bis(methyloxy)quinolin-4-yl]oxy ⁇ phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide.
  • the compound of Formula I, Ia, or Compound 1 is administered as a pharmaceutical composition comprising a pharmaceutically acceptable additive, diluent, or excipient.
  • the invention provides a method for treating osteoblastic bone metastases associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for reducing or stabilizing metastatic bone lesions associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I, Ia or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I, Ia or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for reducing bone pain due to metastatic bone lesions associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for treating or minimizing bone pain due to metastatic bone lesions associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for strengthening bones in patients with metastatic bone lesions associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1. Bone strengthening can occur when the disruption in normal bone remodeling due to bone metastases is minimized, for instance by administering a Compound of Formula I as provided herein.
  • the invention provides a method for preventing osteoblastic bone metastases associated with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for preventing bone metastases in patients with prostate cancer who are castration resistant but have not yet advanced to metastatic disease, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the Compound of Formula I is Compound 1.
  • the invention provides a method for extending the overall survival in patients with CRPC, comprising administering a therapeutically effective amount of a pharmaceutical composition comprising Compound of Formula I or the malate salt of Compound of Formula I or another pharmaceutically acceptable salt of Compound of Formula I, to a patient in need of such treatment.
  • the ability of the compound of Formula I to treat, ameliorate, or reduce the severity of bone metastases can be determined both qualitatively and quantitatively using various physiological markers, such as circulating tumor cell (CTC) counts and imaging technologies.
  • CTC circulating tumor cell
  • the imaging technologies include positron emission tomography (PET) or computerized tomography (CT) and magnetic resonance imaging. By using these imaging techniques, it is possible to monitor and quantify the reduction in tumor size and the reduction in the number and size of bone lesions in response to treatment with the compound of Formula I.
  • shrinkage of soft tissue and visceral lesions has been observed result when the compound of Formula I is administered to patients with CRPC.
  • administration of the compound of Formula I leads to increases in hemoglobin concentration in patients CRPC patients with anemia.
  • FIGS. 1A-C show the bone scan ( FIG. 1A ), bone scan response ( FIG. 1B ), and CT scan data ( FIG. 1C ) for Patient 1.
  • FIGS. 2A-C show the bone scan ( FIG. 2A ), bone scan response ( FIG. 2B ), and CT scan data ( FIG. 2C ) for Patient 2.
  • FIGS. 3A-B show the bone scan ( FIG. 3A ), bone scan response ( FIG. 3B ) for Patient 3.
  • a substituent “R” may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed.
  • a substituent “R” may reside on any atom of the fused ring system, assuming replacement of a depicted hydrogen (for example the —NH— in the formula above), implied hydrogen (for example as in the formula above, where the hydrogens are not shown but understood to be present), or expressly defined hydrogen (for example where in the formula above, “Z” equals ⁇ CH—) from one of the ring atoms, so long as a stable structure is formed.
  • the “R” group may reside on either the 5-membered or the 6-membered ring of the fused ring system.
  • a group “R” is depicted as existing on a ring system containing saturated carbons, as for example in the formula:
  • Halogen or “halo” refers to fluorine, chlorine, bromine or iodine.
  • Yield for each of the reactions described herein is expressed as a percentage of the theoretical yield.
  • Patient for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In another embodiment the patient is a mammal, and in another embodiment the patient is human.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 17′′ ed., Mack Publishing Company, Easton, Pa., 1985, which is incorporated herein by reference or S. M. Berge, et al., “Pharmaceutical Salts,” J. Pharm. Sci., 1977; 66:1-19 both of which are incorporated herein by reference.
  • Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, malic acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid
  • “Prodrug” refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. Common examples include, but are not limited to, ester and amide forms of a compound having an active form bearing a carboxylic acid moiety.
  • Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl.
  • Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons).
  • Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
  • “Therapeutically effective amount” is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease.
  • a therapeutically effective amount is intended to include an amount of a compound alone or in combination with other active ingredients effective to modulate c-Met, and/or VEGFR 2 , or effective to treat or prevent cancer.
  • the amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined by one of ordinary skill in the art having regard to their knowledge and to this disclosure.
  • Treating” or “treatment” of a disease, disorder, or syndrome includes (i) preventing the disease, disorder, or syndrome from occurring in a human, i.e. causing the clinical symptoms of the disease, disorder, or syndrome not to develop in an animal that may be exposed to or predisposed to the disease, disorder, or syndrome but does not yet experience or display symptoms of the disease, disorder, or syndrome; (ii) inhibiting the disease, disorder, or syndrome, i.e., arresting its development; and (iii) relieving the disease, disorder, or syndrome, i.e., causing regression of the disease, disorder, or syndrome.
  • adjustments for systemic versus localized delivery, age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experience.
  • R 1 is halo
  • R 2 is halo
  • Q is CH or N.
  • the compound of Formula I is Compound 1:
  • compound 1 is referred to herein as N-(4- ⁇ [6,7-bis(methyloxy)quinolin-4-yl]oxy ⁇ phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide.
  • WO 2005/030140 discloses Compound 1 and describes how it is made (Example 12, 37, 38, and 48) and also discloses the therapeutic activity of this compound to inhibit, regulate and/or modulate the signal transduction of kinases, (Assays, Table 4, entry 289).
  • Example 48 is on paragraph [0353] in WO 2005/030140.
  • the compound of Formula I, Ia, or Compound 1, or a pharmaceutically acceptable salt thereof is administered as a pharmaceutical composition, wherein the pharmaceutical composition additionally comprises a pharmaceutically acceptable carrier, excipient, or diluent.
  • the Compound of Formula I is Compound 1.
  • the compound of Formula I, Formula Ia and Compound 1, as described herein, includes both the recited compounds as well as individual isomers and mixtures of isomers.
  • the compound of Formula I includes the pharmaceutically acceptable salts, hydrates, and/or solvates of the recited compounds and any individual isomers or mixture of isomers thereof.
  • the compound of Formula I, Ia, or Compound 1 can be the (L)-malate salt.
  • the malate salt of the Compound of Formula I and of Compound 1 is disclosed in PCT/US2010/021194 and 61/325,095.
  • the compound of Formula I can be the (D)-malate salt.
  • the compound of Formula Ia can be malate salt.
  • the compound of Formula Ia can be the (L)-malate salt.
  • Compound 1 can be (D)-malate salt.
  • Compound 1 can be the malate salt.
  • Compound 1 can be the (D)-malate salt.
  • the malate salt is in the crystalline N-1 form of the (L) malate salt and/or the (D) malate salt of the Compound 1 as disclosed in U.S. patent Application Ser. No. 61/325,095. Also see WO 2008/083319 for the properties of crystalline enantiomers, including the N-1 and/or the N-2 crystalline forms of the malate salt of Compound 1. Methods of making and characterizing such forms are fully described in PCT/US10/21194, which is incorporated herein by reference in its entirety.
  • the invention is directed to a method for ameliorating the symptoms of osteoblastic bone metastases, comprising administering to a patient in need of such treatment a therapeutically effective amount of a compound of Formula I in any of the embodiments disclosed herein.
  • the Compound of Formula I is Compound 1.
  • the compound of Formula I is administered post-taxotere treatment.
  • the Compound of Formula I is Compound 1.
  • the compound of Formula I is as effective or more effective than mitoxantrone plus prednisone.
  • the Compound of Formula I is Compound 1.
  • the Compound of Formula I, Ia, or Compound 1 or a pharmaceutically acceptable salt thereof is administered orally once daily as a tablet or capsule.
  • Compound 1 is administered orally as its free base or malate salt as a capsule or tablet.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing up to 100 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 100 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 95 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 90 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 85 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 80 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 75 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 70 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 65 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 60 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 55 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 50 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 45 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 40 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 30 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 25 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 20 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 15 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 10 mg of Compound 1.
  • Compound 1 is administered orally once daily as its free base or as the malate salt as a capsule or tablet containing 5 mg of Compound 1.
  • Compound 1 is administered as its free base or malate salt orally once daily as a tablet as provided in the following table.
  • Compound 1 is administered orally as its free base or malate salt once daily as a tablet as provided in the following table.
  • Compound 1 is administered orally as its free base or malate salt once daily as a tablet as provided in the following table.
  • Theoretical Quantity (mg/unit Ingredient dose) Compound 1 100.0 Microcrystalline Cellulose PH- 155.4 102 Lactose Anhydrous 60M 77.7 Hydroxypropyl Cellulose, EXF 12.0 Croscarmellose Sodium 24 Colloidal Silicon Dioxide 1.2 Magnesium Stearate (Non- 3.0 Bovine) Opadry Yellow 16.0 Total 416
  • any of the tablet formulations provided above can be adjusted according to the dose of Compound 1 desired.
  • the amount of each of the formulation ingredients can be proportionally adjusted to provide a table formulation containing various amounts of Compound 1 as provided in the previous paragraphs.
  • the formulations can contain 20, 40, 60, or 80 mg of Compound 1.
  • Administration of the compound of Formula I, Formula Ia, or Compound 1, or a pharmaceutically acceptable salt thereof, in pure form or in an appropriate pharmaceutical composition can be carried out via any of the accepted modes of administration or agents for serving similar utilities.
  • administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, suppositories, pills, soft elastic and hard gelatin dosages (which can be in capsules or tablets), powders, solutions, suspensions, or aerosols, or the like, specifically in unit dosage forms suitable for simple administration of precise dosages.
  • compositions will include a conventional pharmaceutical carrier or excipient and a compound of Formula I as the/an active agent, and, in addition, may include carriers and adjuvants, etc.
  • Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • a pharmaceutical composition of the compound of Formula I may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
  • compositions depend on various factors such as the mode of drug administration (e.g., for oral administration, compositions in the form of tablets, pills or capsules) and the bioavailability of the drug substance.
  • pharmaceutical compositions have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size.
  • U.S. Pat. No. 4,107,288 describes a pharmaceutical composition having particles in the size range from 10 to 1,000 nm in which the active material is supported on a crosslinked matrix of macromolecules.
  • 5,145,684 describes the production of a pharmaceutical composition in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical composition that exhibits remarkably high bioavailability.
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • One specific route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia
  • humectants as for example, glycerol
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate
  • solution retarders as for example paraffin
  • absorption accelerators as for example, quaternary
  • Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., the compound of Formula I, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administration are, for example, suppositories that can be prepared by mixing the compound of Formula I with, for example, suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • Dosage forms for topical administration of the compound of Formula I include ointments, powders, sprays, and inhalants.
  • the active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required.
  • Ophthalmic compositions, eye ointments, powders, and solutions are also contemplated as being within the scope of this disclosure.
  • Compressed gases may be used to disperse the compound of Formula I in aerosol form.
  • Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc.
  • the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of a compound(s) of Formula I, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a suitable pharmaceutical excipient.
  • the composition will be between about 5% and about 75% by weight of a compound(s) of Formula I, Formula Ia, or Compound 1, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
  • composition to be administered will, in any event, contain a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this disclosure.
  • the compounds of this disclosure are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy.
  • the compound of Formula I, Formula Ia, or Compound 1 can be administered to a patient at dosage levels in the range of about 0.1 to about 1,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example.
  • the specific dosage used can vary.
  • the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used.
  • the determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
  • the compound of Formula I, Formula Ia, or Compound 1 can be administered to the patient concurrently with other cancer treatments.
  • treatments include other cancer chemotherapeutics, hormone replacement therapy, radiation therapy, or immunotherapy, among others.
  • the choice of other therapy will depend on a number of factors including the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy.
  • a reactor was charged sequentially with 6,7-dimethoxy-quinoline-4-ol (10.0 kg) and acetonitrile (64.0 L). The resulting mixture was heated to approximately 65° C. and phosphorus oxychloride (POCl 3 , 50.0 kg) was added. After the addition of POCl 3 , the temperature of the reaction mixture was raised to approximately 80° C. The reaction was deemed complete (approximately 9.0 hours) when less than 2 percent of the starting material remained (in process high-performance liquid chromotography [HPLC] analysis). The reaction mixture was cooled to approximately 10° C.
  • phosphorus oxychloride POCl 3 , 50.0 kg
  • a reactor was sequentially charged with 4-chloro-6,7-dimethoxy-quinoline (8.0 kg), 4 nitrophenol (7.0 kg), 4 dimethylaminopyridine (0.9 kg), and 2,6 lutidine (40.0 kg).
  • the reactor contents were heated to approximately 147° C.
  • the reaction was complete (less than 5 percent starting material remaining as determined by in process HPLC analysis, approximately 20 hours)
  • the reactor contents were allowed to cool to approximately 25° C.
  • Methanol (26.0 kg) was added, followed by potassium carbonate (3.0 kg) dissolved in water (50.0 kg).
  • the reactor contents were stirred for approximately 2 hours.
  • the resulting solid precipitate was filtered, washed with water (67.0 kg), and dried at 25° C. for approximately 12 hours to afford the title compound (4.0 kg).
  • Triethylamine (8.0 kg) was added to a cooled (approximately 4° C.) solution of commercially available cyclopropane-1,1-dicarboxylic acid (2 l, 10.0 kg) in THF (63.0 kg) at a rate such that the batch temperature did not exceed 10° C.
  • the solution was stirred for approximately 30 minutes, and then thionyl chloride (9.0 kg) was added, keeping the batch temperature below 10° C.
  • a solution of 4-fluoroaniline (9.0 kg) in THF (25.0 kg) was added at a rate such that the batch temperature did not exceed 10° C.
  • the mixture was stirred for approximately 4 hours and then diluted with isopropyl acetate (87.0 kg).
  • Oxalyl chloride (1.0 kg) was added to a solution of 1-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (2.0 kg) in a mixture of THF (11 kg) and N,N-dimethylformamide
  • a reactor was charged sequentially with 6,7-dimethoxy-quinoline-4-ol (47.0 kg) and acetonitrile (318.8 kg). The resulting mixture was heated to approximately 60° C. and phosphorus oxychloride (POCl 3 , 130.6 kg) was added. After the addition of POCl 3 , the temperature of the reaction mixture was raised to approximately 77° C. The reaction was deemed complete (approximately 13 hours) when less than 3% of the starting material remained (in-process high-performance liquid chromatography [HPLC] analysis). The reaction mixture was cooled to approximately 2-7° C.
  • POCl 3 phosphorus oxychloride
  • Triethylamine (19.5 kg) was added to a cooled (approximately 5° C.) solution of cyclopropane-1,1-dicarboxylic acid (24.7 kg) in THF (89.6 kg) at a rate such that the batch temperature did not exceed 5° C.
  • the solution was stirred for approximately 1.3 hours, and then thionyl chloride (23.1 kg) was added, keeping the batch temperature below 10° C. When the addition was complete, the solution was stirred for approximately 4 hours keeping temperature below 10° C.
  • a solution of 4-fluoroaniline (18.0 kg) in THF (33.1 kg) was then added at a rate such that the batch temperature did not exceed 10° C. The mixture was stirred for approximately 10 hours after which the reaction was deemed complete.
  • reaction mixture was then diluted with isopropyl acetate (218.1 kg). This solution was washed sequentially with aqueous sodium hydroxide (10.4 kg, 50 percent dissolved in 119 L of water) further diluted with water (415 L), then with water (100 L) and finally with aqueous sodium chloride (20.0 kg dissolved in 100 L of water). The organic solution was concentrated by vacuum distillation (100 L residual volume) below 40° C. followed by the addition of n-heptane (171.4 kg), which resulted in the precipitation of solid.
  • the solid was recovered by filtration and washed with n-heptane (102.4 kg), resulting in wet, crude 1-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (29.0 kg).
  • the crude, 1-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid was dissolved in methanol (139.7 kg) at approximately 25° C. followed by the addition of water (320 L) resulting in slurry which was recovered by filtration, washed sequentially with water (20 L) and n-heptane (103.1 kg) and then dried on the filter at approximately 25° C. under nitrogen to afford the title compound (25.4 kg).
  • Oxalyl chloride (12.6 kg) was added to a solution of 1-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (22.8 kg) in a mixture of THF (96.1 kg) and N,N-dimethylformamide (DMF; 0.23 kg) at a rate such that the batch temperature did not exceed 25° C. This solution was used in the next step without further processing.
  • a reactor was charged with 1-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (35 kg), 344 g DMF, and 175 kg THF.
  • the reaction mixture was adjusted to 12-17° C. and then to the reaction mixture was charged 19.9 kg of oxalyl chloride over a period of 1 hour.
  • the reaction mixture was left stirring at 12-17° C. for 3 to 8 hours. This solution was used in the next step without further processing.
  • the product was recovered by filtration, washed with a pre-made solution of THF (68.6 kg) and water (256 L), and dried first on a filter under nitrogen at approximately 25° C. and then at approximately 45° C. under vacuum to afford the title compound (41.0 kg, 38.1 kg, calculated based on LOD).
  • a reactor was charged with 4-(6,7-dimethoxy-quinoline-4-yloxy)-phenylamine (35.7 kg, 1 equivalent), followed by 412.9 kg THF.
  • To the reaction mixture was charged a solution of 48.3 K 2 CO 3 in 169 kg water.
  • the acid chloride solution of described in the Alternative Preparation of 1-(4-Fluoro-phenylcarbamoyl)-cyclopropanecarbonyl chloride above was transferred to the reactor containing 4-(6,7-dimethoxy-quinoline-4-yloxy)-phenylamine while maintaining the temperature between 20-30° C. over a minimum of two hours.
  • the reaction mixture was stirred at 20-25° C. for a minimum of three hours.
  • the reaction temperature was then adjusted to 30-25° C.
  • the product was filtered and washed with a mixture of 179 kg water and 157.9 kg THF in two portions.
  • the crude product was dried under a vacuum for at least two hours.
  • the dried product was then taken up in 285.1 kg THF.
  • the resulting suspension was transferred to reaction vessel and agitated until the suspension became a clear (dissolved) solution, which required heating to 30-35° C. for approximately 30 minutes.
  • 456 kg water was then added to the solution, as well as 20 kg SDAG-1 ethanol (ethanol denatured with methanol over two hours.
  • the mixture was agitated at 15-25° C. fir at least 16 hours.
  • the product was filtered and washed with a mixture of 143 kg water and 126.7 THF in two portions.
  • the product was dried at a maximum temperature set point of 40° C.
  • reaction temperature during acid chloride formation was adjusted to 10-15° C.
  • the recrystallization temperature was changed from 15-25° C. to 45-50° C. for 1 hour and then cooled to 15-25° C. over 2 hours.
  • Cyclopropane-1,1-dicarboxylic acid [4-(6,7-dimethoxy-quinoline-4-yloxy)-phenyl]-amide(4-fluoro-phenyl)-amide (1-5; 13.3 kg), L-malic acid (4.96 kg), methyl ethyl ketone (MEK; 188.6 kg) and water (37.3 kg) were charged to a reactor and the mixture was heated to reflux (approximately 74° C.) for approximately 2 hours. The reactor temperature was reduced to 50 to 55° C. and the reactor contents were filtered.
  • Cyclopropane-1,1-dicarboxylic acid [4-(6,7-dimethoxy-quinoline-4-yloxy)-phenyl]-amide(4-fluoro-phenyl)-amide (47.9 kg), L-malic acid (17.2), 658.2 kg methyl ethyl ketone, and 129.1 kg water (37.3 kg) were charged to a reactor and the mixture was heated 50-55° C. for approximately 1-3 hours, and then at 55-60° C. for an additional 4-5 hours. The mixture was clarified by filtration through a 1 ⁇ m cartridge. The reactor temperature was adjusted to 20-25° C. and vacuum distilled with a vacuum at 150-200 mm Hg with a maximum jacket temperature of 55° C. to the volume range of 558-731 L.
  • the vacuum distillation was performed two more times with the charge of 380 kg and 380.2 kg methyl ethyl ketone, respectively.
  • the volume of the batch was adjusted to 18 v/w of cyclopropane-1,1-dicarboxylic acid [4-(6,7-dimethoxy-quinoline-4-yloxy)-phenyl]-amide(4-fluoro-phenyl)-amide by charging 159.9 kg methyl ethyl ketone to give a total volume of 880 L.
  • An additional vacuum distillation was carried out by adjusting 245.7 methyl ethyl ketone.
  • the reaction mixture was left with moderate agitation at 20-25° C. for at least 24 hours.
  • the product was filtered and washed with 415.1 kg methyl ethyl ketone in three portions.
  • the product was dried under a vacuum with the jacket temperature set point at 45° C.
  • HGF and MET signaling pathways appear to play important roles in osteoblast and osteoclast function. Strong immunohistochemical staining of MET has been observed in both cell types in developing bone. HGF and MET are expressed by osteoblasts and osteoclasts in vitro and mediate cellular responses such as proliferation, migration, and expression of ALP. Secretion of HGF by osteoblasts has been proposed as a key factor in osteoblast/osteoclast coupling, and in the development of bone metastases by tumor cells that express MET. Osteoblasts and osteoclasts also express VEGF and its receptors, and VEGF signaling in these cells is involved in potential autocrine and/or paracrine feedback mechanisms regulating cell migration, differentiation, and survival.
  • CRPC castration-resistant prostate cancer
  • Activation of the MET and VEGFR signaling pathways is implicated in the development of bone metastases in CRPC.
  • tALP total serum alkaline phosphatase
  • Compound 1 is an orally bioavailable multitargeted tyrosine kinase inhibitor with potent activity against MET and VEGFR2.
  • Compound 1 suppresses MET and VEGFR2 signaling, rapidly induces apoptosis of endothelial cells and tumor cells, and causes tumor regression in xenograft tumor models.
  • Compound 1 also significantly reduces tumor invasiveness and metastasis and substantially improves overall survival in a murine pancreatic neuroendocrine tumor model.
  • Compound 1 was generally well-tolerated, with fatigue, diarrhea, anorexia, rash, and palmar-plantar erythrodysesthesia being the most commonly observed adverse events.
  • Patient 1 was diagnosed with localized prostate cancer in 1993 and treated with radical prostatectomy (Gleason score unavailable; PSA, 0.99 ng/mL).
  • PSA radical prostatectomy
  • PSA 0.99 ng/mL
  • combined androgen blockade (CAB) with leuprolide and bicalutamide was initiated for rising PSA (3.5 ng/mL).
  • diethystillbestrol (DES) was administered briefly.
  • 6 cycles of docetaxel were given for new lung metastases. Rising PSA was unresponsive to antiandrogen withdrawal. Androgen ablation therapy was continued until clinical progression.
  • bone metastasis to the spine associated with impingement on the spinal cord and back pain was treated with radiation therapy (37.5 Gy).
  • Bone scan showed uptake of radiotracer in the left iliac wing, left sacroiliac joint, femoral head, and the pubic symphysis.
  • Biopsy of the left pubic ramus confirmed metastatic adenocarcinoma with mixed lytic and blastic lesions.
  • CAB with leuprolide and bicalutamide and radiation therapy (8 Gy) to the left pubic ramus and acetabulum resulted in bone pain relief and PSA normalization.
  • Rising PSA in November 2009 (16 ng/mL) was unresponsive to antiandrogen withdrawal.
  • a CT scan revealed retroperitoneal lymph node enlargement and liver metastases (PSA, 28.1 ng/mL). Further progression of disease was marked by recurrent bone pain, new lung and hepatic metastases.
  • a repeat bone scan showed new foci, and a CT scan showed an increase in the retroperitoneal, para-aortic, and bilateral common iliac adenopathy. Rising PSA in April 2010 (2.8 ng/mL) and increasing bone pain were unresponsive to antiandrogen withdrawal.
  • Patient 1 started Compound 1 on Feb. 12, 2010. Four weeks later, significant reduction in bone pain was reported. At Week 6, bone scan showed a dramatic decrease in radiotracer uptake by bone metastases ( FIG. 1A ). A CT scan showed a partial response (PR) with a 33% decrease in measurable target lesions ( FIG. 1C ). At Week 12, near complete resolution of bone lesions and a 44% decrease in target lesions was observed and was stable through Week 18. Corresponding with the bone scan response, after an initial rise, serum tALP levels decreased from 689 U/L at baseline to 159 U/L at Week 18 ( FIG. 1B and Table 1). In addition, there was an increase in hemoglobin of 1.4 g/dL at Week 2 compared with baseline (Table 1). PSA decreased from 430 ng/mL at baseline to 93.5 ng/mL at Week 18 ( FIG. 1B and Table 1). The patient was on open-label treatment through Week 18 when he withdrew after developing Grade 3 diarrhea.
  • Patient 2 started Compound 1 on Mar. 31, 2010. At Week 4, reduction in bone pain was reported. At Week 6, bone scan showed a slight flair in radiotracer uptake by bone lesions ( FIG. 2A ), and a CT scan showed a 13% decrease in target lesions ( FIG. 2C ). At Week 12, a substantial reduction of radiotracer uptake ( FIG. 2A ) and a 20% decrease in measurable disease were observed (Table 1). After randomization to placebo at Week 12 the patient developed severe bone pain and sacral nerve root impingement. Radiation to the spine was administered, and the patient crossed over to open-label Compound 1 treatment at Week 15. Serum tALP levels were within the normal range (101-144 U/L) ( FIG. 2B ).
  • Hemoglobin increased by 1.8 g/dL at Week 12 compared with baseline (Table 1).
  • PSA peaked at close to 6-fold of baseline by Week 16, but then decreased to 2-fold of baseline by Week 18 subsequent to crossing over to Compound 1 from placebo ( FIG. 2B and Table 1).
  • the patient continues on Compound 1 treatment as of September 2010.
  • Patient 3 started Compound 1 on Apr. 26, 2010. After three weeks a complete resolution of pain was reported. At Week 6, bone scan showed a dramatic reduction in radiotracer uptake ( FIG. 3A ), and a CT scan showed a PR with a 43% decrease in measurable target lesions. At Week 12 a complete resolution of bone lesions on bone scan ( FIG. 3A ) and a 51% decrease in measurable disease were observed (Table 1 and FIG. 3B )). After an initial rise, serum tALP levels steadily decreased, with tALP at 869 U/L at baseline and 197 U/L at Week 18 ( FIG. 3B and Table 1). Hemoglobin increased 2.2 g/dL at Week 2 compared with baseline (Table 1). PSA decreased from 2.4 ng/mL at screening to 1.2 ng/mL at Week 18 ( FIG. 3B and Table 1). The patient continues on Compound 1 treatment as of September 2010.
  • Uptake of radiotracer in bone depends on both local blood flow and osteoblastic activity, both of which may be pathologically modulated by the tumor cells associated with the bone lesion. Resolving uptake may therefore be attributable to either interruption of local blood flow, direct modulation of osteoblastic activity, a direct effect on the tumor cells in bone, or a combination of these processes.
  • decreased uptake on bone scan in men with CRPC has only been rarely noted with VEGF/VEGFR targeted therapy, despite numerous trials with such agents.
  • observations of decreased uptake on bone scan in CRPC patients have only been reported rarely for abiraterone, which targets the cancer cells directly, and for dasatinib, which targets both cancer cells and osteoclasts.
  • targeting angiogenesis alone, or selectively targeting the tumor cells and/or osteoclasts has not resulted in effects similar to those observed in the patients treated with Compound 1.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Quinoline Compounds (AREA)
  • Steroid Compounds (AREA)
  • Peptides Or Proteins (AREA)
US13/876,290 2010-09-27 2011-09-26 Method of Treating Cancer Abandoned US20140057943A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/876,290 US20140057943A1 (en) 2010-09-27 2011-09-26 Method of Treating Cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US38695910P 2010-09-27 2010-09-27
US201161481671P 2011-05-02 2011-05-02
PCT/US2011/053233 WO2012044572A1 (en) 2010-09-27 2011-09-26 Dual inhibitors of met and vegf for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
US13/876,290 US20140057943A1 (en) 2010-09-27 2011-09-26 Method of Treating Cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/053233 A-371-Of-International WO2012044572A1 (en) 2010-09-27 2011-09-26 Dual inhibitors of met and vegf for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/843,480 Continuation US20160220554A1 (en) 2010-09-27 2015-09-02 Method of Treating Cancer

Publications (1)

Publication Number Publication Date
US20140057943A1 true US20140057943A1 (en) 2014-02-27

Family

ID=44741730

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/876,290 Abandoned US20140057943A1 (en) 2010-09-27 2011-09-26 Method of Treating Cancer
US14/843,480 Abandoned US20160220554A1 (en) 2010-09-27 2015-09-02 Method of Treating Cancer
US16/905,307 Active 2032-05-15 US11612597B2 (en) 2010-09-27 2020-06-18 Method of treating cancer
US18/112,214 Active US11969419B2 (en) 2010-09-27 2023-02-21 Method of treating cancer
US18/423,824 Pending US20240156803A1 (en) 2010-09-27 2024-01-26 Method of Treating Cancer

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/843,480 Abandoned US20160220554A1 (en) 2010-09-27 2015-09-02 Method of Treating Cancer
US16/905,307 Active 2032-05-15 US11612597B2 (en) 2010-09-27 2020-06-18 Method of treating cancer
US18/112,214 Active US11969419B2 (en) 2010-09-27 2023-02-21 Method of treating cancer
US18/423,824 Pending US20240156803A1 (en) 2010-09-27 2024-01-26 Method of Treating Cancer

Country Status (22)

Country Link
US (5) US20140057943A1 (hu)
EP (1) EP2621481B2 (hu)
JP (2) JP6158705B2 (hu)
KR (1) KR101882328B1 (hu)
CN (1) CN103402505A (hu)
AR (1) AR085195A1 (hu)
AU (2) AU2011307304C1 (hu)
BR (1) BR112013007220A2 (hu)
CA (1) CA2812750C (hu)
DK (1) DK2621481T4 (hu)
EA (1) EA027476B1 (hu)
ES (1) ES2754973T5 (hu)
FI (1) FI2621481T4 (hu)
HU (1) HUE045810T2 (hu)
IL (1) IL225507B (hu)
MX (1) MX352926B (hu)
PL (1) PL2621481T5 (hu)
PT (1) PT2621481T (hu)
SI (1) SI2621481T2 (hu)
TW (2) TW201302709A (hu)
UA (1) UA114172C2 (hu)
WO (1) WO2012044572A1 (hu)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9717720B2 (en) 2011-02-10 2017-08-01 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US9861624B2 (en) 2012-05-02 2018-01-09 Exelixis, Inc. Method of treating cancer
US9969692B2 (en) 2011-10-20 2018-05-15 Exelixis, Inc. Process for preparing quinoline derivatives
US10034873B2 (en) 2010-07-16 2018-07-31 Exelixis, Inc. C-met modulator pharmaceutical compositions
US10159666B2 (en) 2014-03-17 2018-12-25 Exelixis, Inc. Dosing of cabozantinib formulations
US10166225B2 (en) 2011-09-22 2019-01-01 Exelixis, Inc. Method for treating osteoporosis
US10273211B2 (en) 2013-03-15 2019-04-30 Exelixis, Inc. Metabolites of N-{4-([6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US10501418B2 (en) 2014-02-14 2019-12-10 Exelixis, Inc. Crystalline solid forms of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US11065240B2 (en) 2014-08-05 2021-07-20 Exelixis, Inc. Drug combinations to treat multiple myeloma
US11116759B2 (en) 2011-04-04 2021-09-14 Exelixis, Inc. Method of treating cancer
US11124481B2 (en) 2014-07-31 2021-09-21 Exelixis, Inc. Method of preparing fluorine-18 labeled Cabozantinib and its analogs
US11141413B2 (en) 2016-04-15 2021-10-12 Exelixis, Inc. Method of treating renal cell carcinoma using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate
US11504363B2 (en) 2011-05-02 2022-11-22 Exelixis, Inc. Method of treating cancer and bone cancer pain
US11564915B2 (en) 2013-04-04 2023-01-31 Exelixis, Inc. Cabozantinib dosage form and use in the treatment of cancer
US11612597B2 (en) 2010-09-27 2023-03-28 Exelixis, Inc. Method of treating cancer

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140323522A1 (en) * 2011-11-08 2014-10-30 Exelixis, Inc. Method of Treating Cancer
CN103664776B (zh) * 2012-09-26 2016-05-04 正大天晴药业集团股份有限公司 一种酪氨酸激酶抑制剂及其中间体的制备方法
JP2016515628A (ja) * 2013-04-04 2016-05-30 エグゼリクシス, インコーポレイテッド 癌を治療するための複合薬
CN104788372B (zh) * 2014-07-25 2018-01-30 上海圣考医药科技有限公司 一种氘代卡博替尼衍生物、其制备方法、应用及其中间体
CN107198770A (zh) * 2016-03-18 2017-09-26 日本医事物理股份有限公司 成骨型骨转移模型动物的制备方法
CN108503650B (zh) * 2017-02-27 2021-02-12 北京赛特明强医药科技有限公司 二噁烷并喹唑啉类化合物或其药用盐或其水合物及其作为酪氨酸激酶抑制剂的应用
AU2018272088C1 (en) 2017-05-26 2022-09-22 Exelixis, Inc. Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-N'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
WO2019133632A1 (en) 2017-12-27 2019-07-04 St. Jude Children's Research Hospital Methods of treating disorders associated with castor
SG11202006047RA (en) 2017-12-27 2020-07-29 St Jude Childrens Res Hospital Inc Small molecule modulators of pantothenate kinases
CA3088200A1 (en) 2018-01-26 2019-08-01 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
JP7022454B2 (ja) * 2018-02-11 2022-02-18 北京賽特明強医薬科技有限公司 ジオキシノキノリン系化合物、その調製方法および使用
EP4262814A1 (en) * 2020-12-16 2023-10-25 St. Jude Children's Research Hospital, Inc. Methods of treating disorders associated with castor

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
EP1447405A4 (en) 2001-10-17 2005-01-12 Kirin Brewery QUINOLINE OR QUINAZOLINE DERIVATIVES INHIBITING THE AUTOPHOSPHORYLATION OF FIBROBLAST GROWTH FACTOR RECEPTORS
GB0219746D0 (en) 2002-08-23 2002-10-02 Inst Of Ex Botany Ascr Azapurine derivatives
EP1559715B1 (en) 2002-10-21 2007-09-26 Kirin Beer Kabushiki Kaisha N-{2-chloro-4-[(6,7-dimethoxy-4-quinolyl)oxy]phenyl}-n'-(5-methyl-3-isoxazolyl)urea salt in crystalline form
EP1566379A4 (en) 2002-10-29 2005-11-09 Kirin Brewery QUINOLINE AND QUINAZOLINE DERIVATIVES INHIBITING THE AUTOPHOSPHORYLATION OF FLT3 AND MEDICAL COMPOSITIONS CONTAINING SAME
PT1562955E (pt) 2002-11-04 2008-05-05 Astrazeneca Ab Derivados de quinazolina como inibidores da scr tirosina-cinase
US7432275B2 (en) 2002-12-13 2008-10-07 Neurogen Corporation Carboxylic acid, phosphate or phosphonate substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
CN1729001A (zh) 2002-12-18 2006-02-01 辉瑞产品公司 用于治疗异常细胞生长的4-苯胺基喹唑啉衍生物
AU2003299943A1 (en) 2002-12-23 2004-07-22 Ariad Pharmaceuticals, Inc. Heterocycles and uses thereof
AU2003292838A1 (en) 2002-12-27 2004-07-29 Kirin Beer Kabushiki Kaisha Therapeutic agent for wet age-related macular degeneration
ES2371383T3 (es) 2003-09-26 2011-12-30 Exelixis, Inc. N-[3-fluoro-4-({6-(metiloxi)-7-[(3-morfolin-4-ilpropil)oxi]quinolin-4-il}oxi)fenil]-n'-(4-fluorofenil)ciclopropan-1,1-dicarboxamida para el tratamiento del cáncer.
CA2572331A1 (en) 2004-07-02 2006-02-09 Exelixis, Inc. C-met modulators and method of use
CA2603748A1 (en) 2005-04-06 2006-10-12 Exelixis, Inc. C-met modulators and methods of use
US20080004273A1 (en) 2006-05-30 2008-01-03 Stephane Raeppel Inhibitors of protein tyrosine kinase activity
CA2671982C (en) 2006-12-14 2016-01-26 Exelixis, Inc. Methods of using mek inhibitors
WO2008083319A1 (en) 2006-12-29 2008-07-10 Il Yang Pharmaceutical Company, Ltd. Solid state forms of enantiopure ilaprazole
WO2009026717A1 (en) * 2007-08-29 2009-03-05 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
TW200922590A (en) * 2007-09-10 2009-06-01 Curis Inc VEGFR inhibitors containing a zinc binding moiety
UY31800A (es) 2008-05-05 2009-11-10 Smithkline Beckman Corp Metodo de tratamiento de cancer usando un inhibidor de cmet y axl y un inhibidor de erbb
JP5699075B2 (ja) * 2008-05-14 2015-04-08 アムジエン・インコーポレーテツド 癌の治療のためのvegf(r)阻害剤および肝細胞増殖因子(c−met)阻害剤との組合せ
TW201035017A (en) 2008-09-26 2010-10-01 Smithkline Beecham Corp Preparation of a quinolinyloxydiphenylcyclopropanedicarboxamide
AU2009300328A1 (en) 2008-10-01 2010-04-08 Ludwig Institute For Cancer Research Methods for the treatment of cancer
JP5486606B2 (ja) 2008-11-13 2014-05-07 エクセリクシス, インク. キノリン誘導体の調製方法
JP2010126530A (ja) * 2008-12-01 2010-06-10 Takeda Chem Ind Ltd 縮合複素環誘導体およびその用途
US20130030172A1 (en) 2008-12-04 2013-01-31 Exelixis, Inc. Methods of Preparing Quinoline Derivatives
CN110818633A (zh) 2009-01-16 2020-02-21 埃克塞里艾克西斯公司 一种苹果酸盐及其晶型
WO2011009095A1 (en) 2009-07-17 2011-01-20 Exelixis, Inc. Crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quin0lin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide
UA108618C2 (uk) 2009-08-07 2015-05-25 Застосування c-met-модуляторів в комбінації з темозоломідом та/або променевою терапією для лікування раку
EP2475390A4 (en) 2009-09-09 2014-01-01 Quintiles Transnat Corp METHODS AND COMPOSITIONS FOR THE TREATMENT OF TYROSINE KINASE RECEPTOR MEDIATION DISEASES OR DISORDERS
EP2545038A1 (en) 2010-03-12 2013-01-16 Exelixis, Inc. Hydrated crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US20120070368A1 (en) 2010-04-16 2012-03-22 Exelixis, Inc. Methods of Using C-Met Modulators
WO2012009723A1 (en) 2010-07-16 2012-01-19 Exelixis, Inc. C-met modulator pharmaceutical compositions
EP4014971A1 (en) 2010-07-16 2022-06-22 Exelixis, Inc. C-met modulator pharmaceutical compositions
JP2013540759A (ja) 2010-09-27 2013-11-07 エクセリクシス, インク. 去勢抵抗性前立腺癌および造骨性転移の治療のためのmetおよびvegfの二元阻害薬
EP2621483A1 (en) 2010-09-27 2013-08-07 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
FI2621481T4 (fi) * 2010-09-27 2023-01-13 Met- ja vegf-kaksoisestäjiä kastraatioresistentin eturauhassyövän ja osteoblastisten luun etäispesäkkeiden hoitamiseen
CN103327979A (zh) 2010-11-22 2013-09-25 葛兰素史密斯克莱知识产权(第2号)有限公司 治疗癌症的方法
CA2826751C (en) 2011-02-10 2021-05-18 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US20120252840A1 (en) 2011-04-04 2012-10-04 Exelixis, Inc. Method of Treating Cancer
MX2013012695A (es) 2011-05-02 2014-03-27 Exelixis Inc Metodo para el tratamiento del cancer y el dolor por cancer de hueso.
TW201306842A (zh) 2011-06-15 2013-02-16 Exelixis Inc 使用pi3k/mtor吡啶並嘧啶酮抑制劑及苯達莫司汀及/或利妥昔單抗治療惡性血液疾病之組合療法
EA201490676A1 (ru) 2011-09-22 2015-02-27 Экселиксис, Инк. Способ лечения остеопороза
CN110511158A (zh) 2011-10-20 2019-11-29 埃克塞里艾克西斯公司 制备喹啉衍生物的方法
US20140323522A1 (en) 2011-11-08 2014-10-30 Exelixis, Inc. Method of Treating Cancer
WO2013166296A1 (en) 2012-05-02 2013-11-07 Exelixis, Inc. A dual met - vegf modulator for treating osteolytic bone metastases
CN104703600A (zh) 2012-09-07 2015-06-10 埃克塞里艾克西斯公司 用于治疗肺腺癌的met、vegfr和ret的抑制剂
US20140221372A1 (en) 2013-02-06 2014-08-07 GlaxoSmithKline Intellectual Property (NO 2.) Limited Method of administration and treatment
CN105121412B (zh) 2013-03-15 2019-07-12 埃克塞里艾克西斯公司 N-(4-{[6,7-双(甲基氧基)喹啉-4-基]氧基}苯基)-n′-(4-氟苯基)环丙烷-1,1-二甲酰胺的代谢物
JP2016515628A (ja) 2013-04-04 2016-05-30 エグゼリクシス, インコーポレイテッド 癌を治療するための複合薬

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Verras et al 'The Androgen Receptor Negatively Regulates the Expression of c-Met: Implications for a Novel Mechanism of Prostate Cancer Progression'Cancer Research, Vol. 67, p. 967-975, 2007. *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11123338B2 (en) 2010-07-16 2021-09-21 Exelixis, Inc. C-met modulator pharmaceutical compositions
US10548888B2 (en) 2010-07-16 2020-02-04 Exelixis C-Met modulator pharmaceutical compositions
US10034873B2 (en) 2010-07-16 2018-07-31 Exelixis, Inc. C-met modulator pharmaceutical compositions
US10039757B2 (en) 2010-07-16 2018-08-07 Exelixis, Inc. C-Met modulator pharmaceutical compositions
US11969419B2 (en) 2010-09-27 2024-04-30 Exelixis, Inc. Method of treating cancer
US11612597B2 (en) 2010-09-27 2023-03-28 Exelixis, Inc. Method of treating cancer
US9717720B2 (en) 2011-02-10 2017-08-01 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US10123999B2 (en) 2011-02-10 2018-11-13 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US10543206B2 (en) 2011-02-10 2020-01-28 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US11298349B2 (en) 2011-02-10 2022-04-12 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US11116759B2 (en) 2011-04-04 2021-09-14 Exelixis, Inc. Method of treating cancer
US11504363B2 (en) 2011-05-02 2022-11-22 Exelixis, Inc. Method of treating cancer and bone cancer pain
US10166225B2 (en) 2011-09-22 2019-01-01 Exelixis, Inc. Method for treating osteoporosis
US9969692B2 (en) 2011-10-20 2018-05-15 Exelixis, Inc. Process for preparing quinoline derivatives
US9861624B2 (en) 2012-05-02 2018-01-09 Exelixis, Inc. Method of treating cancer
US10273211B2 (en) 2013-03-15 2019-04-30 Exelixis, Inc. Metabolites of N-{4-([6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US11564915B2 (en) 2013-04-04 2023-01-31 Exelixis, Inc. Cabozantinib dosage form and use in the treatment of cancer
US10851061B2 (en) 2014-02-14 2020-12-01 Exelixis, Inc. Crystalline solid forms of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US10501418B2 (en) 2014-02-14 2019-12-10 Exelixis, Inc. Crystalline solid forms of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US11724986B2 (en) 2014-02-14 2023-08-15 Exelixis, Inc. Crystalline solid forms of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US11760726B2 (en) 2014-02-14 2023-09-19 Exelixis, Inc. Crystalline solid forms of N-{4-[(6,7-Dimethoxyquinolin-4-yl)oxy]phenyl} -n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US10159666B2 (en) 2014-03-17 2018-12-25 Exelixis, Inc. Dosing of cabozantinib formulations
US11124481B2 (en) 2014-07-31 2021-09-21 Exelixis, Inc. Method of preparing fluorine-18 labeled Cabozantinib and its analogs
US11065240B2 (en) 2014-08-05 2021-07-20 Exelixis, Inc. Drug combinations to treat multiple myeloma
US11141413B2 (en) 2016-04-15 2021-10-12 Exelixis, Inc. Method of treating renal cell carcinoma using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate

Also Published As

Publication number Publication date
HUE045810T2 (hu) 2020-01-28
US11612597B2 (en) 2023-03-28
UA114172C2 (uk) 2017-05-10
EP2621481B1 (en) 2019-08-14
MX2013003594A (es) 2013-08-01
AU2011307304A1 (en) 2013-05-02
MX352926B (es) 2017-12-14
FI2621481T4 (fi) 2023-01-13
US20240156803A1 (en) 2024-05-16
PL2621481T3 (pl) 2020-03-31
US20230301980A1 (en) 2023-09-28
DK2621481T3 (da) 2019-10-21
JP2017149714A (ja) 2017-08-31
KR20140025304A (ko) 2014-03-04
KR101882328B1 (ko) 2018-07-27
WO2012044572A1 (en) 2012-04-05
US11969419B2 (en) 2024-04-30
DK2621481T4 (da) 2023-01-09
AU2011307304C1 (en) 2016-10-20
AR085195A1 (es) 2013-09-18
SI2621481T1 (sl) 2019-11-29
US20160220554A1 (en) 2016-08-04
PT2621481T (pt) 2019-11-19
ES2754973T5 (es) 2023-03-13
BR112013007220A2 (pt) 2017-08-08
PL2621481T5 (pl) 2023-03-13
TW201302709A (zh) 2013-01-16
SI2621481T2 (sl) 2023-02-28
IL225507B (en) 2019-08-29
JP6158705B2 (ja) 2017-07-05
EP2621481A1 (en) 2013-08-07
IL225507A0 (en) 2013-06-27
TW201738213A (zh) 2017-11-01
CA2812750C (en) 2020-10-06
CA2812750A1 (en) 2012-04-05
EA027476B1 (ru) 2017-07-31
JP2013537916A (ja) 2013-10-07
EA201390465A1 (ru) 2013-08-30
CN103402505A (zh) 2013-11-20
AU2016204747A1 (en) 2016-07-28
ES2754973T3 (es) 2020-04-21
AU2011307304B2 (en) 2016-04-14
US20200330450A1 (en) 2020-10-22
EP2621481B2 (en) 2022-10-19

Similar Documents

Publication Publication Date Title
US11969419B2 (en) Method of treating cancer
US11504363B2 (en) Method of treating cancer and bone cancer pain
US9861624B2 (en) Method of treating cancer
US11116759B2 (en) Method of treating cancer
US20140323522A1 (en) Method of Treating Cancer
US20140330170A1 (en) Method of Quantifying Cancer Treatment
NZ716805B2 (en) Method of treating cancer and bone cancer pain
NZ617508B2 (en) Method of treating cancer and bone cancer pain

Legal Events

Date Code Title Description
AS Assignment

Owner name: EXELIXIS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE REGENTS OF THE UNIVERSITY OF MICHIGAN;REEL/FRAME:035711/0339

Effective date: 20110926

Owner name: THE REGENTS OF THE UNIVERSITY OF MICHIGAN, MICHIGA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SMITH, DAVID;HUSSAIN, MAHA;REEL/FRAME:035711/0138

Effective date: 20110927

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: EXELIXIS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REGENTS OF THE UNIVERSITY OF MICHIGAN;REEL/FRAME:052834/0786

Effective date: 20110926

Owner name: EXELIXIS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REGENTS OF THE UNIVERSITY OF MICHIGAN;REEL/FRAME:052834/0545

Effective date: 20110926

Owner name: REGENTS OF THE UNIVERSITY OF MICHIGAN, MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUSSAIN, MAHA;SMITH, DAVID;REEL/FRAME:052834/0751

Effective date: 20110926

Owner name: REGENTS OF THE UNIVERSITY OF MICHIGAN, MICHIGAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUSSAIN, MAHA;SMITH, DAVID;REEL/FRAME:052834/0522

Effective date: 20110926