WO2012103060A1 - Procédé d'administration et de traitement - Google Patents

Procédé d'administration et de traitement Download PDF

Info

Publication number
WO2012103060A1
WO2012103060A1 PCT/US2012/022316 US2012022316W WO2012103060A1 WO 2012103060 A1 WO2012103060 A1 WO 2012103060A1 US 2012022316 W US2012022316 W US 2012022316W WO 2012103060 A1 WO2012103060 A1 WO 2012103060A1
Authority
WO
WIPO (PCT)
Prior art keywords
genotype
patient
cancer
single nucleotide
nucleotide polymorphism
Prior art date
Application number
PCT/US2012/022316
Other languages
English (en)
Inventor
Chun-Fang Xu
Zhengyu XUE
Original Assignee
Glaxo Wellcome Manufacturing Pte Ltd
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 filed Critical Glaxo Wellcome Manufacturing Pte Ltd
Publication of WO2012103060A1 publication Critical patent/WO2012103060A1/fr

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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

Definitions

  • the present invention relates to the administration of drug and its effects on patients with particular genetic variants.
  • Pazopanib is an oral angiogenesis inhibitor targeting vascular endothelial growth factor receptors (VEGFR) -1, -2, and -3; platelet derived growth factor receptors (PDGFR)-a and - ⁇ ; and the stem cell factor receptor, c-Kit.
  • VEGFR vascular endothelial growth factor receptors
  • PDGFR platelet derived growth factor receptors
  • stem cell factor receptor c-Kit
  • Pazopanib-induced hyperbilirubinemia is associated with Gilbert's syndrome UGT1A1 polymorphism Br. J. Cancer, 102: 1371-1377 (2010)].
  • Evidence of association between VEGFA genotypes and median overall survival or PFS has been shown in breast or ovarian cancer patients treated with bevacizumab in combination with chemotherapy agents (Schneider et al 2008, Schultheis et al 2008).
  • IL8 -251T>A polymorphism was seen to be associated with response rate (RR) in patients with ovarian cancer treated with cyclophosphamide and bevacizumab; patients with the variant AA or AT genotypes had a statistically significant lower response rate than those with the corresponding TT genotype (Schultheis et al 2008).
  • RR response rate
  • the inventor has discovered that patients who have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236) and are administered a VEGF or VEGFR inhibitor to treat a condition such as cancer experience a reduced overall survival than patients who have a genotype at the rsl 126647 reference single nucleotide polymorphism other than the TT genotype.
  • a method of administering a VEGF or VEGFR inhibitor to a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), administering to said patient the VEGF or VEGFR inhibitor.
  • a method of prescribing a VEGF or VEGFR inhibitor to a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), prescribing to said patient the VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), administering to said patient a VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof, the patient having been previously genotyped as having a genotype other than the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), includes administering to the patient a VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof includes administering to the patient a VEGF or VEGFR inhibitor, and then determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCB1 1236).
  • the present invention also provides methods comprising determining if patient has the TT, TA or AA genotype at the rs2032582 reference single nucleotide polymorphism (ABCB1 2667) and accordingly administering to the patient a VEGF or VEGFR inhibitor.
  • methods of treating a human with cancer comprising:
  • the human has a neutrophil count ⁇ ULN. In another aspect, the human has BMI of about 26 or greater.
  • methods for treating a human with cancer comprising:
  • the VEGF or VEGFR inhibitor is a compound of formula (I):
  • the human may have renal cancer.
  • the present invention further comprises detecting whether said human has one or more of the following variant alleles: IL8 -251T>A and ABCB1 2677G>T/A.
  • Methods are provided for treating a human with cancer with a VEGF or VEGFR inhibitor if said human has 0, 1 or 2 copies of IL8 -251T>A and ABCB1 26770T/A. According to another aspect of the present invention, methods are provided for treating a human with cancer comprising:
  • Figure 5 Overall survival Kaplan-Meier curves for patients with different numbers of variant alleles of the IL8 2767A>T, FGFR2 IVS2 906OT, NR1I2 -253850T, and ABCBl 1236C>T polymorphisms. Patients were categorized into three groups: 1) those with two or fewer copies of the risk alleles (favorable profile); 2) those with 3 to 5 copies of the risk alleles; 3) those with 6 or 7 copies of the risk alleles (unfavorable profile). CI, confidence interval; HR, hazard ratio; OS, overall survival.
  • solvate refers to a complex of variable stoichiometry formed by a solute (in this invention, compounds of formula (I), (II), (III), 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.
  • suitable pharmaceutically acceptable solvents include, without limitation, water, ethanol and acetic acid.
  • the solvent used is water.
  • One of ordinary skill in the art will readily appreciate how to determine if a solvate of compounds I, ⁇ , and/or I" will form and how to determine the composition of the solvate using standard solvate screening technology understood by those skilled in the art, for example.
  • rsl 128503 reference single nucleotide polymorphism is understood to have the sequence:
  • polymorphisms which are similar to the [C/T] polymorphism shown in the sequence can also exist, namely [C/G] and [C/A].
  • rsl 128503 it is meant to include the [C/T], [C/G], and [C/A] polymorphisms.
  • rsl 128503 reference single nucleotide polymorphisms for which a sequence is shown above can be detected using various oligonucleotides as will be understood by those skilled in the art, including:
  • the rs2032582 reference single nucleotide polymorphisms for which a sequence is shown above can be detected using various oligonucleotides as will be understood by those skilled in the art, including: SNP Oligo sequence
  • wild type refers to a polypeptide or polynucleotide sequence that occurs in a native population without genetic modification.
  • a “variant” includes a polypeptide or polynucleotide sequence having at least one modification to an amino acid or nucleic acid compared to the corresponding amino acid or nucleic acid found in a wild type polypeptide or polynucleotide, respectively. Included in the term variant is Single Nucleotide Polymorphism (SNP) where a single base pair distinction exists in the sequence of a nucleic acid strand compared to the most prevalently found (wild type) nucleic acid strand.
  • SNP Single Nucleotide Polymorphism
  • genetic modification or “genetically modified” refers to, but is not limited to, any suppression, substitution, deletion and/or insertion of one or more bases into DNA sequence(s). Also, as used herein “genetically modified” can refer to a gene encoding a polypeptide or a polypeptide having at least one deletion, substitution or suppression of a nucleic acid or amino acid, respectively.
  • SNPs can be identified by known methods. For example, wild type or SNPs can be identified by DNA amplification and sequencing techniques, DNA and RNA detection techniques, including, but not limited to Northern and Southern blot,
  • WT and mutant polypeptides can be detected by a variety of techniques including, but not limited to immunodiagnostic techniques such as ELISA and western Blot.
  • the process of detecting an allele or polymorphism includes but is not limited to serologic and genetic methods.
  • the allele or polymorphism detected may be functionally involved in affecting an individual's phenotype, or it may be an allele or polymorphism that is in linkage disequilibrium with a functional polymorphism/allele.
  • Polymorphisms/alleles are evidenced in the genomic DNA of a subject, but may also be detectable from RNA, cDNA or protein sequences transcribed or translated from this region, as will be apparent to one skilled in the art.
  • nucleotide and related amino acid sequences obtained from different sources for the same gene may vary both in the numbering scheme and in the precise sequence. Such differences may be due to numbering schemes, inherent sequence variability within the gene, and/or to sequencing errors. Accordingly, reference herein to a particular polymorphic site by number will be understood by those of skill in the art to include those polymorphic sites that correspond in sequence and location within the gene, even where different numbering/nomenclature schemes are used to describe them.
  • genotyping a subject (or DNA or other biological sample) for a polymorphic allele of a gene(s) means detecting which allelic or polymorphic form(s) of the gene(s) or gene expression products (e.g. , hnR A, mR A or protein) are present or absent in a subject (or a sample).
  • Related RNA or protein expressed from such gene may also be used to detect polymorphic variation.
  • an individual may be heterozygous or homozygous for a particular allele. More than two allelic forms may exist, thus there may be more than three possible genotypes.
  • an allele may be 'detected' when other possible allelic variants have been ruled out; e.g., where a specified nucleic acid position is found to be neither adenine (A), thymine (T) or cytosine (C), it can be concluded that guanine (G) is present at that position (i.e., G is 'detected' or 'diagnosed' in a subject).
  • Sequence variations may be detected directly (by, e.g., sequencing) or indirectly (e.g., by restriction fragment length polymorphism analysis, or detection of the hybridization of a probe of known sequence, or reference strand conformation polymorphism), or by using other known methods.
  • a "genetic subset" of a population consists of those members of the population having a particular genotype.
  • a population can potentially be divided into three subsets: homozygous for allele 1 (1,1), heterozygous (1,2), and homozygous for allele 2 (2,2).
  • a 'population' of subjects may be defined using various criteria, e.g., individuals being treated with pazopanib or individuals with cancer.
  • a subject that is "predisposed to” or "at increased risk of a particular phenotypic response based on genotyping will be more likely to display that phenotype than an individual with a different genotype at the target polymorphic locus (or loci).
  • the phenotypic response is based on a multi-allelic polymorphism, or on the genotyping of more than one gene, the relative risk may differ among the multiple possible genotypes.
  • Genetic testing also called genetic screening as used herein refers to the testing of a biological sample from a subject to determine the subject's genotype; and may be utilized to determine if the subject's genotype comprises alleles that either cause, or increase susceptibility to, a particular phenotype (or that are in linkage disequilibrium with allele(s) causing or increasing susceptibility to that phenotype).
  • Linkage disequilibrium refers to the tendency of specific alleles at different genomic locations to occur together more frequently than would be expected by chance. Alleles at given loci are in complete equilibrium if the frequency of any particular set of alleles (or haplotype) is the product of their individual population frequencies. A commonly used measure of linkage disequilibrium is r:
  • n nr 2 has an approximate chi square distribution with 1 degree freedom for biallelic markers. Loci exhibiting an r such that nr 2 is greater than 3.84, corresponding to a significant chi-squared statistic at the 0.05 level, are considered to be in linkage disequilibrium (BS Weir 1996 Genetic Data Analysis II Sinauer Associates, Sunderland, MD).
  • a normalized measure of linkage disequilibrium can be defined as:
  • the value of the D " has a range of -1.0 to 1.0. When statistically significant absolute D " value for two markers is not less than 0.3 they are considered to be in linkage disequilibrium.
  • treatment means any manner in which one or more symptoms associated with the disorder are beneficially altered. Accordingly, the term includes healing or amelioration of a symptom or side effect of the disorder or a decrease in the rate of advancement of the disorder.
  • VEGF vascular endothelial growth factor
  • VEGFR vascular endothelial growth factor receptor
  • VEGFR inhibitor to a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCB1 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCB1 1236), administering to said patient the VEGF or VEGFR inhibitor.
  • a method of prescribing a VEGF or VEGFR inhibitor to a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), prescribing to said patient the VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), administering to said patient a VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof, the patient having been previously genotyped as having a genotype other than the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236), includes administering to the patient a VEGF or VEGFR inhibitor.
  • a method of treating cancer in a patient in need thereof includes administering to the patient a VEGF or VEGFR inhibitor, and then determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236).
  • a method of administering a VEGF or VEGFR inhibitor to a patient in need thereof includes determining whether said patient has the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236) and/or determining whether said patient has has the TT, TA or AA genotype at the rs2032582 reference single nucleotide polymorphism (ABCBl 2667), and if said patient does not have the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCBl 1236) and/or the TT, TA or AA genotype at the rs2032582 reference single nucleotide polymorphism (ABCBl 2667), administering to said patient the VEGF or VEGFR inhibitor.
  • methods for treating a human with cancer comprising:
  • methods further comprising detecting whether said human has one or more of the following variant alleles: IL8 -251T>A and/or ABCBl 2677G>T/A and treating said human with a VEGF or VEGFR inhibitor if said human has 0, 1 or 2 copies of IL8 -251T>A and/or ABCB1 26770T/A.
  • methods for treating a human with cancer comprising:
  • Embodiments of the various aspects of the invention described herein can include discontinuing treatment with the VEGF or VEGFR inhibitor.
  • VEGF or VEGFR inhibitors can be used in the methods of the present invention.
  • the VEGF or VEGFR inhibitor is the compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof:
  • the salt of the compound of formula (I) is a hydrochloride salt.
  • the salt of the compound of formula (I) is a monohydro chloride salt as illustrated by formula ( ⁇ ).
  • the monohydrochloride salt of the compound of formula (I) has the chemical name 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide monohydrochloride.
  • the salt of the compound of formula (I) is a monohydrochloride monohydrate solvate of the compound of formula (I).
  • the monohydrochloride monohydrate solvate of the compound of formula (I) has the chemical name 5-( ⁇ 4-[(2,3-dimethyl-2H-indazol- 6-yl)methylamino]-2-pyrimidinyl ⁇ amino)-2-methylbenzenesulfonamide monohydrochloride monohydrate, as illustrated in formula (I").
  • the free base, salts and solvates of the compound of formula (I) may be prepared, for example, according to the procedures of International Patent Application No. PCT/USO 1/49367 filed December 19, 2001, and published as WO 02/059110 on August 1, 2002, and International Patent Application No. PCT/US03/19211 filed June 17, 2003, and published as WO 03/106416 on December 24, 2003, or according to the methods provided herein.
  • salts may comprise acid addition salts derived from a nitrogen on a substituent in the compound of formula (I).
  • Representative salts include the following salts: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride,
  • the VEGF or VEGFR inhibitor is a monoclonal antibody, such as ranibizumab or bevacizumab.
  • the VEGF or VEGFR inhibitor are multikinase inhibitors for
  • VEGF or VEGF receptors including but not limited to sunitinib, sorafenib, and axitinib.
  • the determination of whether a patient has a particular genotype at a given reference single nucleotide polymorphism includes testing the patient for the particular genotype at the given reference single nucleotide polymorphism.
  • the testing of a patient to determine whether the patient has a particular genotype at a given reference single nucleotide polymorphism can be done by various methods as will be understood by those skilled in the art, for example as described in the Examples section below.
  • the determination of whether a patient has a particular genotype at a given reference single nucleotide polymorphism includes testing the patient for at least one single nucleotide polymorphism that is correlated with the given reference single nucleotide polymorphism.
  • a first reference single nucleotide polymorphism is correlated to a second single nucleotide polymorphism if detection of the first reference single nucleotide
  • polymorphism indicates that the individual would have the second reference single nucleotide polymorphism, or a particular genotype of the second reference single nuclear polymorphism, if the individual were to be tested for the second reference single nucleotide polymorphism or particular genotype thereof.
  • the determination of whether a patient has a particular genotype at a given reference single nucleotide polymorphism includes: a. performing a genotyping technique on a biological sample from the subject to determine whether the subject has a genotype at at least one single nucleotide polymorphism that is correlated with the TT genotype at the rsl 128503 reference single nucleotide polymorphism (ABCB 1 1236);
  • the biological sample is selected from the group consisting of cells, blood, blood components, urine and saliva.
  • embodiments of the invention further provide pharmaceutical compositions, which include therapeutically effective amounts of the VEGF or VEGFR inhibitor, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the carrier(s), diluent(s) or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical formulation including admixing the VEGF or VEGFR inhibitor with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • 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.
  • Capsules are made by preparing a powder mixture as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone
  • a solution retardant such as paraffin
  • a resorption accelerator such as a quaternary salt
  • an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additives such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit formulations for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • Dosage unit forms can also be in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines .
  • VEGF or VEGFR inhibitors can also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled.
  • the compounds may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysme substituted with palmitoyl residues.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986).
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • the formulations are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in- water cream base or a water-in-oil base.
  • Pharmaceutical formulations adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent. Eye-drop formulations are described further herein below.
  • Suitable routes for ocular administration include extraocular and intraocular (including, for example, intra vitreal, subretinal, subscleral, intrachoroidal, and subconjuctival). It will be appreciated that the preferred route may vary with, for example, the condition of the recipient.
  • the pharmaceutical formulations may also be applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • Formulations to be administered to the eye will have ophthalmically compatible pH and osmolality.
  • One or more ophthalmically acceptable pH adjusting agents and/or buffering agents can be included in a composition of the invention, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, and sodium lactate; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids, bases, and buffers can be included in an amount required to maintain pH of the composition in an ophthalmically acceptable range.
  • One or more ophthalmically acceptable salts can be included in the composition in an amount sufficient to bring osmolality of the composition into an
  • Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions.
  • Embodiments of pharmaceutical formulations suitable for ocular administration include the following, Table 1 :
  • formulations are presented as a preservative free, single use eye drop formulations.
  • fill per container is 0.45 mL with a drop weight of ⁇ 40 ⁇
  • density of solution is 1.03 mg/rnL
  • osmolality is -290 mOs m/kg
  • pH is 5.
  • formulations are modified to have a pH down to a value of 4.
  • formulations are modified to have a Captisol concentration up to 10% w/v.
  • the pharmaceutical formulations are adapted for intraocular administration by means of intraocular injection or other device for ocular delivery.
  • ocular devices that may be used in the methods of the invention include periocular or intravitreal devices, contact lenses and liposomes. See, for example, U.S. Pat. Nos. 3,416,530; 3,828,777; 4,014,335; 4,300,557; 4,327,725; 4,853,224; 4,946,450;
  • the ocular delivery device may be designed for the controlled release of one or more therapeutic agents with multiple defined release rates and sustained dose kinetics and permeability. Controlled release may be obtained through the design of polymeric matrices incorporating different choices and properties of biodegradable/bioerodable polymers (e.g. poly(ethylene vinyl) acetate (EVA), superhydrolyzed PVA), hydroxyalkyl cellulose (HPC), methylcellulose (MC), hydroxypropyl methyl cellulose (HPMC), polycaprolactone,
  • EVA poly(ethylene vinyl) acetate
  • HPC hydroxyalkyl cellulose
  • HPMC hydroxypropyl methyl cellulose
  • poly(glycolic) acid poly(lactic) acid, polyanhydride, of polymer molecular weights, polymer crystallinity, copolymer ratios, processing conditions, surface finish, geometry, excipient addition and polymeric coatings that will enhance drug diffusion, erosion, dissolution and osmosis.
  • Formulations for drug delivery using ocular devices may combine one or more active agents and adjuvants appropriate for the indicated route of administration.
  • the active agents may be admixed with any pharmaceutically acceptable excipient, lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, acacia, gelatin, sodium alginate, polyvinylpyrrolidine, and/or polyvinyl alcohol, tableted or encapsulated for conventional administration.
  • the compounds may be dissolved in polyethylene glycol, propylene glycol, carboxymethyl cellulose colloidal solutions, ethanol, corn oil, peanut oil, cottonseed oil, sesame oil, tragacanth gum, and/or various buffers.
  • the compounds may also be mixed with compositions of both biodegradable and non-biodegradable polymers, and a carrier or diluent that has a time delay property.
  • biodegradable compositions can include albumin, gelatin, starch, cellulose, dextrans, polysaccharides, poly (D,L-lactide), poly (D,L-lactide-co-glycolide), poly (glycolide), poly (hydroxybutyrate), poly (alkylcarbonate) and poly (orthoesters) and mixtures thereof.
  • non- biodegradable polymers can include EVA copolymers, silicone rubber and poly
  • compositions for ocular delivery also include in situ gellable aqueous composition.
  • a composition comprises a gelling agent in a concentration effective to promote gelling upon contact with the eye or with lacrimal fluid.
  • Suitable gelling agents include but are not limited to thermosetting polymers.
  • the term "in situ gellable” as used herein is includes not only liquids of low viscosity that form gels upon contact with the eye or with lacrimal fluid, but also includes more viscous liquids such as semi-fluid and thixotropic gels that exhibit substantially increased viscosity or gel stiffness upon administration to the eye. See, for example, Ludwig (2005) Adv. Drug Deliv. Rev. 3;57: 1595-639, herein incorporated by reference for purposes of its teachings of examples of polymers for use in ocular drug delivery.
  • compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • compositions adapted for rectal administration may be presented as suppositories or as enemas.
  • compositions adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
  • Fine particle dusts or mists which may be generated by means of various types of metered, dose pressurised aerosols, nebulizers or insufflators.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • 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 flavouring agents.
  • a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof will depend upon a number of factors including, for example, the age and weight of the animal, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian.
  • an effective amount of a compound of formula (I) or a salt or solvate thereof for the treatment of a cancerous condition such as those described herein will generally be in the range of 0.1 to 100 mg/kg body weight of recipient (mammal) per day and more usually in the range of 1 to 12 mg/kg body weight per day.
  • an effective amount of a salt or solvate thereof can typically be from a lower limit of 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 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, or 400 mg to an upper limit of about 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220,
  • This amount may be given in a single dose per day or in a number (such as two, three, four, five or six) of sub-doses per day such that the total daily dose is the same.
  • An effective amount of a salt or solvate thereof may be determined as a proportion of the effective amount of the compound of formula (I) per se.
  • VEGF or VEGFR inhibitor may be employed alone or in combination with other therapeutic agents for the treatment of the above-mentioned conditions.
  • combination therapies according to the present invention thus comprise the administration of a VEGF or VEGFR inhibitor, and the use of at least one other cancer treatment method, including one or more additional VEGF or VEGFR inhibitors.
  • combination therapies according to the present invention comprise the administration of the VEGF or VEGFR inhibitor, and at least one other pharmaceutically active agent, preferably an anti-neoplastic agent.
  • the VEGF or VEGFR inhibitor and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately this may occur simultaneously or sequentially in any order.
  • the amounts of the VEGF or VEGFR inhibitor and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the VEGF or VEGFR inhibitor and at least one additional cancer treatment therapy may be employed in combination concomitantly or sequentially in any therapeutically appropriate combination with such other anti-cancer therapies.
  • the other anti-cancer therapy is at least one additional chemotherapeutic therapy including administration of at least one anti-neoplastic agent.
  • the administration in combination of a compound of formula (I) or pharmaceutically acceptable salts or solvates thereof with other anti-neoplastic agents may be in combination in accordance with the invention by administration concomitantly in (1) a unitary pharmaceutical composition including both compounds or (2) separate pharmaceutical compositions each including one of the compounds.
  • the combination may be administered separately in a sequential manner wherein one anti-neoplastic agent is administered first and the other second or vice versa. Such sequential administration may be close in time or remote in time.
  • Anti-neoplastic agents may induce anti-neoplastic effects in a cell-cycle specific manner, i.e., are phase specific and act at a specific phase of the cell cycle, or bind DNA and act in a non cell-cycle specific manner, i.e., are non-cell cycle specific and operate by other mechanisms.
  • Anti-neoplastic agents useful in combination with the compound of the VEGF or VEGFR inhibitor can include the following:
  • cell cycle specific anti-neoplastic agents including, but not limited to, diterpenoids such as paclitaxel and its analog docetaxel; vinca alkaloids such as vinblastine, vincristine, vindesine, and vinorelbine; epipodophyllotoxins such as etoposide and teniposide; fluoropyrimidines such as 5-fluorouracil and fluorodeoxyuridine; antimetabolites such as allopurinol, fludurabine, methotrexate, cladrabine, cytarabine, mercaptopurine and thioguanine; and camptothecins such as 9-amino camptothecin, irinotecan, CPT-11 and the various optical forms of 7-(4-methylpiperazino-methylene)-10,l l-ethylenedioxy-20-camptothecin;
  • diterpenoids such as paclitaxel and its analog docetaxel
  • vinca alkaloids such as
  • cytotoxic chemotherapeutic agents including, but not limited to, alkylating agents such as melphalan, chlorambucil, cyclophosphamide, mechlorethamine, hexamethylmelamine, busulfan, carmustine, lomustine, and dacarbazine; anti-tumour antibiotics such as doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dacttinomycin and mithramycin; and platinum coordination complexes such as cisplatin, carboplatin, and oxaliplatin; and
  • anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene
  • progestrogens such as megestrol acetate
  • aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane
  • antiandrogens such as flutamide, nilutamide, bicalutamide, and cyproterone acetate
  • LHRH agonists and antagagonists such as goserelin acetate and luprolide, testosterone 5 a- dihydroreductase inhibitors such as finasteride
  • metalloproteinase inhibitors such as marimastat
  • antiprogestogens urokinase plasminogen activator receptor function inhibitors
  • cyclooxygenase type 2 (COX-2) inhibitors such as celecoxi
  • the VEGF or VEGFR inhibitor can be used to provide additive or synergistic effects with certain existing cancer chemotherapies and radiation, and/or be used to restore effectiveness of certain existing cancer chemotherapies and radiation.
  • the VEGF or VEGFR inhibitor is administered or prescribed in the treatment of disorders mediated by inappropriate VEGF or VEGFR activity.
  • the inappropriate VEGF or VEGFR activity referred to herein is any VEGF or VEGFR activity that deviates from the normal VEGF or VEGFR activity expected in a particular mammalian subject.
  • Inappropriate VEGF or VEGFR activity may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and or control of VEGF or VEGFR activity.
  • Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase or ligand leading to inappropriate or uncontrolled activation of the receptor.
  • unwanted VEGF or VEGFR activity may reside in an abnormal source, such as a malignancy.
  • the level of VEGF or VEGFR activity does not have to be abnormal to be considered inappropriate, rather the activity derives from an abnormal source.
  • the inappropriate angiogenesis referred to herein is any angiogenic activity that deviates from the normal angiogenic activity expected in a particular mammalian subject.
  • Inappropriate angiogenesis may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and or control of angiogenic activity.
  • Such inappropriate activity may result then, for example, from overexpression or mutation of a protein kinase or ligand leading to inappropriate or uncontrolled activation of angiogenesis.
  • unwanted angiogenic activity may reside in an abnormal source, such as a malignancy. That is, the level of angiogenic activity does not have to be abnormal to be considered inappropriate, rather the activity derives from an abnormal source.
  • the disorder is cancer.
  • the cancer is selected from the group consisting of colon cancer, breast cancer, renal cell carcinoma, melanoma, lung cancer including non-small cell lung cancer and adenocarcinoma, gastric cancer, colorectal cancer, neuroendocrine cancer, thyroid cancer, head and neck cancer, brain cancer, cervical cancer, bladder cancer, esophageal cancer, pancreatic cancer, prostate cancer, mesothelioma, liver-hepatobiliary cancer, multiple myeloma, leukemia, thyroid cancer including Hurthle cell, muscle sarcoma (leiomyosarcoma) and bone sarcoma (chonrosarcoma).
  • a further aspect of the present invention provides the use of a VEGF or VEGFR inhibitor, such as the compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for the treatment of a disorder characterized by inappropriate VEGFR2 activity.
  • a VEGF or VEGFR inhibitor such as the compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for the treatment of a disorder characterized by inappropriate VEGFR2 activity.
  • a further aspect of the present invention provides the use of a VEGF or VEGFR inhibitor compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof in the preparation of a medicament for the treatment of cancer and malignant tumours.
  • aspects of the present invention include similar aspects directed to methods of treating neovascular age-related macular degeneration.
  • the neovascular age-related macular degeneration is wet age-related macular degeneration.
  • the age-related macular degeneration is dry age-related macular degeneration and the patient is at increased risk of having age-related macular degeneration progress to wet age- related macular degeneration.
  • the methods according to these aspects of the present invention may also be employed in combination with other methods for the treatment of ocular neovascular disorders.
  • the methods of the invention encompass a combination therapy in which a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof is administered in conjunction with one or more additional therapeutic agents for the treatment of neovascular disorders.
  • additional therapeutic agents include pegaptanib, ranibizumab, bevacizumab, VEGF-TRAP, PKC412, nepafenac, and integrin receptor antagonists (including vitronectin receptor agonists). See, for example, Takahashi et al. (2003) Invest. Ophthalmol. Vis.
  • the therapeutic agents may be administered together or separately.
  • the same means for administration may be used for more than one therapeutic agent of the combination therapy; alternatively, different therapeutic agents of the combination therapy may be administered by different means.
  • the therapeutic agents When the therapeutic agents are administered separately, they may be administered simultaneously or sequentially in any order, both close and remote in time.
  • the amount of administered or prescribed compound according to these aspects of the present invention will depend upon a number of factors including, for example, the age and weight of the patient, the precise condition requiring treatment, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the amount will be at the discretion of the attendant physician.
  • Pharmaceutical formulations for administration to the eye may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • Such a unit may contain, for example, 1 ⁇ g to 1 g, such as 5 ⁇ g to 500 ⁇ g, 10 ⁇ g-250 ⁇ g, 0.5 mg to 700 mg, 2 mg to 350 mg, or 5 mg to 100 mg of a compound of formula (I) or pharmaceutically acceptable salts or solvates thereof depending on the condition being treated, the route of administration and the age, weight and condition of the patient, or pharmaceutical formulations may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • the unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • such pharmaceutical formulations may be prepared by any of the methods well known in the pharmacy art.
  • the compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof is administered or prescribed to be administered one, two, three, four, or more times per day. In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof is administered or prescribed to be administered by administering one, two, three, four or more drops of a suitable pharmaceutical formulation one, two, three, four, or more times per day. In some embodiments of
  • the suitable pharmaceutical formulation comprises between a lower limit of 1, 2, 3, 4, 5, 6, 7, 8, or 9 and an upper limit of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 mg of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof per ml.
  • the first study was a randomized, double-blind, placebo-controlled phase III study, all patients had advanced and/or metastatic renal cell carcinoma (RCC) (hereinafter "first study”). Among 435 participants, 290 patients received pazopanib (800 mg daily) and 145 patients received placebo. A second study was an open-label extension to the first study providing the option for patients who had progressive disease (PD) while on placebo, to receive pazopanib treatment (hereinafter "second study”). As of this data analysis, 79 patients assigned to the placebo arm of the first study participated in the second study and received pazopanib.
  • RCC metastatic renal cell carcinoma
  • Germline DNA was extracted from peripheral blood (QiAamp DNA Blood Kit; Qiagen,
  • the ABCBI polymorphism was genotyped using Illumina GoldenGate genotyping assays (Illumina, San Diego, CA and Shanghai BioChip, Shanghai, China).
  • Each of the following baseline factors was first individually evaluated for association with overall survival (OS) by a univariate Cox model: age, gender, race, body mass index (BMI), MSKCC risk score, Eastern Cooperative Oncology Group (ECOG) performance status, prior nephrectomy status, number of disease sites, time since initial diagnosis, prior therapy status, neutrophil count, platelet count, and study (first or second). All factors significantly associated with OS ( ⁇ 0.05) were further evaluated in a multivariate Cox model. Any baseline factors significant at ⁇ 0.05 in the multivariate Cox model were included as a covariate in PGx analysis. If a covariate violated the proportional hazard assumptions, covariate by survival time interaction was included for evaluation. The median OS in each genotype was estimated using the Kaplan-Meier method.
  • haplotypes with OS was also evaluated for multiple markers in the same gene if at least one of the markers showed nominally significant association with OS ( ⁇ 0.05).
  • Haplotype frequencies were estimated using the R package of haplo. stats (http://mavoresearch.rnayo.edu/schaid lab/software.cfm).
  • SNPs showing nominal significance in single-marker analysis a multivariate analysis including baseline factors and multiple SNPs was applied to evaluate whether they were independent predictive/prognostic factors for OS.
  • This ABCB1 polymorphism was not associated with OS in placebo-arm patients ( Figure 4), although the power of this analysis may be limited because the number of patients receiving placebo who did not cross over to pazoapnib was low.
  • Baseline neutrophil count and body mass index (BMI) were significantly ( ⁇ 0.05) associated with OS (Table 2).
  • HR 0.59
  • CI 95% confidence interval
  • Variables with _P ⁇ 0.05 in the univariate model were selected for evaluation in the multivariate model.
  • Variables with _P ⁇ 0.05 in the multivariate model were included as covariate(s) in the analysis of the effect of each genetic marker.
  • PGx pharmacogenetic
  • BMI body mass index
  • CI confidence interval
  • ECOG Eastern Cooperative
  • the median OS for the pharmacogenetic population was 25 months (95%CI 22-28 months).
  • the variant ABCBl genotypes associated with reduced OS compared with the wild-type genotypes (median, ABCBl 1236TT vs.
  • Multivariate analyses including significant baseline factors and four SNPs (IL8 2767A>T, FGFR2 IVS2 +906OT, NR1I2 -253850T, and ABCBl 12360T) in the model showed these to be independent predictors for OS (Table 4).
  • the second SNP in the IL8 (-251T>A) and ABCBl gene (2677 G>T/A) were not included in the multivariate model because they are strongly correlated with IL8 2767A>T and ABCBl 1236C>T, respectively, therefore are not independent predictors.
  • germline variants in angiogenesis-related eg, IL8, FGFR2
  • exposure -related eg, NR1I2, ABCBl
  • IL8/FGFR2 may therefore serve as a biomarker to identify patients who are more likely to be treatment-resistant and those who will be treatment-responsive. Furthermore, if further studies indicate involvement of these genes in resistance, it is reasonable to consider IL8/FGFR2 as a potential therapeutic target to render tumors durably sensitive to antiangiogenic therapy.
  • Pazopanib is a substrate for P-glycoprotein (data on file), which is encoded by the ABCB1 gene. Therefore polymorphisms in ABCB1 may affect pazopanib absorption and excretion, and consequently, its systemic and cellular (eg, tumor) exposure and effect.
  • van der Veldt et al van der Veldt et al (Janne, et al.
  • NCT00720941 Gaxo SmithKline: Pazopanib versus sunitinib in the treatment of locally advanced and/or metastatic renal cell carcinoma (COMPARZ). Updated July 15, 2010. http://clinicaltrials.gov/ct2/show/NCT00720941) become available.

Abstract

La présente invention concerne des procédés d'administration de pazopanib ou de sels ou de solvates pharmaceutiquement acceptables de celui-ci ainsi que des procédés de traitement du cancer et de la dégénérescence maculaire liée à l'âge chez des patients qui en ont besoin.
PCT/US2012/022316 2011-01-27 2012-01-24 Procédé d'administration et de traitement WO2012103060A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161436686P 2011-01-27 2011-01-27
US61/436,686 2011-01-27
US201161546738P 2011-10-13 2011-10-13
US61/546,738 2011-10-13

Publications (1)

Publication Number Publication Date
WO2012103060A1 true WO2012103060A1 (fr) 2012-08-02

Family

ID=46581133

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/022316 WO2012103060A1 (fr) 2011-01-27 2012-01-24 Procédé d'administration et de traitement

Country Status (1)

Country Link
WO (1) WO2012103060A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015031604A1 (fr) 2013-08-28 2015-03-05 Crown Bioscience, Inc. Signatures d'expression génique permettant de prédire la réponse d'un sujet à un inhibiteur multikinase et leurs procédés d'utilisation
WO2015050504A1 (fr) * 2013-10-03 2015-04-09 Agency For Science, Technology And Research Procédé pour prédire la toxicité et le résultat cliniques
WO2017025928A3 (fr) * 2015-08-12 2017-06-01 Novartis Ag Méthodes de traitement des troubles ophtalmiques
US9968603B2 (en) 2013-03-14 2018-05-15 Forsight Vision4, Inc. Systems for sustained intraocular delivery of low solubility compounds from a port delivery system implant
US10363255B2 (en) 2014-08-08 2019-07-30 Forsight Vision4, Inc. Stable and soluble formulations of receptor tyrosine kinase inhibitors, and methods of preparation thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100184742A1 (en) * 2007-06-12 2010-07-22 Manfred Uhr Polymorphisms in abcb1 associated with a lack of clinical response to medicaments

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100184742A1 (en) * 2007-06-12 2010-07-22 Manfred Uhr Polymorphisms in abcb1 associated with a lack of clinical response to medicaments

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DESAR ET AL.: "Pharmacokinetics of sunitinib in an obese patient with a GIST.", ANNALS ONCOLOGY, vol. 20, no. 3, 19 January 2009 (2009-01-19), pages 599 - 600 *
HENG ET AL.: "Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study.", J CLIN ONCOL, vol. 27, no. 34, 1 December 2009 (2009-12-01), pages 5794 - 5799 *
MARCHETTI ET AL.: "Concise review: Clinical relevance of drug drug and herb drug interactions mediated by the ABC transporter ABCB1 (MDR1, P-glycoprotein).", ONCOLOGIST, vol. 12, no. 8, August 2007 (2007-08-01), pages 927 - 944 *
VAN ERP ET AL.: "Pharmacogenetic pathway analysis for determination of sunitinib-induced toxicity.", J CLIN ONCOL, vol. 27, no. 26, 10 September 2009 (2009-09-10), pages 4406 - 4412 *
WIKIPEDIA.: "Pazopanib", PAZOPANIB, 15 December 2010 (2010-12-15), Retrieved from the Internet <URL:http://en.wikipedia.org/w/index.php?title=Pazopanib&oldid=402519395> [retrieved on 20120306] *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9968603B2 (en) 2013-03-14 2018-05-15 Forsight Vision4, Inc. Systems for sustained intraocular delivery of low solubility compounds from a port delivery system implant
WO2015031604A1 (fr) 2013-08-28 2015-03-05 Crown Bioscience, Inc. Signatures d'expression génique permettant de prédire la réponse d'un sujet à un inhibiteur multikinase et leurs procédés d'utilisation
WO2015050504A1 (fr) * 2013-10-03 2015-04-09 Agency For Science, Technology And Research Procédé pour prédire la toxicité et le résultat cliniques
US10363255B2 (en) 2014-08-08 2019-07-30 Forsight Vision4, Inc. Stable and soluble formulations of receptor tyrosine kinase inhibitors, and methods of preparation thereof
US10765677B2 (en) 2014-08-08 2020-09-08 Forsight Vision4, Inc. Stable and soluble formulations of receptor tyrosine kinase inhibitors, and methods of preparation thereof
WO2017025928A3 (fr) * 2015-08-12 2017-06-01 Novartis Ag Méthodes de traitement des troubles ophtalmiques
US11725246B2 (en) 2015-08-12 2023-08-15 Novartis Ag Methods of treating ophthalmic disorders

Similar Documents

Publication Publication Date Title
US11783366B2 (en) Combination therapies
US20210317534A1 (en) Method for the prognosis and treatment of cancer metastasis
EP3198035B1 (fr) Procédés de prédiction de la réactivité à un médicament
EP2592155B1 (fr) Mutations EGFR
EP1913157B1 (fr) Mutations chez egfr et kras pour predire la reponse d&#39;un patient au traitement avec un inhibiteur d&#39;egfr
US20140221372A1 (en) Method of administration and treatment
JP2019531699A (ja) Nrf2及びその遺伝子の下流標的遺伝子の発現状態及び変異状態によるがんの診断及び治療方法
KR20170132248A (ko) 레티노산 수용체-α 효능제를 이용한 치료에 대해 환자를 계층화하는 방법
EP3260119B1 (fr) Polythérapie pour traiter un cancer
EP2115163A2 (fr) Polymorphismes geniques comme predicteurs specifiques au sexe dans la therapie contre le cancer
US20160032399A1 (en) Method for the Prognosis and Treatment of Renal Cell Carcinoma Metastasis
WO2012103060A1 (fr) Procédé d&#39;administration et de traitement
JP2016144449A (ja) 脈管形成インヒビターに対する応答性
WO2014093750A1 (fr) Méthode d&#39;administration et traitement
US20120232102A1 (en) Methods Of Administration And Treatment
WO2011085288A2 (fr) Procédé d&#39;administration et de traitement
WO2009021683A2 (fr) Marqueur prédictif pour un traitement par un inhibiteur d&#39;egfr
WO2022229846A1 (fr) Traitement du cancer à l&#39;aide d&#39;un inhibiteur du récepteur du facteur de croissance transformant bêta de type 1
CA2695070A1 (fr) Marqueur predictif pour un traitement inhibiteur d&#39;egfr
WO2013172922A1 (fr) Analyse du génotype lmtk3 destinée à être utilisée pour prédire le résultat et la sélection de la thérapie

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: 12739462

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12739462

Country of ref document: EP

Kind code of ref document: A1