US20170000778A1 - Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor - Google Patents

Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor Download PDF

Info

Publication number
US20170000778A1
US20170000778A1 US15/101,155 US201415101155A US2017000778A1 US 20170000778 A1 US20170000778 A1 US 20170000778A1 US 201415101155 A US201415101155 A US 201415101155A US 2017000778 A1 US2017000778 A1 US 2017000778A1
Authority
US
United States
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
patient
formula
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
US15/101,155
Other languages
English (en)
Inventor
Emmanuelle di Tomaso
Marie-Caroline Germa
Cristian Massacesi
Christine Fritsch
Christian Rene Schnell
Ranjana Tavorath
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Individual
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 Individual filed Critical Individual
Assigned to NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC. reassignment NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DI TOMASO, Emmanuelle
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.
Assigned to NOVARTIS PHARMACEUTICALS CORPORATION reassignment NOVARTIS PHARMACEUTICALS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAVORATH, RANJANA
Assigned to NOVARTIS PHARMA S.A.S. reassignment NOVARTIS PHARMA S.A.S. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GERMA, Marie-Caroline, MASSACESI, Cristian
Assigned to NOVARTIS PHARMA AG reassignment NOVARTIS PHARMA AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCHNELL, CHRISTIAN RENE, FRITSCH, CHRISTINE
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMA AG
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMACEUTICALS CORPORATION
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMA S.A.S.
Publication of US20170000778A1 publication Critical patent/US20170000778A1/en
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
    • A61K31/44Non 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to methods of treating or preventing a proliferative disease in a patient in need thereof by orally administering a therapeutically effective amount of an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about two days to about three days between said five-consecutive day cycles; the use of said compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease administered in accordance with said dosage regimen; therapeutic regimen comprising administration of said compound of formula (I) or a pharmaceutically acceptable salt thereof in accordance with said dosage regimen; and related pharmaceutical compositions and packages thereof.
  • Phosphatidylinositol 3-kinases (“PI-3 kinase” or “PI3K”) comprise a family of lipid kinases that catalyze the transfer of phosphate to the D-3′ position of inositol lipids to produce phosphoinositol-3-phosphate (“PIP”), phosphoinositol-3,4-diphosphate (“PIP2”) and phosphoinositol-3,4,5-triphosphate (“PIP3”) that, in turn, act as second messengers in signaling cascades by docking proteins containing pleckstrin-homology, FYVE, Phox and other phospholipid-binding domains into a variety of signaling complexes often at the plasma membrane (Vanhaesebroeck et al., Annu.
  • Human cells contain three genes (PIK3CA, PIK3CB and PIK3CD) encoding the catalytic p110 subunits ( ⁇ , ⁇ , ⁇ isoforms) of class IA PI3K enzymes. These catalytic p110 ⁇ , p110 ⁇ , and p110 ⁇ subunits are constitutively associated with a regulatory subunit that can be p85 ⁇ , p55 ⁇ , p50 ⁇ , p85 ⁇ or p55 ⁇ . p110 ⁇ and p110 ⁇ are expressed in most tissues.
  • Class 1B PI3K has one family member, a heterodimer composed of a catalytic p110 ⁇ subunit associated with one of two regulatory subunits, either the p101 or the p84 (Fruman et al., Annu Rev. Biochem. 67:481 (1998); Suire et al., Curr. Biol. 15:566 (2005)).
  • the modular domains of the p85/55/50 subunits include Src Homology (SH2) domains that bind phosphotyrosine residues in a specific sequence context on activated receptor and cytoplasmic tyrosine kinases, resulting in activation and localization of Class 1A PI3Ks.
  • SH2 Src Homology
  • Class 1B is activated directly by G protein-coupled receptors that bind a diverse repertoire of peptide and non-peptide ligands (Stephens et al., Cell 89:105 (1997)); Katso et al., Annu. Rev. Cell Dev. Biol. 17:615-675 (2001)).
  • PI3K Aberrant regulation of PI3K, which often increases survival through Akt activation, is one of the most prevalent events in human cancer and has been shown to occur at multiple levels.
  • the tumor suppressor gene PTEN which dephosphorylates phosphoinositides at the 3′ position of the inositol ring and in so doing antagonizes PI3K activity, is functionally deleted in a variety of tumors.
  • the genes for the p110 ⁇ isoform, PIK3CA, and for Akt are amplified and increased protein expression of their gene products has been demonstrated in several human cancers. Furthermore, mutations and translocation of p85 ⁇ that serve to up-regulate the p85-p110 complex have been described in human cancers.
  • Deregulation of PI3K is one of the most common deregulations associated with human cancers and proliferative diseases (Parsons et al., Nature 436:792 (2005); Hennessey at el., Nature Rev. Drug Disc. 4:988-1004 (2005)).
  • compound of formula (I) or “Compound A”.
  • compound of formula (I) and pharmaceutically acceptable salts thereof, suitable formulations, and its method of preparation are described in PCT Application WO2010/029082.
  • this alpha-isoform selective PI3K inhibitor compound (S)-Pyrrolidine-1,2-dicarboxylic acid 2-amide 1-( ⁇ 4-methyl-5-[2-(2,2,2-trifluoro-1,1-dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl ⁇ -amide) demonstrated clinical efficacy in the single-agent treatment of patients having advanced solid malignancies carrying an alteration in the PIK3CA gene.
  • the dose escalation phase patients were orally administered this compound either (a) at a dosage ranging from 30 mg to 450 mg once per day (q.d.) on a continuous daily schedule for 28-days, or (b) at a dosage ranging from 120 mg to 200 mg twice per day (b.i.d.) on a continuous daily schedule for 28-days, as guided by Bayesian logistic regression model with overdose control.
  • the dose expansion phase was conducted to additionally treat patients having head and neck cancer with a PIK3CA alteration, patients having solid tumors with PIK3CA alteration, and patients having PIK3CA wildtype ER+/HER2-breast cancer.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I):
  • the present invention relates to a method of treating or preventing a proliferative disease comprising first administering to a patient in need thereof a compound of formula (I) or a pharmaceutically acceptable salt thereof in amount of about 100 mg to about 450 mg daily on a continuous daily schedule via oral administration, second determining said patient has a side effect selected from neutropenia, elevated bilirubin, cardiac toxicity, unstable angina, myocardial infarction, persistent hypertension, peripheral sensory or motor neuropathy/pain, hepatic dysfunction (e.g., liver injury or liver disease, aspartate transaminase level elevation, alanine aminotransferase level elevation, etc.), reduced red and/or white blood cell count, hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mu
  • the present invention relates to a method of reducing at least one side effect selected from neutropenia, elevated bilirubin, cardiac toxicity, unstable angina, myocardial infarction, persistent hypertension, peripheral sensory or motor neuropathy/pain, hepatic dysfunction (e.g., liver injury or liver disease, aspartate transaminase level elevation, alanine aminotransferase level elevation, etc.), reduced red and/or white blood cell count, hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mucositis, pancreatitis, dysgeusia, and dyspepsia from prior treatment with the compound of formula (I) or a pharmaceutically acceptable salt thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I)
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof in a daily dose of about 100 mg to about 450 mg of said compound of formula (I) or a pharmaceutically acceptable salt thereof for at least two five-consecutive day cycles, wherein said medicament is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for treating or preventing a proliferative disease, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a pharmaceutical composition for use in the treatment or prevention of a proliferative disease in a patient in need thereof comprising an amount of about 100 mg to about 450 mg of a compound of formula (I) or pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable excipients, wherein the pharmaceutical composition is orally administered to a patient for at least two five-consecutive day cycles and not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a therapeutic regimen comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to a patient in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a package comprising a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in a daily dose of about 100 mg to about 450 mg together with one or more pharmaceutically acceptable excipients in combination with instructions to orally administer said pharmaceutical composition for at least two five-consecutive day cycles and to not administered said composition for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • a package comprising a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in a daily dose of about 100 mg to about 450 mg together with one or more pharmaceutically acceptable excipients in combination with instructions to orally administer said pharmaceutical composition for at least two five-consecutive day cycles and to not administered said composition for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • FIG. 1 shows concentration-time profiles after oral administration of Compound A at 12.5, 25 and 50 mg/kg qd in nude mice (A) and at 12.5, 25, 40 and 80 mg/kg qd in nude rats (B).
  • FIG. 2 shows observed versus predicted plasma concentrations after oral administration of Compound A at 50 mg/kg qd in nude mice (A) and 40 mg/kg qd in nude rats (B).
  • FIG. 3 shows observed versus predicted plasma concentrations after oral administration of Compound A at 6.25, 12.5, 25 and 50 mg/kg qd in nude mice (A) and at 6.25, 12.5, 25, 40, 50 and 80 mg/kg qd in nude rats (B) on continuous daily schedule.
  • FIGS. 4A and 4B show observed versus predicted plasma concentrations after oral administration of Compound A at 40 mg/kg 2 qd on continuous daily schedule in nude mice in the PK modeling study (A) and the later repeat confirmatory PK modeling study (B).
  • FIG. 5 shows the relationship between tumor tissue concentration and percent S473P-Akt inhibition measured concomitantly in the Rat1-myr P110 ⁇ tumors at different time points post-treatment with Compound A.
  • FIG. 6 shows the relationship between exposure, as measured by time over the in vivo IC80 for S473P-Akt inhibition, and anti-tumor efficacy in Rat1-myr P110 ⁇ tumors treated with Compound A at 50 mg/kg qd.
  • FIG. 7 shows the relationship between the tumor PD marker (pAkt) response and antitumor efficacy observed in mice and rats treated orally qd with various doses of Compound A.
  • FIG. 8 shows observed versus predicted tumor growth inhibition after oral administration of Compound A from 6.25 to 70 mg/kg on continuous daily schedule at various regimen in nude mice and rats.
  • FIG. 9 shows the relationship between plasma Compound A concentrations and plasma insulin levels (A) or blood glucose levels (B) measured in the same probe following Compound A treatment in nude mice.
  • FIG. 10 shows the relationship between plasma Compound A concentrations and plasma insulin levels (A) or blood glucose levels (B) measured in the same probe following Compound A treatment in nude rats.
  • FIG. 11 shows the correlation observed between the fraction of time over plasma hyperglycemia threshold between two consecutive dosing and body weight loss in nude mice and rats.
  • FIG. 12 shows a simulated efficacy curve as determined by the fraction of time above the IC 80 threshold for S473P-Akt and tolerability curve as determined by the duration of exposure above Compound A hyperglycemia threshold in nude mice treated orally qd or 2 qd on continuous daily schedule with increasing doses of Compound A.
  • FIG. 13 shows a simulated efficacy curve as determined by the fraction of time above the IC 80 threshold for S473P-Akt and tolerability curve as determined by the duration of exposure above Compound A hyperglycemia threshold in nude rats treated orally qd or 2 qd with increasing doses of Compound A.
  • FIG. 14 shows a simulated efficacy in Rat1-myr P110 ⁇ tumor bearing nude rats treated orally with Compound A at 20 mg/kg in ALTERNATIVE SCHEDULE 1 (A) or 14 mg/kg qd on continuous daily schedule (B).
  • FIG. 15 shows a simulated plasma PK profiles in nude rats treated orally with Compound A at 20 mg/kg in ALTERNATIVE SCHEDULE 1, as defined in Example 1, or 14 mg/kg qd in continuous daily schedule.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I), as defined herein, or a pharmaceutically acceptable salt thereof to the patient in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about two (2) days to about three (3) days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • a phosphatidylinositol 3-kinase inhibitor or “PI3K inhibitor” is defined herein to refer to a compound which targets, decreases or inhibits activity of the phosphatidylinositol 3-kinase.
  • pharmaceutically acceptable is defined herein to refer to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues a patient without excessive toxicity, irritation allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • treat comprises a treatment or therapeutic regimen relieving, reducing or alleviating at least one symptom in a patient or effecting a delay of progression of a proliferative disorder.
  • treatment can be the diminishment of one or several symptoms of a disorder or complete eradication of a disorder, such as cancer.
  • the term “treat” also denotes to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disorder) and/or reduce the risk of developing or worsening a disorder.
  • prevent comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.
  • clinically effective or “therapeutically effective” is an observable improvement over the baseline clinically observable signs and symptoms of the state, disease or disorder treated with the therapeutic agent.
  • terapéuticaally effective amount is an amount sufficient to provide an observable improvement over the baseline clinically observable signs and symptoms of the state, disease or disorder treated with the therapeutic agent.
  • composition is defined herein to refer to a mixture or solution containing at least one therapeutic agent to be administered to a patient, in order to prevent or treat a particular disease or condition affecting the patient.
  • five-consecutive day cycle means the specified therapeutic agent is administered to the patient during each day for five-consecutive days and then not administered for a period of time before the same therapeutic agent is next administered to the patient. It is understood that the therapeutic agent may be administered each day in a single dosage unit or multiple dosage units and/or administered each day in a single dose (once per day, q.d.) or divided doses (more than once per day, e.g., twice per day, b.i.d.).
  • continuous daily schedule means the therapeutic agent is administered to the patient during each day for at least seven days or for an unspecified period of time or for as long as treatment is necessary. It is understood that the therapeutic agent may be administered each day in a single dosage unit or multiple dosage units and/or administered each day in a single dose (once per day, q.d.) or divided doses (more than once per day, e.g., twice per day, b.i.d.).
  • day refers to either one calendar day or one 24-hour period.
  • the term “combination” is used herein to refer to either a fixed combination in one dosage unit form, a non-fixed combination or a kit of parts for the combined administration where the compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent may be administered simultaneously, independently at the same time or separately within time intervals that allow that the combination partners show a cooperative, e.g., synergistic, effect.
  • the term “fixed combination” means that the therapeutic agents, e.g. the compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one additional therapeutic agent, are both administered to a patient simultaneously in the form of a single entity or dosage unit.
  • non-fixed combination or “kit of parts” means that the therapeutic agents, e.g.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one additional therapeutic agent are both administered to a patient as separate entities or dosage units either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two therapeutic agents in the body of the patient.
  • cocktail therapy e.g. the administration of three or more therapeutic agents.
  • combined administration is defined to encompass the administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • patient is intended to include animals.
  • subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals.
  • the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from a brain tumor disease.
  • the patient or warm-blooded animal is human.
  • WO2010/029082 describes specific 2-carboxamide cycloamino urea derivatives, which have been found to have highly selective inhibitory activity for the alpha-isoform of phosphatidylinositol 3-kinase (PI3K).
  • PI3K phosphatidylinositol 3-kinase
  • the alpha-isoform selective PI3K inhibitor suitable for the present invention is a compound having the following formula (I):
  • compound of formula (I) or “Compound A” or pharmaceutically acceptable salts thereof.
  • the compound of formula (I) is also known as the chemical compound (S)-Pyrrolidine-1, 2-dicarboxylic acid 2-amide 1-( ⁇ 4-methyl-5-[2-(2,2,2-trifluoro-1,1-dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl ⁇ -amide).
  • the compound of formula (I), its pharmaceutically acceptable salts and suitable formulations are described in PCT Application No. WO2010/029082, which is hereby incorporated by reference in its entirety, and methods of its preparation have been described, for example, in Example 15 therein.
  • salts can be present alone or in mixture with free compound of formula (I) and are preferably pharmaceutically acceptable salts.
  • Such salts are formed, for example, as acid addition salts, preferably with organic or inorganic acids, from the compound of formula (I) with a basic nitrogen atom, especially the pharmaceutically acceptable salts.
  • Suitable in-organic acids are, for example, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid.
  • Suitable organic acids are, e.g., carboxylic acids or sulfonic acids, such as fumaric acid or methansulfonic acid.
  • any reference to the free compound hereinbefore and hereinafter is to be understood as referring also to the corresponding salts, as appropriate and expedient.
  • the salts of compound of the formula (I) are preferably pharmaceutically acceptable salts; suitable counter-ions forming pharmaceutically acceptable salts are known in the field.
  • the compound of formula (I) has been previously demonstrated to potently and selectively inhibit the alpha-isoform of the PI3K, including for example, Examples A and C of PCT Application No. WO2010/029082.
  • the compound of formula (I) inhibits the alpha-isoform of PI3K (IC 50 of 0.008 ⁇ mol/L) more potently than the beta-isoform (IC 50 of 1.212 ⁇ mol/L), delta-isoform (IC 50 of 0.077 ⁇ mol/L), and gamma-isoform (IC 50 of 1.097 ⁇ mol/L) in cellular assays and lacks inhibitory activity against the Vps34, mTOR, DNA-PK and ATR.
  • the compound of formula (I) shows inhibitory activity against the wildtype alpha-isoform of PI3K, E545K mutant alpha-isoform of PI3K, and H1047R mutant alpha-isoform of PI3K.
  • the compound of formula (I) or its pharmaceutically acceptable salts may be orally administered at a dosage of about 100 mg to about 450 mg per day to a human patient in need thereof.
  • the term “daily dose” refers to the total dosage amount of the therapeutic agent administered to a specific patient in any single day.
  • the compound of formula (I) may be administered to patient at a daily dose of about 200 to about 400 mg per day, or about 240 mg to about 400 mg per day, or about 300 mg to about 400 mg per day, or about 350 mg to about 400 mg per day.
  • the compound of formula (I) is administered to a human patient at a daily dose of about 350 mg to about 400 mg per day.
  • the daily dose may be administered to the patient in single dose (once per day, q.d.) or divided doses (more than once per day, e.g., twice per day, b.i.d.). In one embodiment, the daily dose is administered in a once per day (q.d.). In a further embodiment, the daily dose is administered twice per day (b.i.d.)
  • the daily dose may be administered to the patient in a single dosage unit or amounts of multiple dosage units to make up the daily dose.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the compound or a pharmaceutically acceptable salt thereof is not administered for about 2 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof once per day (q.d.) at a daily dose of about 100 mg to about 450 mg, preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof twice per day (b.i.d.) at a daily dose of about 100 mg to about 450 mg, preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • dosage regimen of the present invention may be alternatively defined relative to timing of the actual administrations of the compound of formula (I) or its pharmaceutically acceptable salt.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof once per day (q.d.) at a daily dose of about 100 mg to about 450 mg, preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 3 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof twice per day (b.i.d.) at a daily dose of about 100 mg to about 450 mg, preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered for a period of about 2.5 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • Proliferative diseases that may be treated or prevented by the administration of the compound of formula (I) or a pharmaceutically acceptable in accordance with the dosage regimen of the present invention are particularly those mediated by the alpha-isoform of the PI3K. It is understood that one embodiment of the present invention includes the treatment of the proliferative disease and that a further embodiment of the present invention includes the prevention of the proliferative disease.
  • proliferative diseases which may be treated or prevented in accordance with the present invention include, cancer, polycythemia vera, essential thrombocythemia, myelofibrosis with myeloid metaplasia, asthma, COPD, ARDS, Loffler's syndrome, eosinophilic pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma, eosinophil-related disorders affecting the airways occasioned by drug-reaction, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforme, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoi
  • haemolytic anaemia haemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia
  • systemic lupus erythematosus polychondritis, scleroderma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven-Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (e.g.
  • endocrine opthalmopathy Grave's disease, sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, primary biliary cirrhosis, uveitis (anterior and posterior), interstitial lung fibrosis, psoriatic arthritis, glomerulonephritis, cardiovascular diseases, atherosclerosis, hypertension, deep venous thrombosis, stroke, myocardial infarction, unstable angina, thromboembolism, pulmonary embolism, thrombolytic diseases, acute arterial ischemia, peripheral thrombotic occlusions, and coronary artery disease, reperfusion injuries, retinopathy, such as diabetic retinopathy or hyperbaric oxygen-induced retinopathy, and conditions characterized by elevated intraocular pressure or secretion of ocular aqueous humor, such as glaucoma.
  • Grave's disease sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis,
  • the proliferative disease is a cancer.
  • cancer refers to tumors and/or cancerous cell growth preferably mediated by the alpha-isoform of the PI3K.
  • the compounds are useful in the treatment of cancers including, for example, sarcoma, lung, bronchus, prostate, breast (including sporadic breast cancers and sufferers of Cowden disease), pancreas, gastrointestine, colon, rectum, colon carcinoma, colorectal adenoma, thyroid, liver, intrahepatic bile duct, hepatocellular, adrenal gland, stomach, gastric, glioma, glioblastoma, endometrial, melanoma, kidney, renal pelvis, urinary bladder, uterine corpus, uterine cervix, vagina, ovary, multiple myeloma, esophagus, a leukemia, acute myelogenous leukemia, chronic myelogenous leukemia
  • Proliferative diseases mediated by the alpha-subunit of PI3K may include those showing overexpression or amplification of PI3K alpha, somatic mutation of PIK3CA or germline mutations or somatic mutation of PTEN or mutations and translocation of p85 ⁇ that serve to up-regulate the p85-p110 complex.
  • the cancer is a tumor and/or cancerous growth mediated by the alpha isoform of PI3K.
  • the proliferative disease is a cancer selected from a cancer of the lung, bronchus, prostate, breast (including sporadic breast cancers and sufferers of Cowden disease), colon, rectum, colon carcinoma, colorectal adenoma, pancreas, gastrointestine, hepatocellular, stomach, gastric, ovary, squamous cell carcinoma, and head and neck.
  • the proliferative disease is a cancer selected from a cancer of the breast, colon, rectum, colon carcinoma, colorectal adenoma, endometrial, and cervical.
  • the proliferative disease is a cancer selected from a cancer of the lung, breast (including sporadic breast cancers and sufferers of Cowden disease), gastric, ovary and head and neck.
  • the present invention relates to the treatment of a cancer by the administration of the compound of formula (I) or a pharmaceutically acceptable in accordance with the dosage regimen of the present invention.
  • Examples of such side effects which may relieved, reduced, or alleviated by the dosage regimen of the present invention include, but are not limited to, neutropenia, elevated bilirubin, cardiac toxicity, unstable angina, myocardial infarction, persistent hypertension, peripheral sensory or motor neuropathy/pain, hepatic dysfunction (e.g., liver injury or liver disease, aspartate transaminase level elevation, alanine aminotransferase level elevation, etc.), reduced red and/or white blood cell count, hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mucositis, pancreatitis, dysgeusia, and dyspepsia.
  • neutropenia e.g., elevated bilirubin
  • cardiac toxicity e.g., unstable an
  • the side effect relieved, reduced, or alleviated by the dosage regimen of the present invention is a condition selected from hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mucositis, dysgeusia, and dyspepsia. More preferably, the side effect relieved, reduced, or alleviated by the dosage regimen of the present invention is hyperglycemia.
  • the dosage regimen of the present invention results in the beneficial effects described herein before.
  • the person skilled in the art is fully enabled to select a relevant test model to prove such beneficial effects.
  • the pharmacological activity of the compound of formula (I) or its pharmaceutically acceptable salt may, for example, be demonstrated in a clinical study, an animal study or in a test procedure as essentially described hereinafter.
  • Suitable clinical studies are in particular, for example, open label, dose escalation studies in patients with a proliferative disease, including for example a tumor disease, e.g., breast cancer, wherein said patients are orally administered the compound of formula (I) in accordance with the dosage regimen of the present invention.
  • patients are assigned to different groups wherein at least one group is administered the compound of formula (I) on a continuous daily schedule and at least one group is administered the compound of formula (I) in accordance with the dosage regimen of the present invention.
  • Such studies prove in particular the efficacy of the therapeutic agent and its impact on existing or potential side effects.
  • the beneficial effects on a proliferative disease may be determined directly through the results of these studies which are known as such to a person skilled in the art.
  • Such studies may be, in particular, suitable to compare the effects of a continuous daily schedule using the therapeutic agents and the dosing schedule of the present invention.
  • Each patient may receive doses of the compound of formula (I) or its pharmaceutically acceptable salt either once per day or more than once (e.g., twice) per day.
  • the efficacy of the treatment may be determined in such studies, e.g., after 12, 18 or 24 weeks by evaluation of symptom scores and/or tumor size measurements every 6 weeks.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is preferably used or administered in the form of pharmaceutically compositions that contain a therapeutically effective amount of the compound of formula (I) or pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable excipients suitable for oral administration.
  • the pharmaceutical composition may comprise an amount of about 100 mg to about 450 mg of a compound of formula (I) or pharmaceutically acceptable salt thereof to be administered in a single dosage unit.
  • the pharmaceutical composition may comprise an amount of the compound of formula (I) or pharmaceutically acceptable salt thereof which is subdivided into multiple dosage units and administered for a daily dosage of about 100 mg to about 450 mg of the compound of formula (I) or pharmaceutically acceptable salt thereof.
  • compositions used according to the present invention can be prepared in a manner known per se to be suitable for oral administration to mammals (warm-blooded animals), including humans.
  • Pharmaceutical compositions for oral administration may include, for example, those in dosage unit forms, such as sugar-coated tablets, tablets, capsules, sachets and furthermore ampoules. If not indicated otherwise, these are prepared in a manner known per se, for example by means of conventional mixing, granulating, sugar-coating, dissolving or lyophilizing processes. It will be appreciated that the amount of the active ingredient contained in an individual dose or dosage unit need not in itself constitute a therapeutically effective amount since the necessary effective amount can be reached by administration of a plurality of dosage units.
  • the novel pharmaceutical composition may contain, for example, from about 10% to about 100%, preferably from about 20% to about 60%, of the active ingredient.
  • any of the usual pharmaceutically acceptable excipients may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents; or excipients such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed.
  • disintegrants examples include, but are not limited to, starches; clays; celluloses; alginates; gums; cross-linked polymers, e.g., cross-linked polyvinyl pyrrolidone or crospovidone, e.g., POLYPLASDONE XL from International Specialty Products (Wayne, N.J.); cross-linked sodium carboxymethylcellulose or croscarmellose sodium, e.g., AC-DI-SOL from FMC; and cross-linked calcium carboxymethylcellulose; soy polysaccharides; and guar gum.
  • the disintegrant may be present in an amount from about 0% to about 10% by weight of the composition. In one embodiment, the disintegrant is present in an amount from about 0.1% to about 5% by weight of composition.
  • binders examples include, but are not limited to, starches; celluloses and derivatives thereof, for example, microcrystalline cellulose, e.g., AVICEL PH from FMC (Philadelphia, Pa.), hydroxypropyl cellulose hydroxylethyl cellulose and hydroxylpropylmethyl cellulose METHOCEL from Dow Chemical Corp. (Midland, Mich.); sucrose; dextrose; corn syrup; polysaccharides; and gelatin.
  • the binder may be present in an amount from about 0% to about 50%, e.g., 2-20% by weight of the composition.
  • Examples of pharmaceutically acceptable lubricants and pharmaceutically acceptable glidants include, but are not limited to, colloidal silica, magnesium trisilicate, starches, talc, tribasic calcium phosphate, magnesium stearate, aluminum stearate, calcium stearate, magnesium carbonate, magnesium oxide, polyethylene glycol, powdered cellulose and microcrystalline cellulose.
  • the lubricant may be present in an amount from about 0% to about 10% by weight of the composition. In one embodiment, the lubricant may be present in an amount from about 0.1% to about 1.5% by weight of composition.
  • the glidant may be present in an amount from about 0.1% to about 10% by weight.
  • Examples of pharmaceutically acceptable fillers and pharmaceutically acceptable diluents include, but are not limited to, confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, lactose, mannitol, microcrystalline cellulose, powdered cellulose, sorbitol, sucrose and talc.
  • the filler and/or diluent e.g., may be present in an amount from about 0% to about 80% by weight of the composition.
  • a dosage unit form containing the compound of formula (I) or a pharmaceutically acceptable salt thereof may be in the form of micro-tablets enclosed inside a capsule, e.g. a gelatin capsule.
  • a gelatin capsule as is employed in pharmaceutical formulations can be used, such as the hard gelatin capsule known as CAPSUGEL, available from Pfizer.
  • disintegrants examples include, but are not limited to, starches; clays; celluloses; alginates; gums; cross-linked polymers, e.g., cross-linked polyvinyl pyrrolidone or crospovidone, e.g., POLYPLASDONE XL from International Specialty Products (Wayne, N.J.); cross-linked sodium carboxymethylcellulose or croscarmellose sodium, e.g., AC-DI-SOL from FMC; and cross-linked calcium carboxymethylcellulose; soy polysaccharides; and guar gum.
  • the disintegrant may be present in an amount from about 0% to about 10% by weight of the composition. In one embodiment, the disintegrant is present in an amount from about 0.1% to about 5% by weight of composition.
  • binders examples include, but are not limited to, starches; celluloses and derivatives thereof, for example, microcrystalline cellulose, e.g., AVICEL PH from FMC (Philadelphia, Pa.), hydroxypropyl cellulose hydroxylethyl cellulose and hydroxylpropylmethyl cellulose METHOCEL from Dow Chemical Corp. (Midland, Mich.); sucrose; dextrose; corn syrup; polysaccharides; and gelatin.
  • the binder may be present in an amount from about 0% to about 50%, e.g., 2-20% by weight of the composition.
  • Examples of pharmaceutically acceptable lubricants and pharmaceutically acceptable glidants include, but are not limited to, colloidal silica, magnesium trisilicate, starches, talc, tribasic calcium phosphate, magnesium stearate, aluminum stearate, calcium stearate, magnesium carbonate, magnesium oxide, polyethylene glycol, powdered cellulose, Sodium stearyl fumarate and microcrystalline cellulose.
  • the lubricant may be present in an amount from about 0% to about 10% by weight of the composition. In one embodiment, the lubricant may be present in an amount from about 0.1% to about 1.5% by weight of composition.
  • the glidant may be present in an amount from about 0.1% to about 10% by weight.
  • Examples of pharmaceutically acceptable fillers and pharmaceutically acceptable diluents include, but are not limited to, confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, lactose, mannitol, microcrystalline cellulose, powdered cellulose, sorbitol, sucrose and talc.
  • the filler and/or diluent e.g., may be present in an amount from about 0% to about 80% by weight of the composition.
  • the present invention relates to a pharmaceutical composition for use in the treatment or prevention of a proliferative disease in a patient in need thereof comprising an amount of about 100 mg to about 450 mg of a compound of formula (I) or pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable excipients, wherein the pharmaceutical composition is orally administered to a patient for at least two five-consecutive day cycles and not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient in a daily dose of about 100 mg to about 450 mg, preferably about 200 mg to about 400 mg or more preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the compound or a pharmaceutically acceptable salt thereof is not administered for about 2 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient once per day (q.d.) at a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient twice per day (b.i.d.) at a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient once per day (q.d.) at a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 3 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in a patient in need thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to the patient twice per day (b.i.d.) at a daily dose of about 100 mg to about 450 mg, preferably about 350 mg to about 400 mg, for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered for a period of about 2.5 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating or preventing a proliferative disease in accordance with the dosage regimen herein, wherein the compound of formula (I) or its pharmaceutically acceptable salt thereof is administered in two or more of said five-consecutive day cycles until the relief, reduction, or alleviation of the severity, occurrence rate, or frequency of at least one side effect in said patient.
  • the present invention relates to a method of treating or preventing a proliferative disease in accordance with the dosage regimen herein, wherein the compound of formula (I) or its pharmaceutically acceptable salt thereof is administered in two or more of said five-consecutive day cycle until the progression of the disease.
  • the present invention relates to a method of treating or preventing a proliferative disease comprising first administering to a patient in need thereof a compound of formula (I) or a pharmaceutically acceptable salt thereof in amount of about 100 mg to about 450 mg daily on a continuous daily schedule via oral administration, second determining said patient has a side effect selected from neutropenia, elevated bilirubin, cardiac toxicity, unstable angina, myocardial infarction, persistent hypertension, peripheral sensory or motor neuropathy/pain, hepatic dysfunction (e.g., liver injury or liver disease, aspartate transaminase level elevation, alanine aminotransferase level elevation, etc.), reduced red and/or white blood cell count, hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mu
  • the present invention relates to a method of reducing at least one side effect selected from neutropenia, elevated bilirubin, cardiac toxicity, unstable angina, myocardial infarction, persistent hypertension, peripheral sensory or motor neuropathy/pain, hepatic dysfunction (e.g., liver injury or liver disease, aspartate transaminase level elevation, alanine aminotransferase level elevation, etc.), reduced red and/or white blood cell count, hyperglycemia, nausea, decreased appetite, diarrhea, rash (e.g, maculopapular, acneiform, etc.) and hypersensitivity (e.g., increased sensitivity to bruise), photosensitivity, asthenia/fatigue, vomiting, stomatitis, oral mucositis, pancreatitis, dysgeusia, and dyspepsia from prior treatment with the compound of formula (I) or a pharmaceutically acceptable salt thereof, comprising orally administering a therapeutically effective amount of the compound of formula (I)
  • the present invention includes a method of treating or preventing a proliferative disorder in accordance with any other embodiment disclosed above for the present invention.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof in a daily dose of about 100 mg to about 450 mg, preferably about 200 mg to about 400 mg or more preferably about 350 mg to about 400 mg, of said compound of formula (I) or a pharmaceutically acceptable salt thereof for at least two five-consecutive day cycles, wherein said medicament is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof once per day (q.d.) in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof twice per day (b.i.d.) in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof once per day (q.d.) in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 3 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered to a patient in need thereof twice per day (b.i.d.) in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound or a pharmaceutically acceptable salt thereof is not administered for a period of about 2.5 days between the last administration of said compound or a pharmaceutically acceptable salt thereof in one five-consecutive day cycle and the first administration of said compound or a pharmaceutically acceptable salt thereof in its subsequent five-consecutive day cycle.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered in accordance with the dosage regimen herein, wherein the compound of formula (I) or its pharmaceutically acceptable salt thereof is administered in two or more of said five-consecutive day cycles until the relief, reduction, or alleviation of the severity, occurrence rate, or frequency of at least one side effect in said patient.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is orally administered in accordance with the dosage regimen herein, wherein the compound of formula (I) or its pharmaceutically acceptable salt thereof is administered in two or more of said five-consecutive day cycles until the progression of the disease.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, wherein said medicament is first orally administered in amount of about 100 mg to about 450 mg daily dose on a continuous daily schedule and subsequently reduced to an administered amount of about 100 mg to about 450 mg daily dose for at least two five-consecutive day cycles via oral administration, wherein said compound or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention includes any use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease in accordance with the methods of treatment or any embodiment disclosed above for the present invention.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for treating or preventing a proliferative disease, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered to a patient in need thereof in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention includes any use of the compound of formula (I) or a pharmaceutically acceptable salt thereof in accordance with the methods of treatment, uses for the manufacture of a medicament, or any embodiment disclosed above for the present invention.
  • the present invention further relates to a therapeutic regimen comprising orally administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof to a patient in need thereof in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention further relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof administered in combination with at least one additional therapeutic agent for the treatment or prevention of a proliferative disease, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • Suitable therapeutic agents for use in accordance with the present invention include, but are not limited to, kinase inhibitors, anti-estrogens, anti androgens, other inhibitors, cancer chemotherapeutic drugs, alkylating agents, chelating agents, biological response modifiers, cancer vaccines, agents for antisense therapy. Examples are set forth below:
  • EGFR Epidermal Growth Factor Receptor
  • Vascular Endothelial Growth Factor Receptor (VEGFR) kinase inhibitors including SU-11248 (WO 01/60814), SU 5416 (U.S. Pat. No. 5,883,113 and WO 99/61422), SU 6668 (U.S. Pat. No. 5,883,113 and WO 99/61422), CHIR-258 (U.S. Pat. No. 6,605,617 and U.S. Pat. No. 6,774,237), vatalanib or PTK-787 (U.S. Pat. No.
  • VEGFR Vascular Endothelial Growth Factor Receptor
  • VEGF-Trap WO 02/57423
  • B43-Genistein WO-09606116
  • fenretinide retinoic acid p-hydroxyphenylamine
  • IM-862 WO 02/62826
  • bevacizumab or Avastin® WO 94/10202
  • KRN-951 3-[5-(methylsulfonylpiperadine methyl)-indolyl]-quinolone, AG-13736 and AG-13925, pyrrolo[2,1-f][1,2,4]triazines, ZK-304709, Veglin®, VMDA-3601, EG-004, CEP-701 (U.S.
  • Estrogen-targeting agents include Selective Estrogen Receptor Modulators (SERMs) including tamoxifen, toremifene, raloxifene; aromatase inhibitors including Arimidex® or anastrozole; Estrogen Receptor Downregulators (ERDs) including Faslodex® or fulvestrant.
  • SERMs Selective Estrogen Receptor Modulators
  • ESDs Estrogen Receptor Downregulators
  • Anti-Androgens include flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids.
  • Other Inhibitors including Protein farnesyl transferase inhibitors including tipifarnib or R-115777 (US 2003134846 and WO 97/21701), BMS-214662, AZD-3409, and FTI-277; topoisomerase inhibitors including merbarone and diflomotecan (BN-80915); mitotic kinesin spindle protein (KSP) inhibitors including SB-743921 and MKI-833; proteasome modulators such as bortezomib or Velcade® (U.S. Pat. No. 5,780,454), XL-784; cyclooxygenase 2 (COX-2) inhibitors including non-steroidal antiinflammatory drugs I (NSAIDs); letrozole; exemestane; and eribulin.
  • KSP mitotic kinesin spindle protein
  • COX-2 cyclooxygenase 2
  • E. Cancer Chemotherapeutic Drugs including anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), dactinomycin (Act
  • Alkylating Agents including VNP-40101M or cloretizine, oxaliplatin (U.S. Pat. No. 4,169,846, WO 03/24978 and WO 03/04505), glufosfamide, mafosfamide, etopophos (U.S. Pat. No.
  • Chelating Agents including tetrathiomolybdate (WO 01/60814); RP-697; Chimeric T84.66 (cT84.66); gadofosveset (Vasovist®); deferoxamine; and bleomycin optionally in combination with electorporation (EPT).
  • H. Biological Response Modifiers such as immune modulators, including staurosprine and macrocyclic analogs thereof, including UCN-01, CEP-701 and midostaurin (see WO 02/30941, WO 97/07081, WO 89/07105, U.S. Pat. No. 5,621,100, WO 93/07153, WO 01/04125, WO 02/30941, WO 93/08809, WO 94/06799, WO 00/27422, WO 96/13506 and WO 88/07045); squalamine (WO 01/79255); DA-9601 (WO 98/04541 and U.S. Pat. No.
  • alemtuzumab interferons (e.g. IFN-a, IFN-b etc.); interleukins, specifically IL-2 or aldesleukin as well as IL-1, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, and active biological variants thereof having amino acid sequences greater than 70% of the native human sequence; altretamine (Hexalen®); SU 101 or leflunomide (WO 04/06834 and U.S. Pat. No. 6,331,555); imidazoquinolines such as resiquimod and imiquimod (U.S. Pat. Nos.
  • Anticancer vaccines including Avicine® (Tetrahedron Lett. 26:2269-70 (1974)); oregovomab (OvaRex®); Theratope® (STn-KLH); Melanoma Vaccines; GI-4000 series (GI-4014, GI-4015, and GI-4016), which are directed to five mutations in the Ras protein; GlioVax-1; MelaVax; Advexin® or INGN-201 (WO 95/12660); Sig/E7/LAMP-1, encoding HPV-16 E7; MAGE-3 Vaccine or M3TK (WO 94/05304); HER-2VAX; ACTIVE, which stimulates T-cells specific for tumors; GM-CSF cancer vaccine; and Listeria monocytogenes -based vaccines.
  • Avicine® Tetrahedron Lett. 26:2269-70 (1974)
  • oregovomab OvaRex®
  • Theratope® STn-K
  • Anticancer agents including antisense compositions, such as AEG-35156 (GEM-640); AP-12009 and AP-11014 (TGF-beta2-specific antisense oligonucleotides); AVI-4126; AVI-4557; AVI-4472; oblimersen (Genasense®); JFS2; aprinocarsen (WO 97/29780); GTI-2040 (R2 ribonucleotide reductase mRNA antisense oligo) (WO 98/05769); GTI-2501 (WO 98/05769); liposome-encapsulated c-Raf antisense oligodeoxynucleotides (LErafAON) (WO 98/43095); and Sirna-027 (RNAi-based therapeutic targeting VEGFR-1 mRNA).
  • AEG-35156 GEM-640
  • AP-12009 and AP-11014 TGF-beta2-specific antisense
  • the additional therapeutic agent is selected from gefinitib, erlotinib, bevacizumab or Avastin®, pertuzumab, trastuzumab, MEK162, tamoxifen, fulvestrant, capecitabine, cisplatin, carboplatin, cetuximab, paclitaxel, temozolamide, letrozole, or exemestane.
  • the compound of formula (I) and the additional therapeutic agent may be administered together in a single pharmaceutical composition, separately in two or more separate unit dosage forms, or sequentially.
  • the pharmaceutical composition or dosage unit form comprising the additional therapeutic agent may be prepared in a manner known per se and are those suitable for enteral, such as oral or rectal, topical, and parenteral administration to subjects, including mammals (warm-blooded animals) such as humans.
  • a therapeutically effective amount of each of the therapeutic agents may be administered simultaneously or sequentially and in any order, and the components may be administered separately or as a fixed combination.
  • the combination of the present invention may comprise: (i) administration of the first therapeutic agent (a) in free or pharmaceutically acceptable salt form; and (ii) administration of an therapeutic agent (b) in free or pharmaceutically acceptable salt form, simultaneously or sequentially in any order, in jointly therapeutically effective amounts, preferably in synergistically effective amounts, e.g., in daily or intermittent dosages corresponding to the amounts described herein.
  • the individual therapeutic agents of the combination may be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
  • “Synergy” or “synergistic” refers to the action of two therapeutic agents such as, for example, (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof and (b) an aromatase inhibitor, producing an effect, for example, slowing the symptomatic progression of a cancer disease or disorder, particularly cancer, or symptoms thereof, which is greater than the simple addition of the effects of each therapeutic agent administered by themselves.
  • a synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453 (1981)), the equation of Loewe additivity (Loewe, S.
  • each of therapeutic agent (a) or therapeutic agent (b) employed in the combination may vary depending on the particular compound or pharmaceutical composition employed, the mode of administration, the condition being treated, the severity of the condition being treated.
  • the dosage regimen of the combination is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed.
  • a physician, clinician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the therapeutic agent required to prevent, counter or arrest the progress of the condition.
  • Optimal precision in achieving concentration of therapeutic agent within the range that yields efficacy requires a regimen based on the kinetics of the therapeutic agent's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a therapeutic agent.
  • proliferative diseases that may be treated with a combination of a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one additional therapeutic agent include, but not limited to, those set forth above.
  • Suitable clinical studies are in particular, for example, open label, dose escalation studies in patients with a proliferative disease, including for example a tumor disease, e.g., breast cancer. Such studies prove in particular the synergism of the therapeutic agents of the combination of the present invention.
  • the beneficial effects on a proliferative disease may be determined directly through the results of these studies which are known as such to a person skilled in the art.
  • Such studies may be, in particular, suitable to compare the effects of a monotherapy using the therapeutic agents and a combination of the present invention.
  • the dose of the alpha-isoform selective PI3K inhibitor compound of formula (I) or its pharmaceutically acceptable salt is escalated until the Maximum Tolerated Dosage is reached, and the combination partner is administered with a fixed dose.
  • the compound of formula (I) or its pharmaceutically acceptable salt may be administered in a fixed dose and the dose of the combination partner may be escalated.
  • Each patient may receive doses of the compound of formula (I) or its pharmaceutically acceptable salt either once per day or more than once (e.g., twice) per day.
  • the efficacy of the treatment may be determined in such studies, e.g., after 12, 18 or 24 weeks by evaluation of symptom scores every 6 weeks.
  • the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 100 mg to about 450 mg for at least two five-consecutive day cycles, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is not administered to the patient for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • the present invention relates to a method of treating a treating or preventing a proliferative disease by administration in accordance with the dosage regimen of the present invention, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in combination with at least one additional therapeutic agent.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease in accordance with the dosage regimen of the present invention, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in combination with at least one additional therapeutic agent.
  • the present invention relates to the use of the compound of formula (I) or a pharmaceutically acceptable salt thereof for treating or preventing a proliferative disease in accordance with the dosage regimen of the present invention, wherein said compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in combination with at least one additional therapeutic agent.
  • the present invention further relates to a package comprising a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in a daily dose of about 100 mg to about 450 mg together with one or more pharmaceutically acceptable excipients in combination with instructions to orally administer said pharmaceutical composition for at least two five-consecutive day cycles and to not administered said composition for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • a package comprising a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in a daily dose of about 100 mg to about 450 mg together with one or more pharmaceutically acceptable excipients in combination with instructions to orally administer said pharmaceutical composition for at least two five-consecutive day cycles and to not administered said composition for a period of about 2 days to about 3 days between one five-consecutive day cycle and its subsequent five-consecutive day cycle.
  • Utility of the dosage regimen of the compounds of formula (I) of the present invention may be demonstrated in vitro, in animal test methods as well as in clinic studies.
  • Utility of the compounds of formula (I) in accordance with the present invention may be demonstrated in accordance with the methods hereinafter described:
  • Rat1-Myr-p110 ⁇ cells were grown in Dulbecco's Modified Eagle Medium (DMEM) culture medium containing 4.5 g/l glucose supplemented with 10% heat-inactivated fetal calf serum (FCS), 2 mM L-glutamine, 1 mM sodium pyruvate and incubated at 37° C. in a 5% CO 2 humidified atmosphere. Cells were harvested with trypsin-EDTA, re-suspended in culture medium (with additives) and counted with a Casy® system. Finally, cells are centrifuged, suspended in ice-cold Hanks' balanced salt solution (HBSS) at a concentration of 3 ⁇ 10 7 cells/ml. Cell culture reagents were purchased from BioConcept (Allschwil, Switzerland).
  • HBSS Hanks' balanced salt solution
  • Rat1-myr-p110 ⁇ cells were generated by the method described in Maira et al., Molecular Cancer Therapeutics, 11:317-328 (2012), which is incorporated herein by reference in its entirety. Briefly, Rat1 cells were transfected to stably express the constitutively active form of the catalytic PI3K class I p110 isoforms a by addition of a myristylation signal to the N-terminus.
  • Rat1-Myr-p110 ⁇ tumors were established by subcutaneous injection of 5 ⁇ 10 6 cells in 100 ⁇ L HBSS (Sigma #H8264) into the right flank of nude mice or nude rats.
  • treatments were initiated when the mean tumor volumes were approx. 300 mm 3 (14 to 15 days post tumor cells injection).
  • animals were treated once orally with Compound A when the tumors reached a size of approx. 400-500 mm 3 (21 to 23 days post tumor cells injection).
  • Compound A was prepared for dosing as homogenous suspensions in 1% carboxymethyl cellulose: 0.5% Tween® 80: 98.5% deionized water. Fresh suspensions were prepared once every 4 days and stored at 4° C. Compound A or vehicle was administered orally at a volume of 10 mL/kg.
  • Tumor volumes were measured with calipers and determined according to the formula: length ⁇ diameter 2 ⁇ /6.
  • antitumor activity is expressed as T/C % (mean change of tumor volume of treated animals/mean change of tumor volume of control animals) ⁇ 100.
  • Regressions (%) were calculated according to the formula ((mean tumor volume at end of treatment ⁇ mean tumor volume at start of treatment)/mean tumor volume at start of treatment) ⁇ 100.
  • Body weights and tumor volumes were recorded two to three times a week.
  • the tumors were snap-frozen in liquid nitrogen and stored at ⁇ 80° C. Frozen tumors were pulverized using a Retsch ball mixer mill MM20 (Arlesheim, Switzerland) with metal cylinders that were pre-cooled to ⁇ 80° C. in a freezer. Powder was scrapped from metal cylinders on dry ice and transferred into pre-cooled 1.5 mL Eppendorf tubes while avoiding melting.
  • Concentrations of Compound A in plasma and tumor were determined in a separate run by using ultra-high-pressure liquid chromatography/tandem mass spectrometry (UPLC/MS-MS). Following addition of 25 ⁇ L of internal standard (1 ⁇ g/mL) to analytical aliquots (25 ⁇ L) of plasma or (20 mg) tumor powder, the proteins were precipitated by the addition of 200 ⁇ L acetonitrile. The supernatant were transferred in a fresh vial. After evaporation to dryness the samples were re-dissolved in 60 ⁇ L acetonitrile/water (1/1 v/v).
  • UPLC/MS-MS ultra-high-pressure liquid chromatography/tandem mass spectrometry
  • Precursor Product Precursor Product ion ion Internal ion ion [m/z] [m/z] standard [m/z] [m/z] Compound A 442.10 328.10 Compound B 387.10 273.10
  • the limit of quantification (LOQ) for Compound A was set to 2.5 ng/mL (CV and overall bias less than 30%). Regression analysis and further calculations were performed using QuanLynxTM 4.1 (Micromass) and ExcelTM 2007 (Microsoft). Concentrations of unknown samples were back-calculated based on the peak area ratios of analyte/IS from a calibration curve constructed using calibration samples spiked in blank plasma or tumor obtained from animals treated with vehicle.
  • NP40 protein lysis buffer mix (Lysis buffer stock (4° C.): 2.5 mL Tris HCL 2M pH 7.8 RT, 1 mL NP40 (100%) RT, 2.4 mL NaCl 5M RT, 2.5 mL NaF 1M RT, 4 mL 1M beta glycerol phosphate disodium salt penthahydrate ⁇ 20° C., and water till 100 mL; Lysis buffer solution (4° C.): 10 mL lysis buffer stock; 10 uL Na3VO3 100 mM 4° C., 10 uL DTT 1M ⁇ 20° C., 10 uL PMSF 100 mM 4° C., 10 uL Benzamidine 1M ⁇ 20° C., and 10 uL Microcystin ⁇ 20° C.).
  • Each sample was vortexed and centrifuged for 10 minutes at 10,000 rpm. A freezing thawing cycle was performed at ⁇ 80° C. for 30 minutes. The samples were stored, after an additional centrifugation step at 10 000 rpm for 10 minutes, at ⁇ 80° C. for further analysis.
  • the protein concentrations were quantified using the Coomassie Plus Kit (#23236 Thermo Scientific, Rockford, Ill., USA) according to the protocol of the manufacturer.
  • the diluted samples were transferred into a 96 well plate (#269620, NUNC) and the absorbance was measured at 595 nm using a SpectraMAX Plus plate reader from Molecular Devices.
  • the Protein amount was calculated using the Softmax Pro 5.0 software (Molecular Devices, USA) and then normalized on 1 mg/mL using the corresponding lysis buffer.
  • the normalized samples were further diluted 1:10 using the CSBL1 CeLyA spotting buffer (Zeptosens, cat. No. 9020) supplemented with 1 mM Na-orthovanadate (Sigma, cat No. S-6508).
  • the lysate was transferred to a 96-well V-bottom plate (Fisher Scientific, cat. No. 6067Y), followed by a centrifugation step (5 min, 1500 rpm at 19° C. in an Eppendorf 5810R centrifuge) to remove the unlyzed cell debris.
  • the CeLyA blocking buffer BB1 (Zeptosens, cat. No. 9040) is administered via an ultrasonic nebulizer. After 20 minutes of blocking the chips are extensively rinsed with deionized water (Milli-Q quality, 18M′ ⁇ cm) and dried in a nitrogen air flow.
  • the ZeptoMARK® chips were transferred to the ZeptoCARRIER (Zeptosens, cat. No. 1100), and washed twice with 200 ⁇ L CAB1 CeLyA assay buffer (Zeptosens, cat. No. 9032).
  • the assay buffer was then aspirated and each compartment incubated with 100 ⁇ L of the primary target antibody (pAkt Ser473 (lot no. 9)(Cell Signaling Technology, Catalog No. 4060); Akt1 pan (lot no. E0401) (Epitomics, Catalog No. 1085-1); Zenon® Alexa Fluor 647 rabbit (Invitrogen, Catalog No. Z25308)) at room temperature (RT) overnight.
  • the primary antibody was removed, the arrays washed twice with CAB1 buffer and further incubated with 100 ⁇ L of Alexa fluor 647-labeled anti rabbit IgG Fab fragments (Invitrogen; #Z25305) for one hour at RT in the dark. After incubation, the arrays were washed twice with 200 ⁇ L CAB1 buffer.
  • the fluorescence of the target-bound Fab fragments was read out on the ZeptoReader (Zeptosens, Witterswil, Switzerland) using a laser (excitation wavelength 635 nm) and a CCD camera. The fluorescence signal was assessed with exposure times of 1, 3, 5 and 10 seconds, depending on the intensity of the signal.
  • the fluorescence images for each array were analyzed with the ZeptoVIEW Pro 2.0 software (Zeptosens, Witterswil, Switzerland) and the RFI (relative fluorescence intensity) for each signal was calculated.
  • Absolute values for primary tumor growth and body weight were used to make the statistical comparisons between groups (one way ANOVA followed by Dunnett's test for normally distributed data; ANOVA on Ranks for not normally distributed data followed by Dunnett's test for equal group size or Dunn's for unequal group size). The significant level was set at p ⁇ 0.05. Areas under the curve (AUC) recorded for 24 h post last treatment was determined by using the trapezoidal rule method. All statistical calculations were carried out using SigmaStat.
  • the pre-clinical PK-PD-Efficacy-Tolerability model for Compound A was established following the methods set forth above. For this pre-clinical PK-PD-Efficacy-Tolerability model for Compound A:
  • FIGS. 2A and 2B provides a non-parametric superposition model to show the relationship of observed vs. predicted plasma concentrations after oral administration of Compound A at 50 mg/kg qd in nude mice and 40 mg/kg qd in nude rats. ( FIGS. 2 A and B).
  • FIGS. 2 A and B The pharmacokinetics of Compound A were linear over the range of doses tested ( FIG. 1 A: 12.5, 25 and 50 mg/kg qd in nude mice; FIG. 1 B: 12.5, 25, 40 and 80 mg/kg qd in nude rats), and associated with a similar change in AUC between 12.5 and 50 mg/kg in nude mice. A similar relationship was observed in nude rats for doses up to 80 mg/kg.
  • FIGS. 2A and 2B provides a non-parametric superposition model to show the relationship of observed vs. predicted plasma concentrations after oral administration of Compound A at 50 mg/kg qd in nude mice and 40 mg/kg qd in nude rats. (
  • FIG. 4A indicates that this PK model used is also predictive to simulate PK profiles Compound A given twice a day (2 qd) in nude mice.
  • the tumor concentrations giving 50% (in vivo IC 50 ) and 80% (in vivo IC 80 ) S473P-Akt inhibition (0.6 and 4 ⁇ mol/L, respectively) versus controls were determined by measuring the level of Akt phosphorylation using RPPA and the specific tumor drug concentration in matched samples from multiple animals (nude mice and rats) and at multiple time points post-treatment with Compound A ( FIG. 5 ).
  • the in vivo IC 50 (53 nmol/L) and IC 80 (352 nmol/L) values roughly approximate the in vitro cellular IC 50 and IC 80 of 74 nmol/L and 301 nmol/L respectively.
  • Compound A was administered orally to Rat1-myr-p110 ⁇ tumor bearing mice and rats at various doses. Tumor growth inhibition results are summarized below:
  • FIG. 8 provides a comparison of observed tumor growth inhibition and the model prediction tumor growth inhibition after oral administration of Compound A from 6.25 to 70 mg/kg in qd and 2 qd dosing.
  • the model tells us that 80% pAkt inhibition will be achieved for 100% of the time between two consecutive treatments leading to tumor regression.
  • the hyperglycemia threshold was set to 15 ⁇ mol/L
  • the dose of 30 mg/kg qd (no BW loss) will lead to 80% pAkt inhibition for 83% of the time between two consecutive treatments leading to 80% tumor regression ( FIG. 13 , FIG. 7 ).
  • the pre-clinical PK-PD-Efficacy-Tolerability modeling for Compound A described above is a valuable tool to predict efficacy and tolerability of the following dosing schedule of Compound A: oral administration of Compound A once per day (q.d.) or twice per day (b.i.d.) for five-consecutive days followed by no administration of Compound A for two days (CYCLE 1), and then a repeat of the same dosing regimen [i.e, oral administration of Compound A once per day (q.d.) or twice per day (b.i.d.) for five-consecutive days, followed by no administration of Compound A for two days] in one or more subsequent cycles.
  • This alternative dosing schedule is referred to as “ALTERNATIVE SCHEDULE 1”.
  • this model is here used to explore and guide dose scheduling in clinical studies.
  • FIG. 14 provides graphs showing the simulated efficacy of Compound A in Rat1-myr P110 ⁇ tumor bearing nude rats orally with COMPOUND A at 20 mg/kg in ALTERNATIVE SCHEDULE 1 (A) as compared to 14 mg/kg qd in continuous daily schedule (ie., with no drug holiday) (B).
  • FIG. 15 provides the simulated plasma PK profile in Rat1-myr P110 ⁇ tumor bearing nude rats orally with COMPOUND A at 20 mg/kg in ALTERNATIVE SCHEDULE 1 as compared to 14 mg/kg qd in continuous daily schedule (ie., with no drug holiday).
  • ALTERNATIVE SCHEDULE 1 for Compound A can (a) achieve similar or improved anti-tumor efficacy observed in nude rats orally administered Compound A once each day (q.d.) on a continuous daily schedule and (b) achieve at least partial regression (30% tumor regression) over the entire treatment period if Compound A plasma concentration is above the IC 80 on pAkt for 45% of the time between two treatment periods. Based on equivalent AUC, the human dose for Compound A of 300-350 mg/day p.o.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US15/101,155 2013-12-06 2014-12-03 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor Abandoned US20170000778A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP13306679 2013-12-06
EP13306679.5 2013-12-06
PCT/IB2014/066558 WO2015083101A1 (fr) 2013-12-06 2014-12-03 Schéma posologique d'un inhibiteur de la phosphatidylinositol 3-kinase sélectif de l'isoforme alpha

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/066558 A-371-Of-International WO2015083101A1 (fr) 2013-12-06 2014-12-03 Schéma posologique d'un inhibiteur de la phosphatidylinositol 3-kinase sélectif de l'isoforme alpha

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/719,636 Continuation-In-Part US10434092B2 (en) 2013-12-06 2017-09-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US15/719,636 Continuation US10434092B2 (en) 2013-12-06 2017-09-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor

Publications (1)

Publication Number Publication Date
US20170000778A1 true US20170000778A1 (en) 2017-01-05

Family

ID=49876516

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/101,155 Abandoned US20170000778A1 (en) 2013-12-06 2014-12-03 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US15/719,636 Expired - Fee Related US10434092B2 (en) 2013-12-06 2017-09-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US16/556,111 Abandoned US20200069670A1 (en) 2013-12-06 2019-08-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US17/105,453 Abandoned US20210196696A1 (en) 2013-12-06 2020-11-25 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/719,636 Expired - Fee Related US10434092B2 (en) 2013-12-06 2017-09-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US16/556,111 Abandoned US20200069670A1 (en) 2013-12-06 2019-08-29 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
US17/105,453 Abandoned US20210196696A1 (en) 2013-12-06 2020-11-25 Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor

Country Status (23)

Country Link
US (4) US20170000778A1 (fr)
EP (2) EP4000619A1 (fr)
JP (1) JP2016539149A (fr)
KR (1) KR20160095035A (fr)
CN (1) CN105979947A (fr)
AU (2) AU2014358773A1 (fr)
BR (1) BR112016011811A2 (fr)
CA (1) CA2930359C (fr)
CL (1) CL2016001361A1 (fr)
DK (1) DK3076969T3 (fr)
ES (1) ES2899167T3 (fr)
HK (1) HK1223549A1 (fr)
HU (1) HUE057092T2 (fr)
IL (1) IL245709B (fr)
MX (1) MX2016007376A (fr)
PH (1) PH12016501026A1 (fr)
PL (1) PL3076969T3 (fr)
PT (1) PT3076969T (fr)
RU (1) RU2680246C1 (fr)
SI (1) SI3076969T1 (fr)
TN (1) TN2016000179A1 (fr)
TW (1) TWI666019B (fr)
WO (1) WO2015083101A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180073674A (ko) * 2015-11-02 2018-07-02 노파르티스 아게 포스파티딜이노시톨 3-키나제 억제제에 대한 투여 요법
WO2018060833A1 (fr) * 2016-09-27 2018-04-05 Novartis Ag Schéma posologique pour l'alpelisib, un inhibiteur de la phosphatidylinositol 3-kinase spécifique de l'isoforme alpha

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6041077B2 (ja) 1976-09-06 1985-09-13 喜徳 喜谷 1,2‐ジアミノシクロヘキサン異性体のシス白金(2)錯体
US4323581A (en) 1978-07-31 1982-04-06 Johnson & Johnson Method of treating carcinogenesis
IL73534A (en) 1983-11-18 1990-12-23 Riker Laboratories Inc 1h-imidazo(4,5-c)quinoline-4-amines,their preparation and pharmaceutical compositions containing certain such compounds
WO1988007045A1 (fr) 1987-03-09 1988-09-22 Kyowa Hakko Kogyo Co., Ltd. Derives de la substance physiologiquement active k-252
US4904768A (en) 1987-08-04 1990-02-27 Bristol-Myers Company Epipodophyllotoxin glucoside 4'-phosphate derivatives
WO1989007105A1 (fr) 1988-02-04 1989-08-10 Kyowa Hakko Kogyo Co., Ltd. Derives de staurosporine
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
US5389640A (en) 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US6410010B1 (en) 1992-10-13 2002-06-25 Board Of Regents, The University Of Texas System Recombinant P53 adenovirus compositions
US5268376A (en) 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
AU2866992A (en) 1991-10-10 1993-05-03 Schering Corporation 4'-(n-substituted-n-oxide)staurosporine derivatives
US5266575A (en) 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
CA2123096A1 (fr) 1991-11-08 1993-05-13 Beat J. Knusel Compositions contenant des composes k-252 pour la potentialisation de l'activite neurotrophine
AU661533B2 (en) 1992-01-20 1995-07-27 Astrazeneca Ab Quinazoline derivatives
US5948898A (en) 1992-03-16 1999-09-07 Isis Pharmaceuticals, Inc. Methoxyethoxy oligonucleotides for modulation of protein kinase C expression
US5621100A (en) 1992-07-24 1997-04-15 Cephalon, Inc. K-252a derivatives for treatment of neurological disorders
US5756494A (en) 1992-07-24 1998-05-26 Cephalon, Inc. Protein kinase inhibitors for treatment of neurological disorders
ATE280180T1 (de) 1992-08-31 2004-11-15 Ludwig Inst Cancer Res Vom mage-3-gen abgeleitetes und von hla-a1 präsentiertes, isoliertes nonapeptid und dessen anwendungen
CA2123895A1 (fr) 1992-09-21 1994-03-31 Tatsuya Tamaoki Agent pour le traitement de la thrombocytopenie
DK0666868T4 (da) 1992-10-28 2006-09-18 Genentech Inc Anvendelse af anti-VEGF-antistoffer til behandling af cancer
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
JPH09500128A (ja) 1993-07-15 1997-01-07 ミネソタ マイニング アンド マニュファクチャリング カンパニー イミダゾ〔4,5−c〕ピリジン−4−アミン
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5478932A (en) 1993-12-02 1995-12-26 The Board Of Trustees Of The University Of Illinois Ecteinascidins
ES2236702T3 (es) 1993-12-23 2005-07-16 Eli Lilly And Company Inhibidores de la proteina quinasa c.
US5587459A (en) 1994-08-19 1996-12-24 Regents Of The University Of Minnesota Immunoconjugates comprising tyrosine kinase inhibitors
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
US5482936A (en) 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
EP1110953B1 (fr) 1995-03-30 2009-10-28 Pfizer Products Inc. Dérivés de quinazolinone
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
US6331555B1 (en) 1995-06-01 2001-12-18 University Of California Treatment of platelet derived growth factor related disorders such as cancers
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
WO1997007081A2 (fr) 1995-08-11 1997-02-27 Yale University Synthese d'indolocarbazols glycosyles
FR2741881B1 (fr) 1995-12-01 1999-07-30 Centre Nat Rech Scient Nouveaux derives de purine possedant notamment des prorietes anti-proliferatives et leurs applications biologiques
EP0865440B1 (fr) 1995-12-08 2002-04-03 Janssen Pharmaceutica N.V. Derives de la (imidazol-5-yl)methyl-2-quinoleinone comme inhibiteur de la proteine farnesyle-transferase
KR100447918B1 (ko) 1996-07-25 2005-09-28 동아제약주식회사 대장을포함한위장관보호작용을갖는플라본및플라바논화합물
AU738592C (en) 1996-08-02 2002-07-25 Lorus Therapeutics Inc. Antitumor antisense sequences directed against R1 and R2 components of ribonucleotide reductase
CO4950519A1 (es) 1997-02-13 2000-09-01 Novartis Ag Ftalazinas, preparaciones farmaceuticas que las comprenden y proceso para su preparacion
US6126965A (en) 1997-03-21 2000-10-03 Georgetown University School Of Medicine Liposomes containing oligonucleotides
EP1017394B1 (fr) 1997-07-12 2005-12-07 Cancer Research Technology Limited Derives de purine inhibant la kinase dependant de la cycline
GB9800569D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
DK2020408T3 (da) 1998-05-29 2013-09-30 Sugen Inc Pyrrol-substitueret 2-indolinon som proteinkinaseinhibitor
US20030083242A1 (en) 1998-11-06 2003-05-01 Alphonse Galdes Methods and compositions for treating or preventing peripheral neuropathies
CN100340575C (zh) 1999-06-25 2007-10-03 杰南技术公司 人源化抗ErbB2抗体及其在制备药物中的应用
US6806266B1 (en) 1999-07-13 2004-10-19 Kyowa Hakko Kogyo Co., Ltd. Staurosporin derivatives
UA72946C2 (uk) 1999-11-05 2005-05-16 Астразенека Аб Похідні хіназоліну як інгібітори васкулярного ендотеліального фактора росту (vegf)
DE60030770T2 (de) * 1999-12-06 2007-09-06 Rhode Island Hospital Verwendung von taurolidin oder taurultam zur herstellung eines medikaments zur behandlung von ovarialkarzinomen
CA2399358C (fr) 2000-02-15 2006-03-21 Sugen, Inc. Inhibiteurs de la proteine kinase 2-indolinone a substitution pyrrole
PT1274718E (pt) 2000-04-12 2007-01-31 Genaera Corp Processo para a preparação de esteróis 7. alfa-hidroxi 3- aminosubstítuidos utilizando intermediários com um grupo 7. alfa-hidroxilo desprotegido
JP4102185B2 (ja) 2000-06-30 2008-06-18 グラクソ グループ リミテッド キナゾリンジトシル酸塩化合物
EA006711B1 (ru) 2000-09-11 2006-02-24 Чирон Корпорейшн Хинолиноновые производные в качестве ингибиторов тирозинкиназы
US6677450B2 (en) 2000-10-06 2004-01-13 Bristol-Myers Squibb Company Topoisomerase inhibitors
AU2002245272B2 (en) 2001-01-16 2006-06-29 Regeneron Pharmaceuticals, Inc. Isolating cells expressing secreted proteins
WO2002062826A1 (fr) 2001-02-07 2002-08-15 Vadim Viktorovich Novikov Procede de fabrication des peptides
EP1404689A1 (fr) 2001-07-02 2004-04-07 Debiopharm S.A. Substance active a base d'oxaliplatine presentant une faible teneur en acide oxalique
KR100484504B1 (ko) 2001-09-18 2005-04-20 학교법인 포항공과대학교 쿠커비투릴 유도체를 주인 분자로서 포함하고 있는 내포화합물 및 이를 포함한 약제학적 조성물
US20030134846A1 (en) 2001-10-09 2003-07-17 Schering Corporation Treatment of trypanosoma brucei with farnesyl protein transferase inhibitors
WO2003077902A1 (fr) 2002-02-19 2003-09-25 Xenoport, Inc. Procede de synthese de promedicaments a partir de derives de 1-acyl-alkyl et compositions correspondantes
CA2480638C (fr) 2002-03-29 2013-02-12 Chiron Corporation Benzazoles substitues et leur utilisation en tant qu'inhibiteurs de la kinase raf
US6727272B1 (en) 2002-07-15 2004-04-27 Unitech Pharmaceuticals, Inc. Leflunomide analogs for treating rheumatoid arthritis
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
EP1587473A4 (fr) 2002-12-27 2008-08-13 Novartis Vaccines & Diagnostic Thiosemicarbazones antiviraux et immunostimulants
ES2391770T3 (es) 2003-01-21 2012-11-29 Novartis Vaccines And Diagnostics, Inc. Uso de compuestos de triptantrina para la potenciación inmune
US7893096B2 (en) 2003-03-28 2011-02-22 Novartis Vaccines And Diagnostics, Inc. Use of small molecule compounds for immunopotentiation
SI3248600T1 (sl) * 2005-02-18 2020-09-30 Abraxis Bioscience, Llc Kombinacije in načini dajanja terapevtskih sredstev in kombinacijska terapija
UA104147C2 (uk) 2008-09-10 2014-01-10 Новартис Аг Похідна піролідиндикарбонової кислоти та її застосування у лікуванні проліферативних захворювань
CN105412105A (zh) * 2010-11-08 2016-03-23 诺华有限公司 2-甲酰胺环氨基尿素衍生物在治疗egfr依赖性疾病或对靶向egfr家族成员的药剂有获得性耐受性的疾病中的应用
US9006270B2 (en) * 2011-06-21 2015-04-14 Novartis Ag Polymorphs of (S)-pyrrolidine-1,2-dicarboxylic acid 2-amide 1-({4-methyl-5-[2-(2,2,2-trifluoro-1,1-dimethyl-ethyl)-pyridin-4-yl]-thiazol-2-yl}-amide
EP2601965A1 (fr) * 2011-12-06 2013-06-12 Apeiron Biologics AG Compositions pour la prévention ou le traitement des réactions indésirables de l'inhibition d'EGFR
MX360892B (es) 2012-05-16 2018-11-20 Novartis Ag Régimen de dosificación para un inhibidor de cinasa pi-3.

Also Published As

Publication number Publication date
SI3076969T1 (sl) 2021-12-31
CL2016001361A1 (es) 2017-02-10
HK1223549A1 (zh) 2017-08-04
TN2016000179A1 (en) 2017-10-06
PH12016501026A1 (en) 2016-07-04
JP2016539149A (ja) 2016-12-15
EP4000619A1 (fr) 2022-05-25
CA2930359C (fr) 2022-03-01
CN105979947A (zh) 2016-09-28
US20210196696A1 (en) 2021-07-01
KR20160095035A (ko) 2016-08-10
EP3076969B1 (fr) 2021-09-01
IL245709A0 (en) 2016-07-31
AU2017245292A1 (en) 2017-11-02
TWI666019B (zh) 2019-07-21
IL245709B (en) 2021-03-25
US10434092B2 (en) 2019-10-08
AU2017245292B2 (en) 2018-09-27
PT3076969T (pt) 2021-11-23
DK3076969T3 (da) 2021-11-22
TW201609096A (zh) 2016-03-16
PL3076969T3 (pl) 2022-01-17
EP3076969A1 (fr) 2016-10-12
US20200069670A1 (en) 2020-03-05
ES2899167T3 (es) 2022-03-10
HUE057092T2 (hu) 2022-04-28
MX2016007376A (es) 2017-05-11
WO2015083101A1 (fr) 2015-06-11
AU2014358773A1 (en) 2016-06-02
RU2680246C1 (ru) 2019-02-19
BR112016011811A2 (pt) 2017-08-08
CA2930359A1 (fr) 2015-06-11
US20180078540A1 (en) 2018-03-22

Similar Documents

Publication Publication Date Title
RU2607944C2 (ru) Синергические композиции ингибиторов pi3k и мек
AU2019226212B2 (en) Combination of Pl3K inhibitor and c-Met inhibitor
US20210196696A1 (en) Dosage regimen for an alpha-isoform selective phosphatidylinositol 3-kinase inhibitor
KR20160020502A (ko) 제약 조합물
WO2018060833A1 (fr) Schéma posologique pour l'alpelisib, un inhibiteur de la phosphatidylinositol 3-kinase spécifique de l'isoforme alpha
US20180280370A1 (en) Dosage regimen for a phosphatidylinositol 3-kinase inhibitor
KR20230107606A (ko) 저등급 신경교종 치료를 위한 raf 억제제

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DI TOMASO, EMMANUELLE;REEL/FRAME:039981/0980

Effective date: 20140811

Owner name: NOVARTIS PHARMACEUTICALS CORPORATION, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TAVORATH, RANJANA;REEL/FRAME:039982/0053

Effective date: 20140624

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:039982/0268

Effective date: 20140507

Owner name: NOVARTIS PHARMA S.A.S., FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GERMA, MARIE-CAROLINE;MASSACESI, CRISTIAN;REEL/FRAME:039982/0092

Effective date: 20140429

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.;REEL/FRAME:039981/0989

Effective date: 20140821

Owner name: NOVARTIS PHARMA AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRITSCH, CHRISTINE;SCHNELL, CHRISTIAN RENE;SIGNING DATES FROM 20140425 TO 20140428;REEL/FRAME:039982/0206

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMACEUTICALS CORPORATION;REEL/FRAME:040302/0018

Effective date: 20140821

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMA S.A.S.;REEL/FRAME:040302/0079

Effective date: 20140502

STCB Information on status: application discontinuation

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