WO2013116562A1 - Compositions and methods of treating a disease with (s)-4 amino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile - Google Patents

Compositions and methods of treating a disease with (s)-4 amino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile Download PDF

Info

Publication number
WO2013116562A1
WO2013116562A1 PCT/US2013/024216 US2013024216W WO2013116562A1 WO 2013116562 A1 WO2013116562 A1 WO 2013116562A1 US 2013024216 W US2013024216 W US 2013024216W WO 2013116562 A1 WO2013116562 A1 WO 2013116562A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
enantiomer
acceptable salt
administered
Prior art date
Application number
PCT/US2013/024216
Other languages
French (fr)
Inventor
Jerry B. Evarts
Original Assignee
Gilead Calistoga Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Calistoga Llc filed Critical Gilead Calistoga Llc
Publication of WO2013116562A1 publication Critical patent/WO2013116562A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4741Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having oxygen as a ring hetero atom, e.g. tubocuraran derivatives, noscapine, bicuculline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present application is in the field of therapeutics and medicinal chemistry.
  • the present application concerns methods of treatment of asthma that include
  • PI 3 -kinase phosphatidylinositol 3 -kinase
  • PI 3-kinase activation is believed to be involved in a range of cellular responses including cell growth, differentiation, and apoptosis.
  • PI3K participates in cellular pathways involved in hematological malignancy and solid tumor activation.
  • PI3K participates in a cellular pathway that has been implicated in the process of oncogenic
  • T-cell acute lymphoblastic leukemia T-cell acute lymphoblastic leukemia
  • PI3-kinase The initial purification and molecular cloning of PI3-kinase revealed that it was a heterodimer consisting of p85 and pi 10 subunits.
  • Class I PI3Ks Four Class I PI3Ks have been identified and designated as PI3K ⁇ , ⁇ , ⁇ , and ⁇ isomers. Each isomer consists of a distinct pi 10 catalytic subunit and a regulatory subunit. Three catalytic subunits, pi 10a, pi 10 ⁇ and pi 105, each interact with the same regulatory subunit, p85; whereas pi 10 ⁇ interacts with a distinct regulatory subunit, plOl.
  • the expression of each PI3K isoform in human cells and tissues are also distinct.
  • the present application discloses compounds, compositions and methods related to treating a disease or a condition related to PI3K-mediated disorders.
  • a disease may include, for example, asthma.
  • Unexpected effects on PI3K isomers have been found in the compounds disclosed herein.
  • Provided is a compound having the structure of Compound A
  • the compound is the (S)-enantiomer, having the structure of Compound A(S):
  • composition comprising a compound of any of the foregoing embodiments of Compound A, and at least one pharmaceutically acceptable excipient.
  • the composition comprises a therapeutically effective amount of
  • the compound is a racemic mixture of the (R)- and (S)- enantiomers of Compound A.
  • the compound is optically active.
  • the (S)-enantiomer of Compound A having the structure of Compound A(S):
  • the compound is substantially free of Compound A(R).
  • the (S)-enantiomer of Compound A predominates over the (R)-enantiomer of Compound A by a molar ratio of at least 9: 1, at least 19: 1, at least 40: 1, at least 80: 1, at least 160: 1, or at least 320: 1.
  • the compound can also be described by its enantiomeric excess (e.e.). For instance, a compound with 95% (S)-isomer and 5% (R)-isomer will have an e.e. of 90%. In some
  • the compound has an e.e. of at least 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%.
  • the compound is enantiomerically-enriched in the (S)- isomer of Compound A.
  • a disease or a condition related to PI3K-mediated disorders is characterized by histamine release (e.g. , allergic disorders).
  • disorders that may be treated by the methods provided herein may include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, and emphysema.
  • COPD chronic obstructive pulmonary disease
  • asthma ARDS
  • emphysema emphysema
  • the disease is asthma.
  • a method of treating a disease or a condition related to PDK-mediated disorders comprising administering to a patient in need there of an effective amount of any of the foregoing compounds or compositions.
  • Specific embodiments provide a method of treating asthma in a patient, comprising administering to the patient Compou
  • compositions comprising Compound A or a pharmaceutically acceptable salt thereof, optionally admixed with at least one pharmaceutically acceptable excipient.
  • the composition comprises a therapeutically effective amount of a compound of any of the foregoing embodiments of Compound A or a pharmaceutically acceptable salt thereof for the treatment of asthma in a patient.
  • the composition comprises a racemic mixture of Compound A.
  • the composition comprises the (S)-enantiomer of Compound A, having the structure of Compound A(S):
  • composition is substantially free of the (R)-enantiomer of Compound A.
  • Compound A is administered at a dose of about 1 to 4,000 mg/day, about 2,000 to 4,000 mg/day, about 1 to 2,000 mg/day, about 1 to 1,000 mg/day, 10 to 500 mg/day, about 20 to 500 mg/day, about 50 to 300 mg/day, about 75 to 200 mg/day, or about 15-150 mg/day.
  • Compound A is administered at a dose of about 1 to 150 mg twice per day.
  • Compound A is administered at least twice daily.
  • Compound A is administered intermittently or in intervals.
  • the interval may range from one, two, three, four, five, six and seven days.
  • Compound A is administered for at least 21 days, and is then discontinued for at least 7 days.
  • Compound A is
  • the method further comprises reducing the level of PI3K5 activity in the patient in need thereof. In some of the foregoing embodiments, the method further comprises the PBKcc-sparing activity in the patient in need thereof.
  • the method further comprises administering to a patient, in addition to Compound A, a therapeutically effective amount of at least one therapeutic agent selected to treat the PI3K-mediated condition in the patient.
  • the therapeutic agent is selected from the following group consisting of theophylline, nedocromil sodium, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, a beta 2 agonist, an
  • antihistamine an anticholinergic, allergen immunotherapy, albuterol, levalbuterol, pirbutrol, ipratropium bromide, methylprednisolone, prednisolone, prednisone, beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone acetonide, salmeterol, formoterol, montelukast, zafirlukast, mometasone, ciclesonide, alfuzosin, ephedrine, metaproterenol, phenylpropanolamine,
  • FIG. 1 Concentration-time profiles of Compound A(S) in plasma following a 30- minute IV infusion at 0.44 mg/kg in rats. Data is shown for individual rats.
  • FIG. Concentration-time profiles of Compound A(S) in plasma following an oral dose at 1.52 mg/kg in rats. Data is shown for individual rats.
  • novel methods to treat a disease or a condition related to PI3K-mediated disorders using Compound A are novel methods to treat a disease or a condition related to PI3K-mediated disorders using Compound A.
  • the disease is asthma.
  • One aspect provides a compound having the structure of C
  • composition comprising Compound A or a pharmaceutically acceptable salt thereof, optionally admixed with at least one pharmaceutically acceptable excipient.
  • the compound is the (S)-enantiomer, having the structure of Compound A(S):
  • Compound A in which from 1 to n hydrogens attached to a carbon atom may be replaced by deuterium, in which n is the number of hydrogens in the molecule.
  • Such compounds may exhibit increased resistance to metabolism, and thus may be useful for increasing the half life of Compound A when administered to a mammal. See, e.g., Foster, "Deuterium Isotope Effects in Studies of Drug Metabolism", Trends Pharmacol. Sci., 5(12):524-527 (1984).
  • Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium.
  • Compositions comprising Compound A may include racemic mixtures or mixtures containing an enantiomeric excess of one enantiomer or single diastereomers or diastereomeric mixtures. All such isomeric forms of these compounds are expressly included herein the same as if each and every isomeric form were specifically and individually listed.
  • Compound A and compositions thereof for use in the methods described herein may be optically active.
  • Compound A has a single chiral center in the noncyclic linking group between the quinazolinone moiety and the pyrimidine moiety.
  • the preferred enantiomer of Compound A is the (S)-enantiomer depicted above.
  • Optically active forms of Compound A may include predominantly the (S)-enantiomer, although it may also include the (R)-enantiomer of Compound A as a minor component.
  • the dosage refers to the weight of Compound A, including each enantiomer that may be present.
  • a dosage of 100 mg of Compound A refers to the weight of the mixture of enantiomers rather than the weight of the (S)- enantiomer specifically.
  • Compound A may be synthesized in optically active form, or it may be prepared in racemic form (containing equal amounts of R- and (S)-isomers), and then the isomers may be separated.
  • Scheme 1 depicts a chiral synthesis of Compound A that provides the (S) -enantiomer in very high optical purity.
  • the enantiomeric (R)-isomer of Compound A may be excluded.
  • the methods may be practiced with mixtures of R- and (S)- isomers.
  • the methods may be practiced with mixtures of R- and (S)- isomers, in which the (S)-isomer is the major component of the mixture.
  • such mixture may contain no more than about 10% of the R isomer, meaning the ratio of S- to (R)-isomers is at least about 9: 1, and in other embodiments, less than 5% of the (R)-isomer, meaning the ratio of S- to (R)-enantiomers is at least about 19: 1.
  • the (S)-enantiomer predominates over the (R)-enantiomer by a molar ratio of at least 40: 1, at least 80: 1, at least 160: 1, or at least 320: 1.
  • Compound A can also be described by its enantiomeric excess (e.e.). For instance, a compound characterized by 95% (S)-isomer and 5% (R)-isomer will have an e.e. of 90%. In some embodiments, Compound A has an e.e. of at least 60%, at least 75%, at least 80%, at least 90%, at least 95%, at least 98%, or at least 99%.
  • the compound is enantiomerically-enriched in the (S)-isomer of Compound A.
  • the compound may be enriched with the (S)- enantiomer shown here:
  • Compound A is primarily composed of the (S)-enantiomer of Compound A, wherein this isomer comprises at least 66-95%, or about 85-99% of the (S)- enantiomer, in excess over any (R)-enantiomer present. In certain embodiments, Compound A is at least 95% of the (S)-enantiomer of Compound A. In one embodiment, Compound A is 100% of the (S)-enantiomer. In the additional embodiment, the sample of Compound A is at least 99% the (S)-enantiomer, with less than 1% of the (R)-enantiomer.
  • the compounds depicted herein may be present as salts even if the salts are not depicted.
  • the salts of the compounds disclosed in the application are pharmaceutically acceptable salts.
  • the methods described herein are useful to treat a disease or a condition related to PI3K- mediated disorders.
  • the disease is asthma.
  • the application discloses compounds capable of inhibiting the biological activity of human PI3K5.
  • the application provides compounds that inhibit PI3K5 selectively compared to the other PI3K isoforms.
  • the application provides a method of selectively modulating human PI3K5 activity, and thereby treating diseases mediated by PI3K5 dysfunction.
  • the application discloses a method of treating a disease or a condition related to PI3K-mediated disorders.
  • the disease or condition is
  • disorders that may be treated by the methods described herein may include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, and emphysema.
  • COPD chronic obstructive pulmonary disease
  • asthma ARDS
  • emphysema emphysema
  • the disease is asthma.
  • the method of treating a PI3K-mediated disorder or condition comprises administering to a patient in need there of an effective amount of any of the foregoing compounds or compositions.
  • the application discloses a method of disrupting leukocyte function comprising contacting leukocytes with a compound that selectively inhibits phosphatidylinositol 3- kinase delta (PI3K5) activity in the leukocytes.
  • the leukocytes can comprise cells selected from the group consisting of neutrophils, B lymphocytes, T lymphocytes, and basophils.
  • the method comprises disrupting at least one neutrophil function selected from the group consisting of stimulated superoxide release, stimulated exocytosis, and chemotactic migration.
  • the method does not substantially disrupt bacterial phagocytosis or bacterial killing by the neutrophils.
  • the leukocytes comprise B lymphocytes
  • the method comprises disrupting proliferation of the B lymphocytes or antibody production by the B lymphocytes.
  • the leukocytes comprise T lymphocytes
  • the method comprises disrupting proliferation of the T lymphocytes.
  • the leukocytes comprise basophils
  • the method comprises disrupting histamine release by the basophils.
  • the application discloses methods of inhibiting neutrophil functions, without substantially inhibiting phagocytosis of bacteria.
  • Neutrophil functions suitable for inhibition according to the present method include any function mediated by PI3K5 activity or expression. Such functions include, without limitation, stimulated superoxide release, stimulated exocytosis or degranulation, chemotactic migration, adhesion to vascular endothelium (e.g., tethering/rolling of neutrophils, triggering of neutrophil activity, and/or latching of neutrophils to endothelium), transmural diapedesis, or emigration through the endothelium to peripheral tissues.
  • these functions can be collectively termed "inflammatory functions," as they are typically related to neutrophil response to inflammation.
  • the inflammatory functions of neutrophils can be distinguished from the bacterial killing functions exhibited by these cells, e.g., phagocytosis and killing of bacteria. Accordingly, the application further includes methods of treating disease states in which one or more of the inflammatory functions of neutrophils are abnormal or undesirable.
  • the compounds of the present application may be used to inhibit an endogenous immune response stimulated by at least one endogenous factor without substantially inhibiting an exogenous immune response stimulated by at least one exogenous factor as disclosed in US 2005/0043239 Al.
  • the compounds of the present application may also be used to inhibit an endogenous immune response stimulated by at least one endogenous factor without substantially inhibiting immune responsiveness, as disclosed in US 2005/0043239 Al. Accordingly, the compounds of the application advantageously permit treatment of conditions associated with an undesirable endogenous immune response stimulated by at least one endogenous factor without compromising the ability to fight infection.
  • the compounds of the present application may also be used to inhibit leukocyte accumulation as disclosed in US 2005/0054614 Al.
  • the compounds may also be used to inhibit leukocyte tethering to endothelial cells and to inhibit leukocyte transmigration into inflamed tissue, as disclosed in US 2005/0043239 Al . Accordingly, the compounds of the application
  • PI3K5 plays a role in the stimulated proliferation of lymphocytes, including B cells and T cells. Moreover, PI3K5 appears to play a role in stimulated secretion of antibodies by B cells. Selective PI3K5 inhibitor compounds of the present application have been employed to establish that these phenomena can be abrogated by inhibition ofPI3K5.
  • the present application includes methods for inhibiting lymphocyte proliferation, or for inhibiting antibody production by B lymphocytes. Other methods enabled by the present application include methods of treating disease states in which one or more of these lymphocyte functions are abnormal or undesirable.
  • the application includes a method of suppressing a function of basophils and/or mast cells, thereby enabling treatment of diseases, conditions or disorders characterized by excessive or undesirable basophil and/or mast cell activity.
  • a present compound can be used to selectively inhibit the expression or activity of PI3K5 in the basophils and/or mast cells.
  • the method employs a PI3K5 inhibitor in an amount sufficient to inhibit stimulated histamine release by the basophils and/or mast cells.
  • the use of a selective PI3K5 inhibitor can be of value in treating diseases characterized by histamine release (e.g. , allergic disorders).
  • disorders that may be treated by the methods described herein include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, emphysema, and other related disorders.
  • COPD chronic obstructive pulmonary disease
  • ARDS emphysema
  • Compound A is selective for the ⁇ isoform of PI3K over the ⁇ , ⁇ , and ⁇ isoforms.
  • Another aspect includes methods of using Compound A or compositions thereof to treat a condition related to PI3K-mediated disorders such as inflammation or inflammatory disease.
  • Inflammation is a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute or wall off (i.e., sequester) both the injurious agent and the injured tissue.
  • Inflammation or inflammatory disease can be acute or chronic, and often involves the immune response. Inflammation typically results from a cascade of events that includes vasodilation accompanied by increased vascular permeability and exudation of fluid and plasma proteins. The disruption of vascular integrity precedes or coincides with an infiltration of inflammatory cells. Inflammatory mediators generated at the site of the initial lesion serve to recruit inflammatory cells to the site of injury.
  • mediators chemokines such as IL-8, MCP-1, MIP-1, and RANTES, complement fragments and lipid mediators
  • chemokines such as IL-8, MCP-1, MIP-1, and RANTES, complement fragments and lipid mediators
  • Inflammatory disease occurs when the normal discontinuation or attenuation of an inflammatory response does not occur or is incomplete. This includes but not limited to autoimmune disease, allergic disease, arthritic disease, asthma, acne, dermatitis, hypersensitive, transplant rejection, and inflammatory bowel disease.
  • the term 'potency' or variant thereof refers to one compound has an increased levels of activity when compared to other compounds at a specific concentration.
  • the potency of the compound is determined by the IC 50 value, which can be determined using commonly available methods; including in vitro enzyme assays or in vitro protein kinase assays.
  • IC 50 value can be determined using commonly available methods; including in vitro enzyme assays or in vitro protein kinase assays.
  • a compound having a lower IC 50 value is more potent than a compound having higher IC 50 value.
  • the term 'selectivity' or variant thereof refers to one compound has an increased level of activity on one isoform than other isoforms.
  • the selectivity is the activity on some PI3K isoform and not other PI3K isoforms exerted by the compound disclosed herein.
  • the selectivity is determined using the EC 50 value, which can be determined using commonly available methods for cellular assays. As understood by a person skilled in the art, a compound having a lower EC 50 value is more selective than a compound having a higher EC 50 value.
  • the method further comprises reducing the level of PI3K5 activity in the patient. In some embodiments, the method further comprises the PI3K a- sparing activity in the patient in need thereof.
  • the term 'the PDKa-sparing activity' or variant thereof refers to compounds having greater activity in one or more PI3K isoforms of ⁇ , ⁇ , or ⁇ than the PI3K a isoform in cellular assays.
  • compounds having PDKa-sparing activity inhibits PI3K5 more than PDKa, thereby reducing the activity of PI3K5 response more than that of PDKa.
  • treatments of the methods described herein involve administration of Compound A to a subject in need of treatment on a daily basis for at least one week or more than one week.
  • Compound A is administered to a subject in need thereof on a daily basis for 2 to 4 weeks, for 3 to 4 weeks, for 1 month or more, or for 1 year or more.
  • Compound A may be administered in multiple doses each day, in order to maintain efficacious plasma levels over a prolonged period of time. Administration may be done in one dose per day, two doses per day, three doses per day, or four doses per day.
  • Compound A can be administered intravenously at a rate that maintains an efficacious plasma level for a prolonged period of time.
  • the therapeutically effective amount can be determined by one of ordinary skill based on the subject's health, age, body weight, and condition. In some embodiments, the amount is normalized to the subject's body weight. For example, a dosage may be expressed as a number of milligrams of Compound A per kilogram of the subject's body weight (mg/kg). Dosages of between about 0.1 and 150 mg/kg are often appropriate, and in some embodiments, about 0.1 and 100 mg/kg are often appropriate, and in other embodiments a dosage of between 0.5 and 60 mg/kg is used.
  • Normalizing according to the subject's body weight is particularly useful when adjusting dosages between subjects of widely disparate size, such as occurs when using the drug in both children and adult humans or when converting an effective dosage in a non-human subject such as dog to a dosage suitable for a human subject.
  • the daily dosage may be described as a total amount of Compound A administered per dose or per day.
  • Daily dosage of Compound A in some embodiments is between about 1 mg and 4,000 mg.
  • Compound A is administered at a dose of about 2,000 to 4,000 mg/day.
  • Compound A is administered at a dose of about 1 to 2,000 mg/day.
  • Compound A is administered at a dose of about 1 to 1,000 mg/day.
  • Compound A is administered at a dose of about 10 to 500 mg/day.
  • Compound A is administered at a dose of about 20 to 500 mg/day.
  • Compound A is administered at a dose of about 50 to 300 mg/day.
  • Compound A is administered at a dose of about 75 to 200 mg/day.
  • Compound A is administered at a dose of about 15-150 mg/day.
  • Compound A is administered at a dose of about 10-500 mg/day. In a particular embodiment, Compound A is administered at a dose of about 50-300 mg/day. In a particular embodiment, Compound A is administered at a dose of about 75-200 mg/day. In a particular embodiment, Compound A is administered at a dose of about 100-150 mg/day.
  • Compound A is administered at a dose of about 1 to 150 mg per dose, and one to four doses are administered per day (e.g. , QD dosing with about 1 to 150 mg, BID dosing with about 1 to 150 mg, or TID dosing with doses between about 1 to 150 mg, or QID dosing with doses between about 1 to 150 mg).
  • a subject is treated with about 1 mg to 150 mg doses of Compound A once, twice, three, or four times per day.
  • QD refers to dosing once per day
  • BID refers to dosing twice per day
  • TID refers to dosing three times per day
  • QID refers to dosing four times per day.
  • the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of Compound A and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
  • this method comprises continuing to treat the subject by administering Compound A at a dosage where clinical efficacy is achieved for a week or more, or reducing the dose by increments to a level at which safety and efficacy can be maintained.
  • Safety can be monitored by conventional methods such as evaluating serum chemistry and complete blood count parameters.
  • Efficacy can be monitored by conventional methods known in the art.
  • the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of Compound A and increasing the dose to a total dosage of about 50 to 400 mg per day over at least 6 days.
  • the dosage can be further increased to about 750 mg/day.
  • Compound A is administered once daily. In another embodiment, Compound A is administered at least twice daily. In some embodiments Compound A is administered three times per day. In some embodiments, Compound A is administered four times per day, or more than four times per day.
  • Compound A is administered at a rate selected to produce a concentration of compound in the blood between about 40 to 4,000 ng/mL, and maintaining such concentration during a period of about 4 to 12 hours following administration.
  • the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 75 to 2,000 ng/mL and maintain that concentration during a period of about 4 to 12 hours from the time of administration.
  • the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 100 to 1,000 ng/mL following administration.
  • the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 100 to 500 ng/mL over a period of about 12 to 24 hours from the time of administration. In some embodiments, the dose size and frequency are selected to achieve a C max , plasma level of
  • Compound A that is at least about 500 ng/mL and does not exceed about 10,000 ng/mL.
  • Compound A is administered orally, intravenously,
  • Compound A is administered orally in a dose of about 1 mg, 3 mg, 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, 150 mg, 200 mg, or 300 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day. In other embodiments, it is administered orally in a dose of about 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, or 150 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day.
  • Compound A is administered by inhalation in a dose of about 0.1 mg, 0.3 mg, 0.5 mg, 1 mg, 3 mg, 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, 150 mg, 200 mg, or 300 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day.
  • compositions inhalation in a dose of about 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, or 150 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day.
  • Compound A may be administered prior to the onset of respiratory distress or an asthma attack.
  • Compound A may be administered prior to the onset of respiratory distress or an asthma attack.
  • Compound A may be
  • Compound A may be administered following respiratory distress or an asthma attack.
  • the method comprises administering to a patient, in addition to Compound A, a therapeutically effective amount of at least one therapeutic agent selected to treat the cancer in the patient.
  • Compound A may be combined with one or more other active therapeutic agents in a unitary dosage form for simultaneous or sequential administration to a patient.
  • the combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
  • co-administration of Compound A with one or more other active therapeutic agents generally refers to simultaneous or sequential administration of Compound A and one or more other active therapeutic agents, such that therapeutically effective amounts of Compound A and one or more other active therapeutic agents are both present in the body of the patient.
  • Compound A and therapeutic agent(s) are not necessarily both present in the body of the patient but the particular dosing schedule Compound A and therapeutic agents results in synergistic effects.
  • Co-administration includes administration of unit dosages of Compound A before or after administration of unit dosages of one or more other active therapeutic agents; for example, administration of Compound A within seconds, minutes, hours or days of the administration of one or more other active therapeutic agents.
  • a unit dose of Compound A can be administered first, followed within seconds, minutes, hour or days by administration of a unit dose of one or more other active therapeutic agents.
  • a unit dose of one or more other therapeutic agents can be administered first, followed by administration of a unit dose of
  • Compound A within seconds, minutes, hours or days. In some cases, it may be desirable to administer a unit dose of Compound A first, followed, after a period of hours (e.g. , 1 to 12 hours), by administration of a unit dose of one or more other active therapeutic agents. In other cases, it may be desirable to administer a unit dose of one or more other active therapeutic agents first, followed, after a period of hours (e.g. , 1 to 12 hours), by administration of a unit dose of Compound A. In some cases, it may be desirable to administer a unit dose of Compound A first, followed, after a period of days (e.g. , 1 to 14 days), by administration of a unit dose of one or more other active therapeutic agents.
  • the dosing regimen may involve alternating administration of Compound A and therapeutic agent over a period of several days, weeks, or months.
  • the combination therapy may provide "synergy” and "synergistic effect", i.e. , the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. , in separate tablets, pills or capsules, or by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e., serially.
  • the therapeutic agent is selected from the following group consisting of theophylline, nedocromil sodium, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, a beta 2 agonist, an antihistamine, an anticholinergic, allergen immunotherapy, albuterol, levalbuterol, pirbutrol, ipratropium bromide, methylprednisolone, prednisolone, prednisone, beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone acetonide, salmeterol, formoterol, montelukast, zafirlukast, mometasone, ciclesonide, alfuzosin, ephedrine, metaproterenol, phenylpropanolamine, pseudoephedrine, zileuton, omalizumab, and a combination
  • the method further comprises obtaining a biological sample from the subject; and analyzing the biological sample with an analytical procedure selected from the group consisting of blood chemistry analysis, chromosomal translocation analysis, needle biopsy, fluorescence in situ hybridization, laboratory biomarker analysis, immunohistochemistry staining method, flow cytometry, genetic analysis, or a combination thereof. Analysis may provide information about which patients may benefit from therapy, regression or progression of the tumor, an appropriate duration of the treatment, and is useful for determining dosages to administer, for adjusting dosages during a treatment cycle, and for deciding whether to continue or discontinue the treatments.
  • the subject may be any mammal, including human and non-human such as dogs.
  • the subject is a healthy person.
  • the subject is a patient having a disease or a condition related to PI3K-mediated disorders.
  • the subject is a patient having asthma.
  • the method described herein comprises administering to a subject Compound A described herein, in combination with a therapy used to treat a disease or a condition related to PI3K-mediated disorders. In one embodiment, the method described herein comprises administering to a subject Compound A described herein, in combination with a therpay used to treat asthma.
  • the "therapy" used to treat a disease ⁇ e.g., asthma) or a condition related to PI3K-mediated disorders is any well-known or experimental form of treatment used to treat asthma or a condition related to PI3K-mediated disorders that does not include the use of Compound A.
  • the combination of Compound A with a conventional or experimental therapy used to treat a disease ⁇ e.g., asthma) or a condition related to PI3K-mediated disorders provides beneficial and/or desirable treatment results superior to results obtained by treatment without the combination.
  • the therapies used to treat asthma or a condition related to PI3K-mediated disorders are well-known to a person having ordinary skill in the art and are described in the literature.
  • the combination method provides for Compound A administered simultaneously or during the period of administration of the therapy.
  • the combination method provides for Compound A administered prior to or after the administration of the therapy. The exact details regarding the administration of the combination may be determined experimentally. The refinement of sequence and timing of administering Compound A with a selected therapy will be tailored to the individual subject, the nature of the condition to be treated in the subject, and generally, the judgment of the attending practitioner.
  • the compounds described herein may be prepared in the form of prodrugs, i.e., protected forms which release the compounds described herein after administration to the subject.
  • the protecting groups are hydrolyzed in body fluids such as in the bloodstream thus releasing the active compound or are oxidized or reduced in vivo to release the active compound.
  • a discussion of prodrugs is found in Smith and Williams Introduction to the Principles of Drug Design, Smith, H.J.; Wright, 2 nd ed., London (1988).
  • a compound described herein can be administered as the neat chemical, but it is typically preferable to administer the compound in the form of a pharmaceutical composition or formulation.
  • pharmaceutical compositions that comprise Compound A and a biocompatible pharmaceutical carrier, adjuvant, or vehicle.
  • the composition can include the agent as the only active moiety or in combination with other agents, such as oligo- or polynucleotides, oligo- or polypeptides, drugs, or hormones mixed with excipient(s) or other pharmaceutically acceptable carriers. Carriers and other ingredients can be deemed pharmaceutically acceptable insofar as they are compatible with other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions are formulated to contain suitable pharmaceutically acceptable carriers, and can optionally comprise excipients and auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically.
  • the administration modality will generally determine the nature of the carrier.
  • formulations for parenteral administration can comprise aqueous solutions of the active compounds in water-soluble form.
  • Carriers suitable for parenteral administration can be selected from among saline, buffered saline, dextrose, water, and other physiologically compatible solutions.
  • Preferred carriers for parenteral administration are physiologically compatible buffers such as Hank's solution, Ringer' s solution, or physiologically buffered saline.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art.
  • the formulation can include stabilizing materials, such as polyols (e.g. , sucrose) and/or surfactants (e.g. , nonionic surfactants), and the like.
  • formulations for parenteral use can comprise dispersions or suspensions of the active compounds prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, and synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxy-methylcellulose, sorbitol, or dextran.
  • the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Aqueous polymers that provide pH- sensitive solubilization and/or sustained release of the active agent also can be used as coatings or matrix structures, e.g. , methacrylic polymers, such as the EUDRAGITTM series available from Rohm America Inc. (Piscataway, N.J.).
  • Suspensions can contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethlyene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, gum tragacanth, and mixtures thereof.
  • suspending agents such as ethoxylated isostearyl alcohols, polyoxyethlyene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, gum tragacanth, and mixtures thereof.
  • Liposomes containing the active agent also can be employed for parenteral
  • Liposomes generally are derived from phospholipids or other lipid substances.
  • the compositions in liposome form also can contain other ingredients, such as stabilizers,
  • lipids include phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods of forming liposomes are known in the art. See, e.g. , Prescott (Ed.), METHODS IN CELL BIOLOGY, Vol. XIV, p. 33, Academic Press, New York (1976).
  • compositions comprising the agent in dosages suitable for nasal or instillable administration can be formulated using purified aqueous solutions of the active compound with preservative agents such as isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes.
  • the pharmaceutical compositions disclosed herein may be administered into the respiratory system either by inhalation, respiration, nasal administration, or intrapulmonary instillation (into the lungs) of a subject by any suitable means, and they are preferably administered by generating an aerosol or spray comprised of powdered or liquid nasal, intrapulmonary, respirable, or inhalable particles.
  • respirable or inhalable particles comprising the active compound are inhaled by the subject, i.e. by inhalation or by nasal administration or by instillation into the respiratory tract or the lung itself.
  • the formulation may comprise respirable or inhalable liquid or solid particles of the active compound that, in accordance with the present application, include respirable or inhalable particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and continue into the bronchi and alveoli of the lungs.
  • particles may range from about 0.05, about 0.1, about 0.5, about 1, or about 2 to about 4, about 6, about 8, or about 10 ⁇ in diameter. In a particular embodiment, particles range from about 0.5 to less than about 5 ⁇ in diameter. Particles of non-respirable size that are included in an aerosol or spray tend to deposit in the throat and be swallowed. Thus, the quantity of non-respirable particles in the aerosol is preferably minimized.
  • a particle size in the range of about 8, about 10, about 20, or about 25 to about 35, about 50, about 100, about 150, about 250, or about 500 ⁇ diameter is preferred to ensure retention in the nasal cavity or for instillation and direct deposition into the lung.
  • Liquid formulations may be squirted into the respiratory tract (i.e. nose) and the lung, particularly when administered to newborns and infants.
  • Liquid pharmaceutical compositions of active compound for producing an aerosol may be prepared by combining the active ingredient with a stable vehicle, such as sterile pyrogen-free water.
  • Solid particulate compositions containing respirable dry particles of micronized active compound maybe prepared by grinding dry active compound with a mortar and pestle, and then passing the micronized composition through a 400 mesh screen to break up or separate out large agglomerates.
  • a solid particulate composition comprised of the active compound may optionally contain a dispersant that serves to facilitate the formulation of an aerosol.
  • a suitable dispersant is lactose, which may be blended with the active compound in any suitable ratio, e.g., a 1 to 1 ratio by weight.
  • Aerosols of liquid particles comprising the active compound may be produced by any suitable means, such as with a nebulizer.
  • Nebulizers are commercially available devices that transform solutions or suspensions of the active ingredient into a therapeutic aerosol mist either by means of acceleration of a compressed gas, typically air or oxygen, through a narrow venture orifice or by means of ultrasonic agitation.
  • Suitable compositions for use in a nebulizer consist of the active ingredient in liquid carrier, the active ingredient comprising up to 40% w/w composition, but preferably less than 20% w/w, and the carrier being typically water or a dilute aqueous alcoholic solution, preferably made isotonic with body fluids by the addition of, for example, sodium chloride.
  • Optional additives include preservatives if the composition is not prepared sterile, for example, methyl hydroxybenzoate, anti- oxidants, flavoring agents, volatile oils, buffering agents, and surfactants.
  • Aerosols of solid particles comprising the active compound may likewise be produced with any solid particular medicament aerosol generator. Aerosol generators for administering solid particular medicaments to a subject produce particles that are respirable, as explained above, and generate a volume of aerosol containing a predetermined metered dose of a medicament at a rate suitable for human administration. Examples of such aerosol generators include metered dose inhalers and insufflators.
  • composition may be delivered with any delivery device that generates liquid or solid particulate aerosols, such as aerosol or spray generators. These devices produce respirable particles, as explained above, and generate a volume of aerosol or spray containing a
  • an insufflator which are suitable for administration of finely comminuted powders.
  • the powder e.g., a metered dose of the composition effective to carry out the treatments described herein, is contained in a capsule or cartridge.
  • capsules or cartridges are typically made or gelatin, foil, or plastic, and may be pierced or opened in situ, and the powder delivered by air drawn through the device upon inhalation or by means or a manually operated pump.
  • the composition employed in the insufflator may consist either solely of the active compound or of a powder blend comprising the active compound, typically comprising from 0.01 to 100% w/w of the composition.
  • the composition contains the active ingredient in an amount of about 0.01% w/w, about 1% w/w, or about 5% w/w to about 20% w/w, about 40% w/w, or about 99.99% w/w.
  • Other ingredients, and other amounts of the agent are also suitable within the confines of this application.
  • the composition is delivered by a nebulizer.
  • the nebulizer can use any pharmaceutically acceptable carrier, such as a weak saline solution.
  • the nebulizer is the means by which the powder pharmaceutical composition is delivered to the target of the patients or subjects in the airways.
  • the composition is delivered using suspension metered dose inhalation (MDI) formulation.
  • MDI suspension metered dose inhalation
  • HFA hydrofluoroalkane
  • the HFA propellants contain 100 parts per million (ppm) or less of water.
  • the delivery device comprises a dry powder inhalator (DPI) that delivers single or multiple doses of the composition.
  • DPI dry powder inhalator
  • the single dose inhalator may be provided as a disposable kit that is sterilely preloaded with enough formulation for one application.
  • the inhalator may be provided as a pressurized inhalator, and the formulation in a capsule or cartridge that may be pierced or opened.
  • the kit may optionally also comprise in a separate container an agent such as other therapeutic compounds, excipients, surfactants (intended as therapeutic agents as well as formulation ingredients), antioxidants, flavoring and coloring agents, fillers, volatile oils, buffering agents, dispersants, surfactants, bulking agents, propellants, and preservatives, among other suitable additives for the different formulations.
  • compositions comprising the agent in dosages suitable for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art.
  • the preparations formulated for oral administration can be in the form of tablets, pills, capsules, cachets, dragees, lozenges, liquids, gels, syrups, slurries, elixirs, suspensions, or powders.
  • pharmaceutical preparations for oral use can be obtained by combining the active compounds with a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Oral formulations can employ liquid carriers similar in type to those described for parenteral use, e.g., buffered aqueous solutions, suspensions, and the like.
  • Preferred oral formulations include tablets, dragees, and gelatin capsules. These preparations can contain one or excipients, which include, without limitation:
  • diluents such as sugars, including lactose, dextrose, sucrose, mannitol, or sorbitol
  • binders such as magnesium aluminum silicate, starch from corn, wheat, rice,
  • cellulose materials such as methylcellulose, hydroxypropylmethyl cellulose, and
  • sodium carboxymethylcellulose polyvinylpyrrolidone
  • gums such as gum arabic and gum tragacanth
  • proteins such as gelatin and collagen
  • disintegrating or solubilizing agents such as cross-linked polyvinyl pyrrolidone,
  • starches agar, alginic acid or a salt thereof, such as sodium alginate, or effervescent compositions; e) lubricants, such as silica, talc, stearic acid or its magnesium or calcium salt, and polyethylene glycol;
  • colorants or pigments e.g. , to identify the product or to characterize the quantity
  • ingredients such as preservatives, stabilizers, swelling agents, emulsifying agents, solution promoters, salts for regulating osmotic pressure, and buffers.
  • the pharmaceutical composition comprises at least one of the materials from group (a) above, or at least one material from group (b) above, or at least one material from group (c) above, or at least one material from group (d) above, or at least one material from group (e) above.
  • the composition comprises at least one material from each of two groups selected from groups (a)-(e) above.
  • Gelatin capsules include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating such as glycerol or sorbitol.
  • Push-fit capsules can contain the active ingredient(s) mixed with fillers, binders, lubricants, and/or stabilizers, etc.
  • the active compounds can be dissolved or suspended in suitable fluids, such as fatty oils, liquid paraffin, or liquid polyethylene glycol with or without stabilizers.
  • Dragee cores can be provided with suitable coatings such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • suitable coatings such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • the pharmaceutical composition can be provided as a salt of the active agent. Salts tend to be more soluble in aqueous or other protic solvents than the corresponding free acid or base forms.
  • Pharmaceutically acceptable salts are well known in the art. Compounds that contain acidic moieties can form pharmaceutically acceptable salts with suitable cations. Suitable pharmaceutically acceptable cations include, for example, alkali metal (e.g. , sodium or potassium) and alkaline earth (e.g. , calcium or magnesium) cations.
  • Compound A may form pharmaceutically acceptable acid addition salts with suitable acids.
  • suitable acids for example, Berge, et ah, describe pharmaceutically acceptable salts in detail in J. Pharm. ScL, 66: 1 (1977).
  • the salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting Compound A with a suitable acid.
  • Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorolsulfonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate (isothionate), lactate, maleate, methanesulfonate or sulfate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate or hydrogen phosphate, glutamate, bicarbonate, p
  • compositions comprising a compound described herein formulated in a pharmaceutical acceptable carrier can be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Accordingly, there also is contemplated an article of manufacture, such as a container comprising a dosage form of a compound described herein and a label containing instructions for use of the compound. Kits are also contemplated for the compounds and methods described herein. For example, the kit can comprise a dosage form of a pharmaceutical
  • the kit comprises Compound A, and at least one therapeutic agent disclosed herein. In certain embodiments, the kit may further comprise at least one
  • compositions comprising Compound A can be administered to the subject by any conventional method, including parenteral and enteral techniques. Parenteral
  • administration modalities include those in which the composition is administered by a route other than through the gastrointestinal tract, for example, intravenous, intraarterial, intraperitoneal, intramedullarly, intramuscular, intraarticular, intrathecal, and intraventricular injections.
  • Enteral administration modalities include, for example, oral (including buccal and sublingual) and rectal administration.
  • Transepithelial administration modalities include, for example, transmucosal administration and transdermal administration.
  • Transmucosal administration includes, for example, enteral administration as well as nasal, inhalation, and deep lung administration; vaginal administration; and rectal administration.
  • Transdermal administration includes passive or active transdermal or transcutaneous modalities, including, for example, patches and iontophoresis devices, as well as topical application of pastes, salves, or ointments.
  • Parenteral administration also can be accomplished using a high-pressure technique, e.g., POWDERJECTTM.
  • Surgical techniques include implantation of depot (reservoir) compositions, osmotic pumps, and the like.
  • a preferred route of administration for treatment of inflammation can be local or topical delivery for localized disorders such as arthritis, or systemic delivery for distributed disorders, e.g., intravenous delivery for reperfusion injury or for systemic conditions such as septicemia.
  • administration can be accomplished by inhalation or deep lung administration of sprays, aerosols, powders, and the like.
  • the characteristics of the agent itself and the formulation of the agent can influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agent.
  • Such pharmacokinetic and pharmacodynamic information can be collected through preclinical in vitro and in vivo studies, later confirmed in humans during the course of clinical trials.
  • a therapeutically effective dose can be estimated initially from biochemical and/or cell-based assays.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the "therapeutic index," which typically is expressed as the ratio LD 50 /ED 50 .
  • Compounds that exhibit large therapeutic indices i.e., the toxic dose is substantially higher than the effective dose) are preferred.
  • the data obtained from such cell culture assays and additional animal studies can be used in formulating a range of dosage for human use.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • any effective administration regimen regulating the timing and sequence of doses can be used.
  • Doses of the agent preferably include pharmaceutical dosage units comprising an effective amount of the agent.
  • effective amount refers to an amount sufficient to modulate the PDKa-sparing activity or any combination of ⁇ , ⁇ , and ⁇ expression or activity and/or derive a measurable change in a physiological parameter of the subject through administration of one or more of the pharmaceutical dosage units.
  • Effective amount can also refer to the amount required to ameliorate a disease, condition or disorder in a subject.
  • Suitable dosage ranges for Compound A may vary according to these considerations, but in general, Compound A may be administered in the range of about 10.0 ⁇ g/kg to 15 mg/kg of body weight; about 1.0 ⁇ g/kg to 10 mg/kg of body weight, or about 0.5 mg/kg to 5 mg/kg of body weight.
  • the dosage range is from about 700 ⁇ g to 1050 mg; about 70 ⁇ g to 700 mg; or about 35mg to 350 mg per dose, and two or more doses may be administered per day. Dosages may be higher when Compound A is administered orally or transdermally as compared to, for example, IV administration.
  • the treatment of asthma comprises oral administration of up to about 750 mg/day of Compound A.
  • the reduced toxicity of this compound permits the therapeutic administration of relatively high doses.
  • the reduced toxicity of Compound A permits the therapeutic administration of relatively high doses.
  • Compound A is administered orally, in three to five doses per day, using about 20 to 150 mg per dose for a total daily dose between about 60 to 750 mg.
  • the total daily dose is between about 100 to 500 mg, and in some embodiments the normalized daily dosage (adjusted for subject' s body weight) is up to about 60 mg per kg of the treated subject's body weight.
  • Compound A may be administered as a single bolus dose, a dose over time, as in IV or transdermal administration, or in multiple dosages.
  • a dosage may be delivered over a prolonged period of time, and may be selected or adjusted to produce a desired plasma level of the active compound.
  • the desired plasma level is at least about 1 micromolar, or at least about 10 micromolar.
  • Compound A When Compound A is administered orally, it is preferably administered one time per day or in two or more doses per day. In some embodiments, three doses per day are administered. In some embodiments four doses per day are administered.
  • Dosing may be continued for one day or for multiple days, such as about 7 days. In some embodiments, daily dosing is continued for about 14 days or about 28 days. In some embodiments,
  • dosing is continued for about 28 days and is then discontinued for about 7 days; the efficacy of the treatment can be assessed during the break, when treatment with Compound A has been stopped, and if the assessment shows that the treatment is achieving a desired effect, another cycle of about 7 to 28 days of treatment with Compound A can be initiated.
  • a suitable dose can be calculated according to body weight, body surface area, or organ size.
  • the final dosage regimen will be determined by the attending physician in view of good medical practice, considering various factors that modify the action of drugs, e.g. , the agent' s specific activity, the identity and severity of the disease state, the responsiveness of the patient, the age, condition, body weight, sex, and diet of the patient, and the severity of the disease related to PI3K-mediated disorders (e.g. asthma), a condition related to PI3K-mediated disorders, or any infection.
  • Additional factors that can be taken into account include comorbidities, prior therapies, the time and frequency of administration, drug combinations, reaction sensitivities, and tolerance/response to therapy. Further refinement of the dosage appropriate for treatment involving any of the formulations mentioned herein is done routinely by the skilled practitioner without undue experimentation, especially in light of the dosage information and assays disclosed, as well as the pharmacokinetic data observed in human clinical trials. Appropriate dosages can be ascertained through use of established assays for determining concentration of the agent in a body fluid or other sample together with dose -response data.
  • the frequency of dosing will depend on the pharmacokinetic parameters of the agent and the route of administration. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Accordingly, the pharmaceutical compositions can be administered in a single dose, multiple discrete doses, continuous infusion, sustained release depots, or combinations thereof, as required to maintain desired minimum level of the agent. Short- acting pharmaceutical compositions (i.e. , short half-life) can be administered once a day or more than once a day (e.g. , two, three, or four times a day). Long acting pharmaceutical compositions might be administered every 3 to 4 days, every week, or once every two weeks to 12 weeks.
  • Pumps such as subcutaneous, intraperitoneal, or subdural pumps, can be preferred for continuous infusion.
  • Subjects that will respond favorably to the methods described herein include medical and veterinary subjects generally, including human patients. Among other subjects for whom the methods described herein are useful are cats, dogs, large animals, avians such as chickens, and the like. In general, any subject who would benefit from Compound A is appropriate for
  • the reaction was stirred at -15°C for 30 minutes, then added with 2-amino-6-chloro-N-phenylbenzamide (3) in THF (1.0 mL). The reaction mixture was slowly warmed to 21°C. When about 10% conversion was observed, the reaction mixture was warmed to 60°C for 30 minutes. The reaction mixture was poured into ethyl acetate (150 mL), and washed with water (50 mL) twice and brine (30 mL). The organic layer was dried over sodium sulfate, filtered, and rotary evaporated to remove the solvent. Chromatography was performed using 90 g silica gel with 1:4 (v/v) ethyl acetate: hexane to yield white crystals.
  • reaction mixture was diluted with toluene (100 mL), and solvents were removed by rotary evaporation. The remaining solid was dissolved in ethyl acetate (300 mL), and washed with sodium bicarbonate (100 mL), water (100 mL), and brine (100 mL). The organic layer was dried over magnesium sulfate, filtered, and rotary evaporated to remove solvent. Chromatography was performed using 40 g silica gel with chloroform and a slow gradient to 10% methanol (containing 10% ammonium hydroxide)-chloroform to yield (S)-2-(l- aminoethyl)-5-chloro-3-phenylquinazolin-4(3H)-one (5).
  • Compound A is the optically active form that predominantly includes the (S)-enantiomer.
  • Compound A(S) was formulated for IV infusion in a sterile solution of 5% ethanol, 75% PEG 400, and 20% citrate buffer (pH 3.0) at 0.09 mg/mL.
  • Compound A(S) was formulated for oral dosing (PO) in 5% ethanol, 87% PEG 400, 4% solutol, and 4% water at 0.23 mg/mL.
  • Each dosing group consisted of three Sprague Dawley rats. At dosing, the animals weighed between 0.26 and 0.27 kg.
  • test article was administered to the rats via infusion over 30-minutes at 5.0 mL/kg for a dose of 0.44 mg/kg.
  • test article was administered to the rats via oral gavage at 10.0 mL/kg for a dose of 1.52 mg/kg.
  • Blood samples (approximately 0.35 mL each) were taken at specified time points after dosing from each animal, as shown in Tables 1 and 2. The blood samples were collected into VACUTAINER tubes containing EDTA-K 2 as the anti-coagulant and were immediately placed on wet ice pending centrifugation for plasma.
  • Table 2 Plasma concentration-time data of Compound A(S) in plasma after an oral of Compound A(S) at 1.52 mg/kg in rats.
  • Rats 4 and 5 were observed with diarrhea at 6, 8, and 12 hours postdose.
  • Rat 6 was observed with very soft stool at 4 and 6 hours postdose, and with diarrhea at 8 and 12 hours postdose.
  • the column used was a HyPurity C 18 HLPC column (30 X 2.1 mm, 5 ⁇ ) (Thermo-Hypersil, Part #22105-032130).
  • Mobile phase A contained 1% acetonitrile in 10 mM ammonium formate aqueous solution
  • mobile phase B contained 80% acetonitrile in 10 mM ammonium formate aqueous solution.
  • a Cohesive LX-2 multiplex with two identical Agilent 1200 series binary pumps (P/N G1312A Bin Pump) was used for elution.
  • An HTS PAL autosampler was used (LEAP Technologies, Carrboro, NC).
  • FIG. 1 A concentration-time profile of Compound A(S) in plasma following the 30-minute IV infusion at 0.44 mg/kg and the oral dose at 1.52 mg/kg in rats is shown in Figure 1.
  • Figures 2 and 3 show the individual rat concentration-time profile for IV and oral administration, respectively.
  • V ss (L/kg) 2.00 2.16 3.75 2.64 0.97
  • IC 50 IC 50 values for inhibiting PI3K isoforms were determined using in vitro
  • IC 50 values for Compounds A-G for various isoforms of PI3K are shown in Table 6.
  • EC '50 To analyze PI3K pi 105 and pi 10 ⁇ signaling, basophil activation was measured in isolated PBMC or whole blood using the Flow2 CAST kit according to the manufacture' s standardized methods (Buhlman Laboratories AG, Switzerland). Briefly, ⁇ ⁇ ⁇ was activated with anti-FCeRI and pi 10 ⁇ was activated with fMLP (2 ⁇ ) in the absence or presence of compounds. To monitor the basophil cell population and cellular activation, anti-CD63-FITC and anti-CCR3- PE antibodies were added to each sample. Cells were fixed and analyzed on a FC500MPL flow cytometer (Beckman Coulter, Brea, CA).
  • the EC 50 value for Compound A(S) in PI3K p 110 ⁇ was 1.0 nM.
  • the EC 50 value for Compound A(S) in PI3K pi 10 ⁇ was 50 nM.
  • the potency of Compound A(S) is greater for the ⁇ -isoform than for the ⁇ -isoform.

Abstract

Provided are methods that relate to a novel therapeutic strategy for the treatment of asthma. In particular, the method comprises administration of Compound A, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such compound admixed with at least one pharmaceutically acceptable excipient.

Description

COMPOSITIONS AND METHODS OF TREATING A DISEASE WITH (S)-4-AMINO-6- ((l-(5-CHLORO-4-OXO-3-PHENYL-3,4-DIHYDROQUINAZOLIN-2- YL)ETHYL)AMINO)PYRIMIDINE-5-CARBONITRILE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. provisional patent application Serial No.
61/594,868, filed February 3, 2012, the entire disclosure of which is incorporated herein by reference.
FIELD
[0002] The present application is in the field of therapeutics and medicinal chemistry. In particular, the present application concerns methods of treatment of asthma that include
administration of certain quinazolinone derivatives.
BACKGROUND
[0003] Cell signaling via 3 '-phosphorylated phosphoinositides has been implicated in a variety of cellular processes, e.g. , malignant transformation, growth factor signaling, inflammation, and immunity. The enzyme responsible for generating these phosphorylated signaling products, phosphatidylinositol 3 -kinase (PI 3 -kinase; PI3K), was originally identified as an activity associated with viral oncoproteins and growth factor receptor tyrosine kinases that phosphorylates phosphatidylinositol (PI) and its phosphorylated derivatives at the 3 '-hydroxyl of the inositol ring.
[0004] PI 3-kinase activation is believed to be involved in a range of cellular responses including cell growth, differentiation, and apoptosis. In some instances, PI3K participates in cellular pathways involved in hematological malignancy and solid tumor activation. For example, PI3K participates in a cellular pathway that has been implicated in the process of oncogenic
transformation and in promoting the growth, proliferation, and survival of various types of cancers, such as T-cell acute lymphoblastic leukemia.
[0005] The initial purification and molecular cloning of PI3-kinase revealed that it was a heterodimer consisting of p85 and pi 10 subunits. Four Class I PI3Ks have been identified and designated as PI3K α, β, δ, and γ isomers. Each isomer consists of a distinct pi 10 catalytic subunit and a regulatory subunit. Three catalytic subunits, pi 10a, pi 10β and pi 105, each interact with the same regulatory subunit, p85; whereas pi 10γ interacts with a distinct regulatory subunit, plOl. The expression of each PI3K isoform in human cells and tissues are also distinct.
[0006] Identification of the pi 105 isoform of PI-3-kinase is described in Chantry et ah, J. Biol. Chem., 272: 19236-41 (1997). It was observed that the human PI3K pi 105 isoform was expressed in a tissue-restricted fashion; for example, PI3K pi 105 expressed at high levels in lymphocytes and lymphoid tissues. This suggests that PI3K5 might play a role in the PI3-kinase-mediated signaling in the immune system. The pi 10β isoform of PI3K may also play a role in the PI3K- mediated signaling in certain cancers.
SUMMARY
[0007] The present application discloses compounds, compositions and methods related to treating a disease or a condition related to PI3K-mediated disorders. Such disease may include, for example, asthma. Unexpected effects on PI3K isomers have been found in the compounds disclosed herein. Provided is a compound having the structure of Compound A
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof. Provided are also all stereoisomeric forms, individual diastereoisomers and enantiomers, as well as racemic and non-racemic mixtures of Compound A or a pharmaceutically acceptable salt thereof.
[0008] In specific embodiments, the compound is the (S)-enantiomer, having the structure of Compound A(S):
Figure imgf000003_0002
or a pharmaceutically acceptable salt thereof. [0009] Provided is also a composition comprising a compound of any of the foregoing embodiments of Compound A, and at least one pharmaceutically acceptable excipient. In particular embodiments, the composition comprises a therapeutically effective amount of
Compound A for the treatment of asthma in a patient.
[0010] In some embodiments, the compound is a racemic mixture of the (R)- and (S)- enantiomers of Compound A. In other embodiments, the compound is optically active. In specific embodiments, the (S)-enantiomer of Compound A, having the structure of Compound A(S):
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof; is present in excess of Compound A(R)
Figure imgf000004_0002
[0011] In further embodiments, the compound is substantially free of Compound A(R). In some embodiments, the (S)-enantiomer of Compound A predominates over the (R)-enantiomer of Compound A by a molar ratio of at least 9: 1, at least 19: 1, at least 40: 1, at least 80: 1, at least 160: 1, or at least 320: 1.
[0012] The compound can also be described by its enantiomeric excess (e.e.). For instance, a compound with 95% (S)-isomer and 5% (R)-isomer will have an e.e. of 90%. In some
embodiments, the compound has an e.e. of at least 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%. In some of the foregoing embodiments, the compound is enantiomerically-enriched in the (S)- isomer of Compound A.
[0013] Provided is also a method of treating a disease or a condition related to PI3K-mediated disorders. In some embodiments, the disease or condition is characterized by histamine release (e.g. , allergic disorders). In certain embodiments, disorders that may be treated by the methods provided herein may include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, and emphysema. In one embodiment, the disease is asthma. In some embodiments, provided is a method of treating a disease or a condition related to PDK-mediated disorders comprising administering to a patient in need there of an effective amount of any of the foregoing compounds or compositions.
[0014] Specific embodiments provide a method of treating asthma in a patient, comprising administering to the patient Compou
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, including all stereoisomeric forms, individual diastereoisomers and enantiomers, as well as racemic and non-racemic mixtures of Compound A or a pharmaceutically acceptable salt thereof. Specific embodiments provide a method of treating asthma in a patient, comprising administering to the patient a pharmaceutical composition comprising Compound A or a pharmaceutically acceptable salt thereof, optionally admixed with at least one pharmaceutically acceptable excipient. In particular embodiments, the composition comprises a therapeutically effective amount of a compound of any of the foregoing embodiments of Compound A or a pharmaceutically acceptable salt thereof for the treatment of asthma in a patient.
[0015] In some embodiments, the composition comprises a racemic mixture of Compound A. In a specific embodiment, the composition comprises the (S)-enantiomer of Compound A, having the structure of Compound A(S):
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof, wherein Compound A(S) is present in excess of the (R)-enantiomer of Compound A, having the structure of Compound A(R)
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof.
[0016] In specific embodiments, the composition is substantially free of the (R)-enantiomer of Compound A.
[0017] In some of the foregoing embodiments, Compound A is administered at a dose of about 1 to 4,000 mg/day, about 2,000 to 4,000 mg/day, about 1 to 2,000 mg/day, about 1 to 1,000 mg/day, 10 to 500 mg/day, about 20 to 500 mg/day, about 50 to 300 mg/day, about 75 to 200 mg/day, or about 15-150 mg/day. In other embodiments, Compound A is administered at a dose of about 1 to 150 mg twice per day. In yet other embodiments, Compound A is administered at least twice daily. In certain embodiments, Compound A is administered intermittently or in intervals.
Depending on the treatment and the patient's condition, the interval may range from one, two, three, four, five, six and seven days. In one example, Compound A is administered for at least 21 days, and is then discontinued for at least 7 days. In another example, Compound A is
administered for about 21 days, and is then discontinued for about 7 days.
[0018] In some of the foregoing embodiments, the method further comprises reducing the level of PI3K5 activity in the patient in need thereof. In some of the foregoing embodiments, the method further comprises the PBKcc-sparing activity in the patient in need thereof.
[0019] In some of the foregoing embodiments, the method further comprises administering to a patient, in addition to Compound A, a therapeutically effective amount of at least one therapeutic agent selected to treat the PI3K-mediated condition in the patient. In some embodiments, the therapeutic agent is selected from the following group consisting of theophylline, nedocromil sodium, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, a beta 2 agonist, an
antihistamine, an anticholinergic, allergen immunotherapy, albuterol, levalbuterol, pirbutrol, ipratropium bromide, methylprednisolone, prednisolone, prednisone, beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone acetonide, salmeterol, formoterol, montelukast, zafirlukast, mometasone, ciclesonide, alfuzosin, ephedrine, metaproterenol, phenylpropanolamine,
pseudoephedrine, zileuton, omalizumab, and a combination thereof. BRIEF DESCRIPTION OF THE FIGURES
[0020] Figure 1. Concentration-time profiles of Compound A(S) in plasma following a 30- minute IV infusion at 0.44 mg/kg and an oral dose at 1.52 mg/kg in rats (mean + SD, n=3).
[0021] Figure 2. Concentration-time profiles of Compound A(S) in plasma following a 30- minute IV infusion at 0.44 mg/kg in rats. Data is shown for individual rats.
[0022] Figure 3. Concentration-time profiles of Compound A(S) in plasma following an oral dose at 1.52 mg/kg in rats. Data is shown for individual rats.
DETAILED DESCRIPTION
[0023] Unless otherwise described, all terms of art, notations and other scientific terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this present disclosure pertains. In some cases, terms with commonly understood meanings are described herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. Many of the techniques and procedures described or referenced herein are well understood and commonly employed using conventional methodology by those skilled in the art. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer described protocols and/or parameters unless otherwise noted.
[0024] The discussion of the general methods given herein is intended for illustrative purposes only. Other alternative methods and embodiments will be apparent to those of skill in the art upon review of this disclosure.
[0025] A group of items linked with the conjunction "or" should not be read as requiring mutual exclusivity among that group, but rather should also be read as "and/or" unless expressly stated otherwise. Although items, elements, or components of the present disclosure may be described or claimed in the singular, the plural is contemplated to be within the scope thereof unless limitation to the singular is explicitly stated.
[0026] Throughout the specification, various publications, patents, and patent applications are disclosed. All of these are hereby incorporated by reference in their entirety for all purposes. Compound A
[0027] Provided are novel methods to treat a disease or a condition related to PI3K-mediated disorders using Compound A. In one embodiment, the disease is asthma. One aspect provides a compound having the structure of C
Figure imgf000008_0001
or a pharmaceutically acceptable salt thereof, including all stereoisomeric forms, individual diastereoisomers and enantiomers, as well as racemic and non-racemic mixtures of Compound A or a pharmaceutically acceptable salt thereof. Another aspect provides a pharmaceutical composition comprising Compound A or a pharmaceutically acceptable salt thereof, optionally admixed with at least one pharmaceutically acceptable excipient.
[0028] In specific embodiments, the compound is the (S)-enantiomer, having the structure of Compound A(S):
Figure imgf000008_0002
[0029] Provided is also Compound A in which from 1 to n hydrogens attached to a carbon atom may be replaced by deuterium, in which n is the number of hydrogens in the molecule. Such compounds may exhibit increased resistance to metabolism, and thus may be useful for increasing the half life of Compound A when administered to a mammal. See, e.g., Foster, "Deuterium Isotope Effects in Studies of Drug Metabolism", Trends Pharmacol. Sci., 5(12):524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium.
[0030] Compositions comprising Compound A may include racemic mixtures or mixtures containing an enantiomeric excess of one enantiomer or single diastereomers or diastereomeric mixtures. All such isomeric forms of these compounds are expressly included herein the same as if each and every isomeric form were specifically and individually listed.
[0031] In some embodiments, Compound A and compositions thereof for use in the methods described herein may be optically active. Compound A has a single chiral center in the noncyclic linking group between the quinazolinone moiety and the pyrimidine moiety. In some
embodiments, the preferred enantiomer of Compound A is the (S)-enantiomer depicted above. Optically active forms of Compound A may include predominantly the (S)-enantiomer, although it may also include the (R)-enantiomer of Compound A as a minor component. For clarity, where a dosage of Compound A is described herein, the dosage refers to the weight of Compound A, including each enantiomer that may be present. Thus, a dosage of 100 mg of Compound A, for example, refers to the weight of the mixture of enantiomers rather than the weight of the (S)- enantiomer specifically. It could, for example, refer to 100 mg of a 9: 1 mixture of S- and (R)- enantiomers, which would contain about 90 mg of the (S)-enantiomer, or to 100 mg of a 19: 1 mixture of S- and (R)-enantiomers, which would contain about 95 mg of the (S)-enantiomer.
[0032] Compound A may be synthesized in optically active form, or it may be prepared in racemic form (containing equal amounts of R- and (S)-isomers), and then the isomers may be separated. Scheme 1 depicts a chiral synthesis of Compound A that provides the (S) -enantiomer in very high optical purity. In some embodiments, the enantiomeric (R)-isomer of Compound A may be excluded. In other embodiments, the methods may be practiced with mixtures of R- and (S)- isomers. In yet other embodiments, the methods may be practiced with mixtures of R- and (S)- isomers, in which the (S)-isomer is the major component of the mixture. In embodiments where the (S)-isomer is the major component of the mixture, such mixture may contain no more than about 10% of the R isomer, meaning the ratio of S- to (R)-isomers is at least about 9: 1, and in other embodiments, less than 5% of the (R)-isomer, meaning the ratio of S- to (R)-enantiomers is at least about 19: 1. In some embodiments, the (S)-enantiomer predominates over the (R)-enantiomer by a molar ratio of at least 40: 1, at least 80: 1, at least 160: 1, or at least 320: 1.
[0033] Compound A can also be described by its enantiomeric excess (e.e.). For instance, a compound characterized by 95% (S)-isomer and 5% (R)-isomer will have an e.e. of 90%. In some embodiments, Compound A has an e.e. of at least 60%, at least 75%, at least 80%, at least 90%, at least 95%, at least 98%, or at least 99%.
[0034] In some of the foregoing embodiments, the compound is enantiomerically-enriched in the (S)-isomer of Compound A. In certain embodiments, the compound may be enriched with the (S)- enantiomer shown here:
Figure imgf000010_0001
and preferably, is at least 90% (S)-enantiomer of Compound A, containing no more than about 10% of the enantiomeric (R)-enantiomer of Compound A.
[0035] In certain embodiments, Compound A is primarily composed of the (S)-enantiomer of Compound A, wherein this isomer comprises at least 66-95%, or about 85-99% of the (S)- enantiomer, in excess over any (R)-enantiomer present. In certain embodiments, Compound A is at least 95% of the (S)-enantiomer of Compound A. In one embodiment, Compound A is 100% of the (S)-enantiomer. In the additional embodiment, the sample of Compound A is at least 99% the (S)-enantiomer, with less than 1% of the (R)-enantiomer.
[0036] The compounds depicted herein may be present as salts even if the salts are not depicted. In some embodiments, the salts of the compounds disclosed in the application are pharmaceutically acceptable salts.
Methods of Treatment
[0037] The methods described herein are useful to treat a disease or a condition related to PI3K- mediated disorders. In one embodiment, the disease is asthma.
[0038] In one aspect, the application discloses compounds capable of inhibiting the biological activity of human PI3K5. In another aspect, the application provides compounds that inhibit PI3K5 selectively compared to the other PI3K isoforms. Still in another aspect, the application provides a method of selectively modulating human PI3K5 activity, and thereby treating diseases mediated by PI3K5 dysfunction.
[0039] In one aspect, the application discloses a method of treating a disease or a condition related to PI3K-mediated disorders. In some embodiments, the disease or condition is
characterized by histamine release (e.g. , allergic disorders). In certain embodiments, disorders that may be treated by the methods described herein may include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, and emphysema. In some embodiments, the disease is asthma. In certain embodiments, the method of treating a PI3K-mediated disorder or condition comprises administering to a patient in need there of an effective amount of any of the foregoing compounds or compositions. [0040] In one aspect, the application discloses a method of disrupting leukocyte function comprising contacting leukocytes with a compound that selectively inhibits phosphatidylinositol 3- kinase delta (PI3K5) activity in the leukocytes. The leukocytes can comprise cells selected from the group consisting of neutrophils, B lymphocytes, T lymphocytes, and basophils.
[0041] For example, in cases wherein the leukocytes comprise neutrophils, the method comprises disrupting at least one neutrophil function selected from the group consisting of stimulated superoxide release, stimulated exocytosis, and chemotactic migration. Preferably, the method does not substantially disrupt bacterial phagocytosis or bacterial killing by the neutrophils. In cases wherein the leukocytes comprise B lymphocytes, the method comprises disrupting proliferation of the B lymphocytes or antibody production by the B lymphocytes. In cases wherein the leukocytes comprise T lymphocytes, the method comprises disrupting proliferation of the T lymphocytes. In cases wherein the leukocytes comprise basophils, the method comprises disrupting histamine release by the basophils.
[0042] In one aspect, the application discloses methods of inhibiting neutrophil functions, without substantially inhibiting phagocytosis of bacteria. Neutrophil functions suitable for inhibition according to the present method include any function mediated by PI3K5 activity or expression. Such functions include, without limitation, stimulated superoxide release, stimulated exocytosis or degranulation, chemotactic migration, adhesion to vascular endothelium (e.g., tethering/rolling of neutrophils, triggering of neutrophil activity, and/or latching of neutrophils to endothelium), transmural diapedesis, or emigration through the endothelium to peripheral tissues. In general, these functions can be collectively termed "inflammatory functions," as they are typically related to neutrophil response to inflammation. The inflammatory functions of neutrophils can be distinguished from the bacterial killing functions exhibited by these cells, e.g., phagocytosis and killing of bacteria. Accordingly, the application further includes methods of treating disease states in which one or more of the inflammatory functions of neutrophils are abnormal or undesirable.
[0043] The compounds of the present application may be used to inhibit an endogenous immune response stimulated by at least one endogenous factor without substantially inhibiting an exogenous immune response stimulated by at least one exogenous factor as disclosed in US 2005/0043239 Al. The compounds of the present application may also be used to inhibit an endogenous immune response stimulated by at least one endogenous factor without substantially inhibiting immune responsiveness, as disclosed in US 2005/0043239 Al. Accordingly, the compounds of the application advantageously permit treatment of conditions associated with an undesirable endogenous immune response stimulated by at least one endogenous factor without compromising the ability to fight infection.
[0044] The compounds of the present application may also be used to inhibit leukocyte accumulation as disclosed in US 2005/0054614 Al. The compounds may also be used to inhibit leukocyte tethering to endothelial cells and to inhibit leukocyte transmigration into inflamed tissue, as disclosed in US 2005/0043239 Al . Accordingly, the compounds of the application
advantageously permit treatment of individuals having an inflammatory condition where leukocytes are found to be at the site of insult or inflamed tissue.
[0045] It further has been established that PI3K5 plays a role in the stimulated proliferation of lymphocytes, including B cells and T cells. Moreover, PI3K5 appears to play a role in stimulated secretion of antibodies by B cells. Selective PI3K5 inhibitor compounds of the present application have been employed to establish that these phenomena can be abrogated by inhibition ofPI3K5. Thus, the present application includes methods for inhibiting lymphocyte proliferation, or for inhibiting antibody production by B lymphocytes. Other methods enabled by the present application include methods of treating disease states in which one or more of these lymphocyte functions are abnormal or undesirable.
[0046] In another aspect, the application includes a method of suppressing a function of basophils and/or mast cells, thereby enabling treatment of diseases, conditions or disorders characterized by excessive or undesirable basophil and/or mast cell activity. According to the method, a present compound can be used to selectively inhibit the expression or activity of PI3K5 in the basophils and/or mast cells. Preferably, the method employs a PI3K5 inhibitor in an amount sufficient to inhibit stimulated histamine release by the basophils and/or mast cells. Accordingly, the use of a selective PI3K5 inhibitor can be of value in treating diseases characterized by histamine release (e.g. , allergic disorders). In certain embodiments, disorders that may be treated by the methods described herein include, for example, chronic obstructive pulmonary disease (COPD), asthma, ARDS, emphysema, and other related disorders. As shown in the examples provided herein, Compound A is selective for the δ isoform of PI3K over the α, β, and γ isoforms.
[0047] Another aspect includes methods of using Compound A or compositions thereof to treat a condition related to PI3K-mediated disorders such as inflammation or inflammatory disease.
Inflammation is a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute or wall off (i.e., sequester) both the injurious agent and the injured tissue. Inflammation or inflammatory disease can be acute or chronic, and often involves the immune response. Inflammation typically results from a cascade of events that includes vasodilation accompanied by increased vascular permeability and exudation of fluid and plasma proteins. The disruption of vascular integrity precedes or coincides with an infiltration of inflammatory cells. Inflammatory mediators generated at the site of the initial lesion serve to recruit inflammatory cells to the site of injury. These mediators (chemokines such as IL-8, MCP-1, MIP-1, and RANTES, complement fragments and lipid mediators) have chemotactic activity for leukocytes and attract the inflammatory cells to the inflamed lesion. These chemotactic mediators, which cause circulating leukocytes to localize at the site of inflammation, require the cells to cross the vascular
endothelium at a precise location. This leukocyte recruitment is accomplished by a process called cell adhesion. Inflammatory disease occurs when the normal discontinuation or attenuation of an inflammatory response does not occur or is incomplete. This includes but not limited to autoimmune disease, allergic disease, arthritic disease, asthma, acne, dermatitis, hypersensitive, transplant rejection, and inflammatory bowel disease.
[0048] In some embodiments, the term 'potency' or variant thereof refers to one compound has an increased levels of activity when compared to other compounds at a specific concentration. By way of example, the potency of the compound is determined by the IC50 value, which can be determined using commonly available methods; including in vitro enzyme assays or in vitro protein kinase assays. As understood by a person skilled in the art, a compound having a lower IC50 value is more potent than a compound having higher IC50 value. Also used herein, the term 'selectivity' or variant thereof refers to one compound has an increased level of activity on one isoform than other isoforms. In one preferred embodiment, the selectivity is the activity on some PI3K isoform and not other PI3K isoforms exerted by the compound disclosed herein. By way of example, the selectivity is determined using the EC50 value, which can be determined using commonly available methods for cellular assays. As understood by a person skilled in the art, a compound having a lower EC50 value is more selective than a compound having a higher EC50 value.
[0049] In some of the foregoing embodiments, the method further comprises reducing the level of PI3K5 activity in the patient. In some embodiments, the method further comprises the PI3K a- sparing activity in the patient in need thereof.
[0050] As used herein, the term 'the PDKa-sparing activity' or variant thereof refers to compounds having greater activity in one or more PI3K isoforms of β, δ, or γ than the PI3K a isoform in cellular assays. For example, compounds having PDKa-sparing activity inhibits PI3K5 more than PDKa, thereby reducing the activity of PI3K5 response more than that of PDKa. Dosing and Modes of Administration
[0051] In some embodiments, treatments of the methods described herein involve administration of Compound A to a subject in need of treatment on a daily basis for at least one week or more than one week. For example, Compound A is administered to a subject in need thereof on a daily basis for 2 to 4 weeks, for 3 to 4 weeks, for 1 month or more, or for 1 year or more. In some embodiments, Compound A may be administered in multiple doses each day, in order to maintain efficacious plasma levels over a prolonged period of time. Administration may be done in one dose per day, two doses per day, three doses per day, or four doses per day. Alternatively, Compound A can be administered intravenously at a rate that maintains an efficacious plasma level for a prolonged period of time.
[0052] The therapeutically effective amount can be determined by one of ordinary skill based on the subject's health, age, body weight, and condition. In some embodiments, the amount is normalized to the subject's body weight. For example, a dosage may be expressed as a number of milligrams of Compound A per kilogram of the subject's body weight (mg/kg). Dosages of between about 0.1 and 150 mg/kg are often appropriate, and in some embodiments, about 0.1 and 100 mg/kg are often appropriate, and in other embodiments a dosage of between 0.5 and 60 mg/kg is used. Normalizing according to the subject's body weight is particularly useful when adjusting dosages between subjects of widely disparate size, such as occurs when using the drug in both children and adult humans or when converting an effective dosage in a non-human subject such as dog to a dosage suitable for a human subject.
[0053] In other embodiments, the daily dosage may be described as a total amount of Compound A administered per dose or per day. Daily dosage of Compound A in some embodiments is between about 1 mg and 4,000 mg. In some embodiments, Compound A is administered at a dose of about 2,000 to 4,000 mg/day. In other embodiments, Compound A is administered at a dose of about 1 to 2,000 mg/day. In yet other embodiments, Compound A is administered at a dose of about 1 to 1,000 mg/day. In addition embodiments, Compound A is administered at a dose of about 10 to 500 mg/day. In other embodiment, Compound A is administered at a dose of about 20 to 500 mg/day. In other embodiments, Compound A is administered at a dose of about 50 to 300 mg/day. In yet another embodiments, Compound A is administered at a dose of about 75 to 200 mg/day. In other embodiment, Compound A is administered at a dose of about 15-150 mg/day.
[0054] When administered orally, the total daily dosage for a human subject in some
embodiments is between 1 mg and 1,000 mg. In a particular embodiment, Compound A is administered at a dose of about 10-500 mg/day. In a particular embodiment, Compound A is administered at a dose of about 50-300 mg/day. In a particular embodiment, Compound A is administered at a dose of about 75-200 mg/day. In a particular embodiment, Compound A is administered at a dose of about 100-150 mg/day.
[0055] In a particular embodiment, Compound A is administered at a dose of about 1 to 150 mg per dose, and one to four doses are administered per day (e.g. , QD dosing with about 1 to 150 mg, BID dosing with about 1 to 150 mg, or TID dosing with doses between about 1 to 150 mg, or QID dosing with doses between about 1 to 150 mg). In a preferred embodiment, a subject is treated with about 1 mg to 150 mg doses of Compound A once, twice, three, or four times per day.
Typically, the term QD refers to dosing once per day, BID refers to dosing twice per day, TID refers to dosing three times per day and QID refers to dosing four times per day.
[0056] In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of Compound A and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
[0057] In a particular embodiment, this method comprises continuing to treat the subject by administering Compound A at a dosage where clinical efficacy is achieved for a week or more, or reducing the dose by increments to a level at which safety and efficacy can be maintained. Safety can be monitored by conventional methods such as evaluating serum chemistry and complete blood count parameters. Efficacy can be monitored by conventional methods known in the art.
[0058] In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 1 to 500 mg of Compound A and increasing the dose to a total dosage of about 50 to 400 mg per day over at least 6 days. Optionally, the dosage can be further increased to about 750 mg/day.
[0059] In a particular embodiment, Compound A is administered once daily. In another embodiment, Compound A is administered at least twice daily. In some embodiments Compound A is administered three times per day. In some embodiments, Compound A is administered four times per day, or more than four times per day.
[0060] In a particular embodiment, Compound A is administered at a rate selected to produce a concentration of compound in the blood between about 40 to 4,000 ng/mL, and maintaining such concentration during a period of about 4 to 12 hours following administration. In another particular embodiment, the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 75 to 2,000 ng/mL and maintain that concentration during a period of about 4 to 12 hours from the time of administration. In some embodiments, the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 100 to 1,000 ng/mL following administration. In some embodiments, the dose size and frequency are selected to achieve a concentration of compound in the blood that is between about 100 to 500 ng/mL over a period of about 12 to 24 hours from the time of administration. In some embodiments, the dose size and frequency are selected to achieve a Cmax, plasma level of
Compound A that is at least about 500 ng/mL and does not exceed about 10,000 ng/mL.
[0061] In certain embodiments, Compound A is administered orally, intravenously,
transdermally, or by inhalation. In some embodiments, Compound A is administered orally in a dose of about 1 mg, 3 mg, 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, 150 mg, 200 mg, or 300 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day. In other embodiments, it is administered orally in a dose of about 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, or 150 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day.
[0062] In some embodiments, Compound A is administered by inhalation in a dose of about 0.1 mg, 0.3 mg, 0.5 mg, 1 mg, 3 mg, 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, 150 mg, 200 mg, or 300 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day. In other embodiments, it is administered by inhalation in a dose of about 15 mg, 20 mg, 25 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, or 100 mg, 125 mg, or 150 mg per dose, and the dose may be administered at a frequency of once per day, twice per day, three times per day, or four times per day. In a particular embodiment, Compound A may be administered prior to the onset of respiratory distress or an asthma attack. In another embodiment, Compound A may be
administered during respiratory distress or an asthma attack. In yet another embodiment,
Compound A may be administered following respiratory distress or an asthma attack.
[0063] In a particular embodiment, the method comprises administering to a patient, in addition to Compound A, a therapeutically effective amount of at least one therapeutic agent selected to treat the cancer in the patient. In certain embodiments, Compound A may be combined with one or more other active therapeutic agents in a unitary dosage form for simultaneous or sequential administration to a patient. The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. [0064] In one embodiment, co-administration of Compound A with one or more other active therapeutic agents generally refers to simultaneous or sequential administration of Compound A and one or more other active therapeutic agents, such that therapeutically effective amounts of Compound A and one or more other active therapeutic agents are both present in the body of the patient. In an alternative embodiment, Compound A and therapeutic agent(s) are not necessarily both present in the body of the patient but the particular dosing schedule Compound A and therapeutic agents results in synergistic effects.
[0065] Co-administration includes administration of unit dosages of Compound A before or after administration of unit dosages of one or more other active therapeutic agents; for example, administration of Compound A within seconds, minutes, hours or days of the administration of one or more other active therapeutic agents. For example, a unit dose of Compound A can be administered first, followed within seconds, minutes, hour or days by administration of a unit dose of one or more other active therapeutic agents. Alternatively, a unit dose of one or more other therapeutic agents can be administered first, followed by administration of a unit dose of
Compound A within seconds, minutes, hours or days. In some cases, it may be desirable to administer a unit dose of Compound A first, followed, after a period of hours (e.g. , 1 to 12 hours), by administration of a unit dose of one or more other active therapeutic agents. In other cases, it may be desirable to administer a unit dose of one or more other active therapeutic agents first, followed, after a period of hours (e.g. , 1 to 12 hours), by administration of a unit dose of Compound A. In some cases, it may be desirable to administer a unit dose of Compound A first, followed, after a period of days (e.g. , 1 to 14 days), by administration of a unit dose of one or more other active therapeutic agents. In other cases, it may be desirable to administer a unit dose of one or more other active therapeutic agents first, followed, after a period of days (e.g. , 1 to 14 days), by administration of a unit dose of Compound A. In some embodiments, the dosing regimen may involve alternating administration of Compound A and therapeutic agent over a period of several days, weeks, or months.
[0066] The combination therapy may provide "synergy" and "synergistic effect", i.e. , the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. , in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially.
[0067] In some embodiments, the therapeutic agent is selected from the following group consisting of theophylline, nedocromil sodium, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, a beta 2 agonist, an antihistamine, an anticholinergic, allergen immunotherapy, albuterol, levalbuterol, pirbutrol, ipratropium bromide, methylprednisolone, prednisolone, prednisone, beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone acetonide, salmeterol, formoterol, montelukast, zafirlukast, mometasone, ciclesonide, alfuzosin, ephedrine, metaproterenol, phenylpropanolamine, pseudoephedrine, zileuton, omalizumab, and a combination thereof.
[0068] In a particular embodiment, the method further comprises obtaining a biological sample from the subject; and analyzing the biological sample with an analytical procedure selected from the group consisting of blood chemistry analysis, chromosomal translocation analysis, needle biopsy, fluorescence in situ hybridization, laboratory biomarker analysis, immunohistochemistry staining method, flow cytometry, genetic analysis, or a combination thereof. Analysis may provide information about which patients may benefit from therapy, regression or progression of the tumor, an appropriate duration of the treatment, and is useful for determining dosages to administer, for adjusting dosages during a treatment cycle, and for deciding whether to continue or discontinue the treatments. The subject may be any mammal, including human and non-human such as dogs. In some embodiments, the subject is a healthy person. In other embodiment, the subject is a patient having a disease or a condition related to PI3K-mediated disorders. In one embodiment, the subject is a patient having asthma.
[0069] In one embodiment, the method described herein comprises administering to a subject Compound A described herein, in combination with a therapy used to treat a disease or a condition related to PI3K-mediated disorders. In one embodiment, the method described herein comprises administering to a subject Compound A described herein, in combination with a therpay used to treat asthma. In some embodiments, the "therapy" used to treat a disease {e.g., asthma) or a condition related to PI3K-mediated disorders is any well-known or experimental form of treatment used to treat asthma or a condition related to PI3K-mediated disorders that does not include the use of Compound A. In certain embodiments, the combination of Compound A with a conventional or experimental therapy used to treat a disease {e.g., asthma) or a condition related to PI3K-mediated disorders provides beneficial and/or desirable treatment results superior to results obtained by treatment without the combination. In certain embodiments, the therapies used to treat asthma or a condition related to PI3K-mediated disorders are well-known to a person having ordinary skill in the art and are described in the literature. In certain embodiments, the combination method provides for Compound A administered simultaneously or during the period of administration of the therapy. In certain embodiments, the combination method provides for Compound A administered prior to or after the administration of the therapy. The exact details regarding the administration of the combination may be determined experimentally. The refinement of sequence and timing of administering Compound A with a selected therapy will be tailored to the individual subject, the nature of the condition to be treated in the subject, and generally, the judgment of the attending practitioner.
[0070] The compounds described herein may be formulated for administration to animal subjects using commonly understood formulation techniques well known in the art. Formulations which are suitable for particular modes of administration and for Compound A may be found in
Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Company, Easton, PA.
[0071] The compounds described herein may be prepared in the form of prodrugs, i.e., protected forms which release the compounds described herein after administration to the subject. Typically, the protecting groups are hydrolyzed in body fluids such as in the bloodstream thus releasing the active compound or are oxidized or reduced in vivo to release the active compound. A discussion of prodrugs is found in Smith and Williams Introduction to the Principles of Drug Design, Smith, H.J.; Wright, 2nd ed., London (1988).
[0072] A compound described herein can be administered as the neat chemical, but it is typically preferable to administer the compound in the form of a pharmaceutical composition or formulation. Accordingly, also provided are pharmaceutical compositions that comprise Compound A and a biocompatible pharmaceutical carrier, adjuvant, or vehicle. The composition can include the agent as the only active moiety or in combination with other agents, such as oligo- or polynucleotides, oligo- or polypeptides, drugs, or hormones mixed with excipient(s) or other pharmaceutically acceptable carriers. Carriers and other ingredients can be deemed pharmaceutically acceptable insofar as they are compatible with other ingredients of the formulation and not deleterious to the recipient thereof.
[0073] The pharmaceutical compositions are formulated to contain suitable pharmaceutically acceptable carriers, and can optionally comprise excipients and auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically. The administration modality will generally determine the nature of the carrier. For example, formulations for parenteral administration can comprise aqueous solutions of the active compounds in water-soluble form. Carriers suitable for parenteral administration can be selected from among saline, buffered saline, dextrose, water, and other physiologically compatible solutions. Preferred carriers for parenteral administration are physiologically compatible buffers such as Hank's solution, Ringer' s solution, or physiologically buffered saline. For tissue or cellular administration, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. For preparations comprising proteins, the formulation can include stabilizing materials, such as polyols (e.g. , sucrose) and/or surfactants (e.g. , nonionic surfactants), and the like.
[0074] Alternatively, formulations for parenteral use can comprise dispersions or suspensions of the active compounds prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, and synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxy-methylcellulose, sorbitol, or dextran. Optionally, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Aqueous polymers that provide pH- sensitive solubilization and/or sustained release of the active agent also can be used as coatings or matrix structures, e.g. , methacrylic polymers, such as the EUDRAGIT™ series available from Rohm America Inc. (Piscataway, N.J.). Emulsions, e.g. , oil-in-water and water-in-oil dispersions, also can be used, optionally stabilized by an emulsifying agent or dispersant (surface active materials; surfactants). Suspensions can contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethlyene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, gum tragacanth, and mixtures thereof.
[0075] Liposomes containing the active agent also can be employed for parenteral
administration. Liposomes generally are derived from phospholipids or other lipid substances. The compositions in liposome form also can contain other ingredients, such as stabilizers,
preservatives, excipients, and the like. Preferred lipids include phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods of forming liposomes are known in the art. See, e.g. , Prescott (Ed.), METHODS IN CELL BIOLOGY, Vol. XIV, p. 33, Academic Press, New York (1976).
[0076] The pharmaceutical compositions comprising the agent in dosages suitable for nasal or instillable administration can be formulated using purified aqueous solutions of the active compound with preservative agents such as isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes. [0077] The pharmaceutical compositions disclosed herein may be administered into the respiratory system either by inhalation, respiration, nasal administration, or intrapulmonary instillation (into the lungs) of a subject by any suitable means, and they are preferably administered by generating an aerosol or spray comprised of powdered or liquid nasal, intrapulmonary, respirable, or inhalable particles. The respirable or inhalable particles comprising the active compound are inhaled by the subject, i.e. by inhalation or by nasal administration or by instillation into the respiratory tract or the lung itself. The formulation may comprise respirable or inhalable liquid or solid particles of the active compound that, in accordance with the present application, include respirable or inhalable particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and continue into the bronchi and alveoli of the lungs. In some
embodiments, particles may range from about 0.05, about 0.1, about 0.5, about 1, or about 2 to about 4, about 6, about 8, or about 10 μιη in diameter. In a particular embodiment, particles range from about 0.5 to less than about 5 μιη in diameter. Particles of non-respirable size that are included in an aerosol or spray tend to deposit in the throat and be swallowed. Thus, the quantity of non-respirable particles in the aerosol is preferably minimized. For nasal administration or intrapulmonary instillation, a particle size in the range of about 8, about 10, about 20, or about 25 to about 35, about 50, about 100, about 150, about 250, or about 500 μιη diameter is preferred to ensure retention in the nasal cavity or for instillation and direct deposition into the lung. Liquid formulations may be squirted into the respiratory tract (i.e. nose) and the lung, particularly when administered to newborns and infants.
[0078] Liquid pharmaceutical compositions of active compound for producing an aerosol may be prepared by combining the active ingredient with a stable vehicle, such as sterile pyrogen-free water. Solid particulate compositions containing respirable dry particles of micronized active compound maybe prepared by grinding dry active compound with a mortar and pestle, and then passing the micronized composition through a 400 mesh screen to break up or separate out large agglomerates. A solid particulate composition comprised of the active compound may optionally contain a dispersant that serves to facilitate the formulation of an aerosol. A suitable dispersant is lactose, which may be blended with the active compound in any suitable ratio, e.g., a 1 to 1 ratio by weight.
[0079] Aerosols of liquid particles comprising the active compound may be produced by any suitable means, such as with a nebulizer. Nebulizers are commercially available devices that transform solutions or suspensions of the active ingredient into a therapeutic aerosol mist either by means of acceleration of a compressed gas, typically air or oxygen, through a narrow venture orifice or by means of ultrasonic agitation. Suitable compositions for use in a nebulizer consist of the active ingredient in liquid carrier, the active ingredient comprising up to 40% w/w composition, but preferably less than 20% w/w, and the carrier being typically water or a dilute aqueous alcoholic solution, preferably made isotonic with body fluids by the addition of, for example, sodium chloride. Optional additives include preservatives if the composition is not prepared sterile, for example, methyl hydroxybenzoate, anti- oxidants, flavoring agents, volatile oils, buffering agents, and surfactants. Aerosols of solid particles comprising the active compound may likewise be produced with any solid particular medicament aerosol generator. Aerosol generators for administering solid particular medicaments to a subject produce particles that are respirable, as explained above, and generate a volume of aerosol containing a predetermined metered dose of a medicament at a rate suitable for human administration. Examples of such aerosol generators include metered dose inhalers and insufflators.
[0080] The composition may be delivered with any delivery device that generates liquid or solid particulate aerosols, such as aerosol or spray generators. These devices produce respirable particles, as explained above, and generate a volume of aerosol or spray containing a
predetermined metered dose or a medicament at a rate suitable for human or animal administration. One illustrative type of solid particulate aerosol or spray generator is an insufflator, which are suitable for administration of finely comminuted powders. In the insufflator, the powder, e.g., a metered dose of the composition effective to carry out the treatments described herein, is contained in a capsule or cartridge. These capsules or cartridges are typically made or gelatin, foil, or plastic, and may be pierced or opened in situ, and the powder delivered by air drawn through the device upon inhalation or by means or a manually operated pump. The composition employed in the insufflator may consist either solely of the active compound or of a powder blend comprising the active compound, typically comprising from 0.01 to 100% w/w of the composition. In some embodiments, the composition contains the active ingredient in an amount of about 0.01% w/w, about 1% w/w, or about 5% w/w to about 20% w/w, about 40% w/w, or about 99.99% w/w. Other ingredients, and other amounts of the agent, however, are also suitable within the confines of this application.
[0081] In one embodiment, the composition is delivered by a nebulizer. This means is especially useful for patients or subjects who are unable to inhale or respire the composition under their own efforts. The nebulizer can use any pharmaceutically acceptable carrier, such as a weak saline solution. The nebulizer is the means by which the powder pharmaceutical composition is delivered to the target of the patients or subjects in the airways. [0082] In one embodiment, the composition is delivered using suspension metered dose inhalation (MDI) formulation. Such a MDI formulation can be delivered using a delivery device using a propellant such as hydrofluoroalkane (HFA). Preferably, the HFA propellants contain 100 parts per million (ppm) or less of water.
[0083] In one embodiment, the delivery device comprises a dry powder inhalator (DPI) that delivers single or multiple doses of the composition. The single dose inhalator may be provided as a disposable kit that is sterilely preloaded with enough formulation for one application. The inhalator may be provided as a pressurized inhalator, and the formulation in a capsule or cartridge that may be pierced or opened. The kit may optionally also comprise in a separate container an agent such as other therapeutic compounds, excipients, surfactants (intended as therapeutic agents as well as formulation ingredients), antioxidants, flavoring and coloring agents, fillers, volatile oils, buffering agents, dispersants, surfactants, bulking agents, propellants, and preservatives, among other suitable additives for the different formulations.
[0084] The pharmaceutical compositions comprising the agent in dosages suitable for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art. The preparations formulated for oral administration can be in the form of tablets, pills, capsules, cachets, dragees, lozenges, liquids, gels, syrups, slurries, elixirs, suspensions, or powders. To illustrate, pharmaceutical preparations for oral use can be obtained by combining the active compounds with a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Oral formulations can employ liquid carriers similar in type to those described for parenteral use, e.g., buffered aqueous solutions, suspensions, and the like.
[0085] Preferred oral formulations include tablets, dragees, and gelatin capsules. These preparations can contain one or excipients, which include, without limitation:
a) diluents, such as sugars, including lactose, dextrose, sucrose, mannitol, or sorbitol; b) binders, such as magnesium aluminum silicate, starch from corn, wheat, rice,
potato, etc.;
c) cellulose materials, such as methylcellulose, hydroxypropylmethyl cellulose, and
sodium carboxymethylcellulose, polyvinylpyrrolidone, gums, such as gum arabic and gum tragacanth, and proteins, such as gelatin and collagen;
d) disintegrating or solubilizing agents such as cross-linked polyvinyl pyrrolidone,
starches, agar, alginic acid or a salt thereof, such as sodium alginate, or effervescent compositions; e) lubricants, such as silica, talc, stearic acid or its magnesium or calcium salt, and polyethylene glycol;
f) flavorants and sweeteners;
g) colorants or pigments, e.g. , to identify the product or to characterize the quantity
(dosage) of active compound; and
h) other ingredients, such as preservatives, stabilizers, swelling agents, emulsifying agents, solution promoters, salts for regulating osmotic pressure, and buffers.
[0086] In some preferred oral formulations, the pharmaceutical composition comprises at least one of the materials from group (a) above, or at least one material from group (b) above, or at least one material from group (c) above, or at least one material from group (d) above, or at least one material from group (e) above. Preferably, the composition comprises at least one material from each of two groups selected from groups (a)-(e) above.
[0087] Gelatin capsules include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating such as glycerol or sorbitol. Push-fit capsules can contain the active ingredient(s) mixed with fillers, binders, lubricants, and/or stabilizers, etc. In soft capsules, the active compounds can be dissolved or suspended in suitable fluids, such as fatty oils, liquid paraffin, or liquid polyethylene glycol with or without stabilizers.
[0088] Dragee cores can be provided with suitable coatings such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
[0089] The pharmaceutical composition can be provided as a salt of the active agent. Salts tend to be more soluble in aqueous or other protic solvents than the corresponding free acid or base forms. Pharmaceutically acceptable salts are well known in the art. Compounds that contain acidic moieties can form pharmaceutically acceptable salts with suitable cations. Suitable pharmaceutically acceptable cations include, for example, alkali metal (e.g. , sodium or potassium) and alkaline earth (e.g. , calcium or magnesium) cations.
[0090] Compound A may form pharmaceutically acceptable acid addition salts with suitable acids. For example, Berge, et ah, describe pharmaceutically acceptable salts in detail in J. Pharm. ScL, 66: 1 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting Compound A with a suitable acid.
[0091] Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorolsulfonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate (isothionate), lactate, maleate, methanesulfonate or sulfate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate or hydrogen phosphate, glutamate, bicarbonate, p-toluenesulfonate, and undecanoate. Examples of acids that can be employed to form pharmaceutically acceptable acid addition salts include, without limitation, such inorganic acids as hydrochloric acid, hydrobromic acid, sulfuric acid, and phosphoric acid, and such organic acids as oxalic acid, maleic acid, succinic acid, and citric acid.
[0092] Compositions comprising a compound described herein formulated in a pharmaceutical acceptable carrier can be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Accordingly, there also is contemplated an article of manufacture, such as a container comprising a dosage form of a compound described herein and a label containing instructions for use of the compound. Kits are also contemplated for the compounds and methods described herein. For example, the kit can comprise a dosage form of a pharmaceutical
composition and a package insert containing instructions for use of the composition in treatment of a medical condition. In either case, conditions indicated on the label can include treatment of asthma. In certain embodiments, the kit comprises Compound A, and at least one therapeutic agent disclosed herein. In certain embodiments, the kit may further comprise at least one
pharmaceutically acceptable excipient.
[0093] Pharmaceutical compositions comprising Compound A can be administered to the subject by any conventional method, including parenteral and enteral techniques. Parenteral
administration modalities include those in which the composition is administered by a route other than through the gastrointestinal tract, for example, intravenous, intraarterial, intraperitoneal, intramedullarly, intramuscular, intraarticular, intrathecal, and intraventricular injections. Enteral administration modalities include, for example, oral (including buccal and sublingual) and rectal administration. Transepithelial administration modalities include, for example, transmucosal administration and transdermal administration. Transmucosal administration includes, for example, enteral administration as well as nasal, inhalation, and deep lung administration; vaginal administration; and rectal administration. Transdermal administration includes passive or active transdermal or transcutaneous modalities, including, for example, patches and iontophoresis devices, as well as topical application of pastes, salves, or ointments. Parenteral administration also can be accomplished using a high-pressure technique, e.g., POWDERJECT™. [0094] Surgical techniques include implantation of depot (reservoir) compositions, osmotic pumps, and the like. A preferred route of administration for treatment of inflammation can be local or topical delivery for localized disorders such as arthritis, or systemic delivery for distributed disorders, e.g., intravenous delivery for reperfusion injury or for systemic conditions such as septicemia. For other diseases, including those involving the respiratory tract, e.g., chronic obstructive pulmonary disease, asthma, and emphysema, administration can be accomplished by inhalation or deep lung administration of sprays, aerosols, powders, and the like.
[0095] The characteristics of the agent itself and the formulation of the agent can influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agent. Such pharmacokinetic and pharmacodynamic information can be collected through preclinical in vitro and in vivo studies, later confirmed in humans during the course of clinical trials. Thus, for any compound used in the method described herein, a therapeutically effective dose can be estimated initially from biochemical and/or cell-based assays.
[0096] Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the "therapeutic index," which typically is expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices (i.e., the toxic dose is substantially higher than the effective dose) are preferred. The data obtained from such cell culture assays and additional animal studies can be used in formulating a range of dosage for human use. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
[0097] For the methods described herein, any effective administration regimen regulating the timing and sequence of doses can be used. Doses of the agent preferably include pharmaceutical dosage units comprising an effective amount of the agent. In some embodiments, "effective amount" refers to an amount sufficient to modulate the PDKa-sparing activity or any combination of ΡΒΚδ, ΡΒΚγ, and ΡΒΚβ expression or activity and/or derive a measurable change in a physiological parameter of the subject through administration of one or more of the pharmaceutical dosage units. "Effective amount" can also refer to the amount required to ameliorate a disease, condition or disorder in a subject.
[0098] Suitable dosage ranges for Compound A may vary according to these considerations, but in general, Compound A may be administered in the range of about 10.0 μg/kg to 15 mg/kg of body weight; about 1.0 μg/kg to 10 mg/kg of body weight, or about 0.5 mg/kg to 5 mg/kg of body weight. For a typical about 70-kg human subject, thus, the dosage range is from about 700 μg to 1050 mg; about 70 μg to 700 mg; or about 35mg to 350 mg per dose, and two or more doses may be administered per day. Dosages may be higher when Compound A is administered orally or transdermally as compared to, for example, IV administration. In certain embodiments, the treatment of asthma comprises oral administration of up to about 750 mg/day of Compound A. The reduced toxicity of this compound permits the therapeutic administration of relatively high doses. The reduced toxicity of Compound A, permits the therapeutic administration of relatively high doses. In some embodiments, Compound A is administered orally, in three to five doses per day, using about 20 to 150 mg per dose for a total daily dose between about 60 to 750 mg. In some embodiments, the total daily dose is between about 100 to 500 mg, and in some embodiments the normalized daily dosage (adjusted for subject' s body weight) is up to about 60 mg per kg of the treated subject's body weight.
[0099] Compound A may be administered as a single bolus dose, a dose over time, as in IV or transdermal administration, or in multiple dosages. For IV or transdermal delivery, a dosage may be delivered over a prolonged period of time, and may be selected or adjusted to produce a desired plasma level of the active compound. In some embodiments, the desired plasma level is at least about 1 micromolar, or at least about 10 micromolar.
[0100] When Compound A is administered orally, it is preferably administered one time per day or in two or more doses per day. In some embodiments, three doses per day are administered. In some embodiments four doses per day are administered.
[0101] Dosing may be continued for one day or for multiple days, such as about 7 days. In some embodiments, daily dosing is continued for about 14 days or about 28 days. In some
embodiments, dosing is continued for about 28 days and is then discontinued for about 7 days; the efficacy of the treatment can be assessed during the break, when treatment with Compound A has been stopped, and if the assessment shows that the treatment is achieving a desired effect, another cycle of about 7 to 28 days of treatment with Compound A can be initiated.
[0102] Depending on the route of administration, a suitable dose can be calculated according to body weight, body surface area, or organ size. The final dosage regimen will be determined by the attending physician in view of good medical practice, considering various factors that modify the action of drugs, e.g. , the agent' s specific activity, the identity and severity of the disease state, the responsiveness of the patient, the age, condition, body weight, sex, and diet of the patient, and the severity of the disease related to PI3K-mediated disorders (e.g. asthma), a condition related to PI3K-mediated disorders, or any infection. Additional factors that can be taken into account include comorbidities, prior therapies, the time and frequency of administration, drug combinations, reaction sensitivities, and tolerance/response to therapy. Further refinement of the dosage appropriate for treatment involving any of the formulations mentioned herein is done routinely by the skilled practitioner without undue experimentation, especially in light of the dosage information and assays disclosed, as well as the pharmacokinetic data observed in human clinical trials. Appropriate dosages can be ascertained through use of established assays for determining concentration of the agent in a body fluid or other sample together with dose -response data.
[0103] The frequency of dosing will depend on the pharmacokinetic parameters of the agent and the route of administration. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Accordingly, the pharmaceutical compositions can be administered in a single dose, multiple discrete doses, continuous infusion, sustained release depots, or combinations thereof, as required to maintain desired minimum level of the agent. Short- acting pharmaceutical compositions (i.e. , short half-life) can be administered once a day or more than once a day (e.g. , two, three, or four times a day). Long acting pharmaceutical compositions might be administered every 3 to 4 days, every week, or once every two weeks to 12 weeks.
Pumps, such as subcutaneous, intraperitoneal, or subdural pumps, can be preferred for continuous infusion.
[0104] Subjects that will respond favorably to the methods described herein include medical and veterinary subjects generally, including human patients. Among other subjects for whom the methods described herein are useful are cats, dogs, large animals, avians such as chickens, and the like. In general, any subject who would benefit from Compound A is appropriate for
administration of the method described herein.
[0105] The present disclosure will be understood more readily by reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present disclosure.
EXAMPLES
Example 1
Preparation of Compound A
[0106] The (S)-enantiomer of Compound A was prepared as shown in Scheme 1. Scheme
Figure imgf000029_0001
(A(S))
Preparation of 5-chloro-2H-3,l-benzoxazine-2,4(lH)-dione (2)
[0107] 2-amino-6-chlorobenzoic acid (1) (10 g, 1 equiv) was dissolved in acetonitrile (58.1 mL, 19.1 equiv) at 50°C, and added with pyridine (9.4 mL, 2 equiv). Then, triphosgene (5.7 g, 0.33 equiv) in methylene chloride (30 mL, 9 equiv) was added dropwise with stirring. The reaction mixture was stirred for 2 hours at 50°C. The solvent was removed by rotary evaporation. The residue was then dispersed in 50 mL water and filtered. The tan solid was washed with a minimal amount of acetonitrile to remove color, and dried to produce an off-white solid powder. HPLC RT was 4.41 minutes. All compounds were characterized using high performance liquid
chromatograph (HPLC), with elution from the Zorbax Cg column using a gradient of 0-100% acetonitrile in water containing 0.07% trifluoroacetic acid (TFA) and detection using absorbance at 210 nm and 254 nm. Preparation of 2-amino-6-chloro-N-phenylbenzamide (3)
[0108] 5-chloro-2H-3,l-benzoxazine-2,4(lH)-dione (2) (2.00 g, 1 equiv) was dissolved in dioxane (15 mL, 19 equiv) at 40°C. The aniline (1.38 mL, 1 equiv) was added dropwise over 30 minutes, gradually warming to 100°C. The reaction mixture was stirred for 4 hours then cooled to ambient temperature (25°C + 5°C). The solvent was removed by evaporation. Chromatography was performed using 90 g silica gel with 1: 1 (v/v) ethyl acetate: hexane to yield a white solid. HPLC RT was 5.46 minutes.
Preparation of (S)-tert-butyl l-(3-chloro-2-(phenylcarbamoyl)phenylamino)-l-oxopropan-2- ylcarbamate (4)
[0109] (S)-2-(tert-butoxycarbonylamino)propanoic acid (0.4 g, 2 equiv) was dissolved in dry tetrahydrofuran (THF)(3 mL, 40 equiv), and 4-methylmorpholine (0.256 mL, 2.2 equiv) was added. The reaction mixture was then cooled to -15°C in an ethylene glycol/C02 bath. A solution of isobutyl chloroformate (0.274 mL, 2 equiv) in dry THF (1 mL) was added dropwise to the reaction mixture, and stirred for 30 minutes. The reaction was stirred at -15°C for 30 minutes, then added with 2-amino-6-chloro-N-phenylbenzamide (3) in THF (1.0 mL). The reaction mixture was slowly warmed to 21°C. When about 10% conversion was observed, the reaction mixture was warmed to 60°C for 30 minutes. The reaction mixture was poured into ethyl acetate (150 mL), and washed with water (50 mL) twice and brine (30 mL). The organic layer was dried over sodium sulfate, filtered, and rotary evaporated to remove the solvent. Chromatography was performed using 90 g silica gel with 1:4 (v/v) ethyl acetate: hexane to yield white crystals.
Preparation of (S)-2-(l-aminoethyl)-5-chloro-3-phenylquinazolin-4(3H)-one (5)
[0110] (S)-tert-butyl-l-(3-chloro-2-(phenylcarbamoyl)phenylamino)-l-oxopropan-2-ylcarbamate (4) (5 g, 1 equiv) was dissolved in acetonitrile (300 mL, 500 equiv) under a nitrogen atmosphere. Triethylamine (79.21 mL, 47.5 equiv) was added with stirring, followed by the dropwise addition of chlorotrimethylsilane (22.78 mL, 15 equiv). The flask was sealed, and placed in an oil bath and heated to 90°C for 48 hours. HPLC RT was 6.66 minutes. The solvents were evaporated, and the dark residue was dissolved in ethyl acetate (350 mL) and washed with sodium bicarbonate (100 mL), water (100 mL) and brine (100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated by rotary evaporation to yield a brown solid (tert-butyl [(lS)-l-(5-chloro- 4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl]carbamate). [0111] The brown solid was then dissolved in methylene chloride (15 mL). TFA (6 mL) was added at 21°C and stirred for 2 hours. The reaction mixture was diluted with toluene (100 mL), and solvents were removed by rotary evaporation. The remaining solid was dissolved in ethyl acetate (300 mL), and washed with sodium bicarbonate (100 mL), water (100 mL), and brine (100 mL). The organic layer was dried over magnesium sulfate, filtered, and rotary evaporated to remove solvent. Chromatography was performed using 40 g silica gel with chloroform and a slow gradient to 10% methanol (containing 10% ammonium hydroxide)-chloroform to yield (S)-2-(l- aminoethyl)-5-chloro-3-phenylquinazolin-4(3H)-one (5).
Preparation of (S)-4-amino-6-((l-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2- yl)ethyl)amino)pyrimidine-5-carbonitrile (A(S))
[0112] To 200 mg (0.66 mmol) of (5) and 103 mg (0.66 mmol) of (6) in a lOmL microwave vial was added 0.23 mL iPr2NEt (1.2 mmol, 2 equiv.) and 4 mL of 2-propanol. The reaction mixture was heated under microwave irradiation for 20 min and 130° C. The contents of the vial were concentrated to dryness by rotary evaporation and dissolved in DMSO and filtered. Compound (A(S)) was purified by reverse phase preparative chromatography. The fractions containing the product were poured into 50 mL 2.5M K2C03 and extracted with chloroform. The chloroform layer was separated and dried with MgS04 and concentrated to yield Compound A(S).
[0113] In the examples below, unless stated otherwise, Compound A is the optically active form that predominantly includes the (S)-enantiomer.
Example 2
In Vivo Pharmacokinetics of Compound A in Rats
[0114] Compound A(S) was formulated for IV infusion in a sterile solution of 5% ethanol, 75% PEG 400, and 20% citrate buffer (pH 3.0) at 0.09 mg/mL. Compound A(S) was formulated for oral dosing (PO) in 5% ethanol, 87% PEG 400, 4% solutol, and 4% water at 0.23 mg/mL.
[0115] Each dosing group consisted of three Sprague Dawley rats. At dosing, the animals weighed between 0.26 and 0.27 kg.
[0116] For the IV infusion group, the test article was administered to the rats via infusion over 30-minutes at 5.0 mL/kg for a dose of 0.44 mg/kg. For the PO dosing group, the test article was administered to the rats via oral gavage at 10.0 mL/kg for a dose of 1.52 mg/kg.
[0117] Blood samples (approximately 0.35 mL each) were taken at specified time points after dosing from each animal, as shown in Tables 1 and 2. The blood samples were collected into VACUTAINER tubes containing EDTA-K2 as the anti-coagulant and were immediately placed on wet ice pending centrifugation for plasma.
[0118] Table 1. Plasma concentration-time data of Compound A(S) in plasma after a 30-minute
IV infusion of Compound A(S) at 0.44 mg/kg in rats.
Time (hr) Rat 1 (nM) Rat 2 (nM) Rat 3 (nM) Mean (nM) SD (nM)
0.0 BLQ BLQ BLQ NC NC
0.25 343 340 280 321 35.8
0.48 652 556 538 582 61.5
0.58 509 383 386 426 72.0
0.75 353 303 236 297 58.4
1.5 224 205 158 196 34.0
3.0 158 98.8 93.6 117 35.6
6.0 40.9 13.0 37.1 30.3 15.1
8.0 21.0 10.8 26.7 19.5 8.1
12.0 2.3 1.6 3.5 2.5 1.0
24.0 BLQ BLQ 1.5 NC NC
BLQ: Below the limit of quantification
NC: Not calculated due to insufficient data
[0119] Table 2. , Plasma concentration-time data of Compound A(S) in plasma after an oral of Compound A(S) at 1.52 mg/kg in rats.
Time (hr) Rat 4 (nM) Rat 5 (nM) Rat 6 (nM) Mean (nM) SD (nM)
0.0 BLQ BLQ BLQ NC NC
0.25 261 561 134 318 219
0.50 312 794 211 439 312
1.0 364 711 247 441 242
2.0 326 703 499 509 189
4.0 209 318 385 304 88.5
6.0 104 163 225 164 60.4
8.0 98.1 101 93.5 97.6 3.9
12.0 5.1 24.2 26.9 18.7 11.8
24.0 1.2 BLQ BLQ NC NC
BLQ: Below the limit of quantification
NC: Not calculated due to insufficient data
[0120] Rats 4 and 5 were observed with diarrhea at 6, 8, and 12 hours postdose. Rat 6 was observed with very soft stool at 4 and 6 hours postdose, and with diarrhea at 8 and 12 hours postdose.
[0121] An LC/MS/MS method was used to measure the concentrations of Compound A(S) in plasma. An aliquot of 50 μΐ^ of each plasma sample was treated with 100 μΐ^ of acetonitrile (ACN) containing internal standard. The above solution was centrifuged at 3000 RPM for 20 minutes and 75 μΐ^ of supernatant was transferred to a clean 96-well plate, followed by the addition of 75 μΐ^ of water. An aliquot of 20 μΐ^ was injected to the TSQ Quantum LC/MS/MS system (Thermo Finnigan, San Jose, CA). The column used was a HyPurity C18 HLPC column (30 X 2.1 mm, 5 μ) (Thermo-Hypersil, Part #22105-032130). Mobile phase A contained 1% acetonitrile in 10 mM ammonium formate aqueous solution, and mobile phase B contained 80% acetonitrile in 10 mM ammonium formate aqueous solution. A Cohesive LX-2 multiplex with two identical Agilent 1200 series binary pumps (P/N G1312A Bin Pump) was used for elution. An HTS PAL autosampler was used (LEAP Technologies, Carrboro, NC).
[0122] Un-used portions of the dosing solutions were saved. The concentration of Compound A(S) in the dosing solution was measured with an HPLC-UV method. The target and measured concentrations of Compound A(S) are listed in Table 3.
[0123] Table 3. Concentrations of Compound A(S) in dosing solutions
Animal Target Measured Actual
Animal Dose Vol. Actual Dose
Route Weight Cone. Cone. Dose
ID (mL) (nmoles/kg)
(kg) (mg/mL) (mg/mL) (mg/kg)
Rat 1 IV 0.27 0.10 0.09 1.40 0.46 1107
Rat 2 IV 0.27 0.10 0.09 1.30 0.44 1045
Rat 3 IV 0.27 0.10 0.09 1.30 0.43 1038
Rat 4 PO 0.27 0.30 0.23 1.80 1.54 3694
Rat 5 PO 0.27 0.30 0.23 1.80 1.53 3666
Rat 6 PO 0.26 0.30 0.23 1.70 1.48 3533
[0124] A concentration-time profile of Compound A(S) in plasma following the 30-minute IV infusion at 0.44 mg/kg and the oral dose at 1.52 mg/kg in rats is shown in Figure 1. Figures 2 and 3 show the individual rat concentration-time profile for IV and oral administration, respectively.
[0125] The apparent systemic clearance for Compound A(S) was low/intermediate relative to live blood flow in rats (CL = 1.12 + 0.57 L/hr/kg). The volume of distribution was much larger than the volume of total body water (Vss = 2.64 + 0.97 L/kg). The half-life of Compound A(S) was 2.6 + 1.7 hr, while the mean residence time (MRT) was 2.3 + 0.7 hr. The bioavailability of Compound A(S) was approximately 75% + 21%. The Cmax was 552 + 220 nM at 1.2 + 0.8 hr. The pharmacokinetic parameters are summarized in Tables 4 and 5. [0126] Table 4. Pharmacokinetic parameters of Compound A(S) in plasma following a 30-min
IV infusion of Compound A(S) at 0.44 mg/kg in rats.
IV Compound A(S)
Plasma Rat i Rat 2 Rat 3 Mean SD
AUCiast (nM-hr) 1198 886 916 1000 112
AUCinf (nM-hr) 1203 890 926 1006 111
Cmax (nM) 652 556 538 582 61.5
max (hr) 0.48 0.48 0.48 0.48 0.00
MRT (hr) 2.2 1.7 3.1 2.3 0.7 ti/2 (hr) 1.4 1.9 4.5 2.6 1.7
Vss (L/kg) 2.00 2.16 3.75 2.64 0.97
CL (L/hr/kg) 0.92 1.24 1.20 1.12 0.17
[0127] Table 5. Pharmacokinetic parameters of Compound A(S) in plasma following an oi dose of Compound A(S) at 1.52 mg/kg in rats.
PO Compound A(S)
Plasma Rat 4 Rat 5 Rat 6 Mean SD
AUCiast (nM-hr) 1913 3340 2600 2618 713
AUCinf (nM-hr) 1918 3415 2677 2670 748
Cmax (nM) 364 794 499 552 220
max (hr) 1.0 0.5 2.0 1.2 0.8 ti/2 (hr) 2.9 2.1 2.0 2.3 0.5
F% 55% 96% 75% 75% 21%
Example 3
In vitro Potency and Specificity of Compound A
[0128] The potency and selectivity of Compound A was compared to other inhibitors having PI3K inhibitory activity, such as Compounds B-G. The structure of Compounds B-G are each shown below.
Figure imgf000035_0001
[0129] IC50. IC50 values for inhibiting PI3K isoforms were determined using in vitro
SelectScreen kinase inhibitor assay service (Invitrogen Ltd.). Compounds were diluted in 10 mM of DMSO, and measured for 10-point kinase inhibitory activities over a range of concentration from 5 to 104 nM with ATP concentration consistent with each enzyme' s Km.
[0130] IC50 values for Compounds A-G for various isoforms of PI3K are shown in Table 6.
[0131] Table 6. Comparison of IC50 values of Compounds A-G for different isoforms of PI3K
(nM)
Figure imgf000035_0002
[0132] EC '50. To analyze PI3K pi 105 and pi 10γ signaling, basophil activation was measured in isolated PBMC or whole blood using the Flow2 CAST kit according to the manufacture' s standardized methods (Buhlman Laboratories AG, Switzerland). Briefly, ρ ΐ ΐθδ was activated with anti-FCeRI and pi 10γ was activated with fMLP (2 μΜ) in the absence or presence of compounds. To monitor the basophil cell population and cellular activation, anti-CD63-FITC and anti-CCR3- PE antibodies were added to each sample. Cells were fixed and analyzed on a FC500MPL flow cytometer (Beckman Coulter, Brea, CA).
[0133] The EC50 value for Compound A(S) in PI3K p 110δ was 1.0 nM. The EC50 value for Compound A(S) in PI3K pi 10γ was 50 nM. Thus, the potency of Compound A(S) is greater for the δ-isoform than for the γ-isoform.
Example 4
Metabolic Stability of Compound A
[0134] The in vitro metabolic stability of Compounds A(S) and G was measured in human, rat, and dog microsomes using procedures known in the art. The half-lives of these compounds in the microsomes are provided in Table 7.
[0135] Table 7. Half-lives of Compounds A(S) and G in human, rat, and dog microsomes (min).
Figure imgf000036_0001
[0136] The half-life of Compound A(S) in humans and rats is two to four times longer than the half-life of Compound G in humans and rats.

Claims

1. A compound having the structure of Compound A
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound or a pharmaceutically acceptable salt thereof is the (S)-enantiomer.
3. A composition comprising the compound according to claim 1 or a
pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
4. The composition according to claim 3, wherein the composition comprises the (S)- enantiomer of the compound or a pharmaceutically acceptable salt thereof, wherein the (S)- enantiomer of the compound is present in excess of the (R)-enantiomer of the compound.
5. The composition according to claim 4, wherein the composition is substantially free of the (R)-enantiomer of the compound or a pharmaceutically acceptable salt thereof.
6. A method of treating a PI3K-mediated condition in a patient, comprising administering to the patient a composition comprising the compound according to claim 1 and at least one pharmaceutically acceptable excipient.
7. The method according to claim 6, wherein the composition comprises the (S)- enantiomer of the compound or a pharmaceutically acceptable salt thereof, wherein the (S)- enantiomer of the compound or a pharmaceutically acceptable salt thereof is present in excess of the (R)-enantiomer of the compound or a pharmaceutically acceptable salt thereof.
8. The method according to claim 7, wherein the composition is substantially free of the (R)-enantiomer of the compound or a pharmaceutically acceptable salt thereof.
9. The method according to claim 7, wherein the (S)-enantiomer of the compound or a pharmaceutically acceptable salt thereof predominates over the (R)-enantiomer of the compound or a pharmaceutically acceptable salt thereof by a ratio of at least 9: 1.
10. The method according to claim 6, wherein the condition is characterized by histamine release.
11. The method according to claim 6, wherein the condition is an allergic disorder.
12. The method according to claim 6, wherein the condition is selected from the group consisting of asthma, chronic obstructive pulmonary disease, ARDS, and emphysema.
13. The method according to claim 12, wherein the condition is asthma.
14. The method according to claim 6, wherein the compound is administered at a dose of about 20-500 mg/day.
15. The method according to claim 6, wherein the compound is administered at least twice daily.
16. The method according to claim 6, further comprising reducing the level of PI3K5, ΡΒΚγ, or ΡΙ3Κβ activity in the patient.
17. The method according to claim 6, further comprising administering to the patient, in addition to the compound or a pharmaceutically acceptable salt thereof, a therapeutically effective amount of at least one therapeutic agent selected to treat asthma in the patient.
18. The method according to claim 17, wherein the therapeutic agent is selected from the following group consisting of theophylline, nedocromil sodium, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, a beta 2 agonist, an antihistamine, an anticholinergic, allergen immunotherapy, albuterol, levalbuterol, pirbutrol, ipratropium bromide, methylprednisolone, prednisolone, prednisone, beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone acetonide, salmeterol, formoterol, montelukast, zafirlukast, mometasone, ciclesonide, alfuzosin, ephedrine, metaproterenol, phenylpropanolamine, pseudoephedrine, zileuton, omalizumab, and a combination thereof.
19. The method of claim 6, wherein the compound selectively inhibits PI3K5.
20. A kit comprising the compound according to claim 1 or a pharmaceutically acceptable salt thereof, and instructions for use of the composition in treatment of asthma.
PCT/US2013/024216 2012-02-03 2013-01-31 Compositions and methods of treating a disease with (s)-4 amino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile WO2013116562A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261594868P 2012-02-03 2012-02-03
US61/594,868 2012-02-03

Publications (1)

Publication Number Publication Date
WO2013116562A1 true WO2013116562A1 (en) 2013-08-08

Family

ID=48905846

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/024216 WO2013116562A1 (en) 2012-02-03 2013-01-31 Compositions and methods of treating a disease with (s)-4 amino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile

Country Status (1)

Country Link
WO (1) WO2013116562A1 (en)

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014072937A1 (en) 2012-11-08 2014-05-15 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
CN104262344A (en) * 2014-08-22 2015-01-07 苏州明锐医药科技有限公司 A preparing method of Idelalisib
WO2015001491A1 (en) 2013-07-02 2015-01-08 Rhizen Pharmaceuticals Sa Pi3k protein kinase inhibitors, particularly delta and/or gamma inhibitors
CN104418849A (en) * 2013-09-02 2015-03-18 广东东阳光药业有限公司 Substituted aminopyrimidine compound as well as using method and application thereof
WO2015042077A1 (en) 2013-09-22 2015-03-26 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
WO2015168079A1 (en) * 2014-04-29 2015-11-05 Infinity Pharmaceuticals, Inc. Pyrimidine or pyridine derivatives useful as pi3k inhibitors
WO2016126552A1 (en) 2015-02-03 2016-08-11 Gilead Sciences, Inc. Combination therapies for treating cancers
WO2016134668A1 (en) * 2015-02-28 2016-09-01 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine compounds and methods of use
CN105924434A (en) * 2015-02-28 2016-09-07 广东东阳光药业有限公司 Substituted aminopyrimidine compound and usage method and application thereof
WO2016149160A1 (en) * 2015-03-15 2016-09-22 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine compounds and methods of use
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
US9546180B2 (en) 2011-08-29 2017-01-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2017059224A2 (en) 2015-10-01 2017-04-06 Gilead Sciences, Inc. Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
WO2017106556A1 (en) 2015-12-17 2017-06-22 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US9738644B2 (en) 2010-05-21 2017-08-22 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
WO2017152062A1 (en) 2016-03-04 2017-09-08 Gilead Sciences, Inc. Compositions and combinations of autotaxin inhibitors
WO2017177179A1 (en) 2016-04-08 2017-10-12 Gilead Sciences, Inc. Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
WO2017215590A1 (en) 2016-06-13 2017-12-21 I-Mab Anti-pd-l1 antibodies and uses thereof
WO2018026835A1 (en) 2016-08-04 2018-02-08 Gilead Sciences, Inc. Cobicistat for use in cancer treatments
US9944639B2 (en) 2014-07-04 2018-04-17 Lupin Limited Quinolizinone derivatives as PI3K inhibitors
WO2018085069A1 (en) 2016-11-03 2018-05-11 Gilead Sciences, Inc. Combination of a bcl-2 inhibitor and a bromodomain inhibitor for treating cancer
WO2018097977A1 (en) 2016-11-22 2018-05-31 Gilead Sciences, Inc. Crystalline forms of a phosphate complex of a bet inhibitor
WO2018137598A1 (en) 2017-01-24 2018-08-02 I-Mab Anti-cd73 antibodies and uses thereof
WO2018156895A1 (en) 2017-02-24 2018-08-30 Gilead Sciences, Inc. Inhibitors of bruton's tyrosine kinase
WO2018156901A1 (en) 2017-02-24 2018-08-30 Gilead Sciences, Inc. Inhibitors of bruton's tyrosine kinase
WO2018195321A1 (en) 2017-04-20 2018-10-25 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2019160882A1 (en) 2018-02-13 2019-08-22 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
WO2019204609A1 (en) 2018-04-19 2019-10-24 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
WO2019217780A1 (en) 2018-05-11 2019-11-14 Phosphorex, Inc. Microparticles and nanoparticles having negative surface charges
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2019222112A1 (en) 2018-05-14 2019-11-21 Gilead Sciences, Inc. Mcl-1 inhibitors
WO2020014643A1 (en) 2018-07-13 2020-01-16 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2020072656A1 (en) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
WO2020086556A1 (en) 2018-10-24 2020-04-30 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
WO2021076908A1 (en) 2019-10-18 2021-04-22 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
WO2021087064A1 (en) 2019-10-31 2021-05-06 Forty Seven, Inc. Anti-cd47 and anti-cd20 based treatment of blood cancer
WO2021096860A1 (en) 2019-11-12 2021-05-20 Gilead Sciences, Inc. Mcl1 inhibitors
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
WO2021163064A2 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
WO2021222522A1 (en) 2020-05-01 2021-11-04 Gilead Sciences, Inc. Cd73 inhibiting 2,4-dioxopyrimidine compounds
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2022221304A1 (en) 2021-04-14 2022-10-20 Gilead Sciences, Inc. CO-INHIBITION OF CD47/SIRPα BINDING AND NEDD8-ACTIVATING ENZYME E1 REGULATORY SUBUNIT FOR THE TREATMENT OF CANCER
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023107956A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16 and 5t4
WO2023107954A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Antibodies targeting 5t4 and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023183817A1 (en) 2022-03-24 2023-09-28 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023196784A1 (en) 2022-04-05 2023-10-12 Gilead Sciences, Inc. Combinations of antibody therapies for treating colorectal cancer
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
WO2024064668A1 (en) 2022-09-21 2024-03-28 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPα DISRUPTION ANTICANCER COMBINATION THERAPY

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US20050054614A1 (en) * 2003-08-14 2005-03-10 Diacovo Thomas G. Methods of inhibiting leukocyte accumulation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
US20050054614A1 (en) * 2003-08-14 2005-03-10 Diacovo Thomas G. Methods of inhibiting leukocyte accumulation

Cited By (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9738644B2 (en) 2010-05-21 2017-08-22 Infinity Pharmaceuticals, Inc. Chemical compounds, compositions and methods for kinase modulation
US9546180B2 (en) 2011-08-29 2017-01-17 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
EA035391B1 (en) * 2012-11-08 2020-06-05 Ризен Фармасьютикалз Са Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US10058539B2 (en) 2012-11-08 2018-08-28 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a PDE4 inhibitor and a PI3 delta or dual PI3 delta-gamma kinase inhibitor
US10413532B2 (en) 2012-11-08 2019-09-17 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a PDE4 inhibitor and a PI3 delta or dual PI3 delta-gamma kinase inhibitor
US9737521B2 (en) 2012-11-08 2017-08-22 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a PDE4 inhibitor and a PI3 delta or dual PI3 delta-gamma kinase inhibitor
WO2014072937A1 (en) 2012-11-08 2014-05-15 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US11065236B2 (en) 2012-11-08 2021-07-20 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a PDE4 inhibitor and a PI3 delta or dual PI3 delta-gamma kinase inhibitor
US9481667B2 (en) 2013-03-15 2016-11-01 Infinity Pharmaceuticals, Inc. Salts and solid forms of isoquinolinones and composition comprising and methods of using the same
WO2015001491A1 (en) 2013-07-02 2015-01-08 Rhizen Pharmaceuticals Sa Pi3k protein kinase inhibitors, particularly delta and/or gamma inhibitors
CN104418849A (en) * 2013-09-02 2015-03-18 广东东阳光药业有限公司 Substituted aminopyrimidine compound as well as using method and application thereof
CN104513235B (en) * 2013-09-02 2017-12-05 广东东阳光药业有限公司 Substituted amino-metadiazine compound and its application method and purposes
CN104513235A (en) * 2013-09-02 2015-04-15 广东东阳光药业有限公司 Substituted aminopyrimidine compound and use thereof
CN104447727A (en) * 2013-09-02 2015-03-25 广东东阳光药业有限公司 Substituted aminopyrimidine compound as well as use method and application thereof
RU2675105C9 (en) * 2013-09-22 2019-01-09 Саншайн Лейк Фарма Ко., Лтд. Substituted aminopyrimidine compounds and methods of use
RU2675105C2 (en) * 2013-09-22 2018-12-17 Саншайн Лейк Фарма Ко., Лтд. Substituted aminopyrimidine compounds and methods of use
JP2016534146A (en) * 2013-09-22 2016-11-04 キャリター・サイエンシーズ・リミテッド・ライアビリティ・カンパニーCalitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
US9512114B2 (en) 2013-09-22 2016-12-06 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
US9518046B2 (en) 2013-09-22 2016-12-13 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
WO2015042077A1 (en) 2013-09-22 2015-03-26 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
EP3046563A4 (en) * 2013-09-22 2017-02-15 Calitor Sciences, LLC Substituted aminopyrimidine compounds and methods of use
JP2016530338A (en) * 2013-09-22 2016-09-29 キャリター・サイエンシーズ・リミテッド・ライアビリティ・カンパニーCalitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
US9657007B2 (en) 2013-09-22 2017-05-23 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
US9670194B2 (en) 2013-09-22 2017-06-06 Calitor Sciences, Llc Substituted aminopyrimidine compounds and methods of use
WO2015168079A1 (en) * 2014-04-29 2015-11-05 Infinity Pharmaceuticals, Inc. Pyrimidine or pyridine derivatives useful as pi3k inhibitors
US9944639B2 (en) 2014-07-04 2018-04-17 Lupin Limited Quinolizinone derivatives as PI3K inhibitors
CN104262344B (en) * 2014-08-22 2015-11-04 苏州明锐医药科技有限公司 The preparation method of Chinese mugwort Dana Delany
CN104262344A (en) * 2014-08-22 2015-01-07 苏州明锐医药科技有限公司 A preparing method of Idelalisib
WO2016126552A1 (en) 2015-02-03 2016-08-11 Gilead Sciences, Inc. Combination therapies for treating cancers
CN105924434A (en) * 2015-02-28 2016-09-07 广东东阳光药业有限公司 Substituted aminopyrimidine compound and usage method and application thereof
WO2016134668A1 (en) * 2015-02-28 2016-09-01 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine compounds and methods of use
CN105924433A (en) * 2015-02-28 2016-09-07 广东东阳光药业有限公司 Substituted aminopyrimidine compound and usage method and application thereof
WO2016149160A1 (en) * 2015-03-15 2016-09-22 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine compounds and methods of use
WO2017059224A2 (en) 2015-10-01 2017-04-06 Gilead Sciences, Inc. Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
WO2017106556A1 (en) 2015-12-17 2017-06-22 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
WO2017152062A1 (en) 2016-03-04 2017-09-08 Gilead Sciences, Inc. Compositions and combinations of autotaxin inhibitors
WO2017177179A1 (en) 2016-04-08 2017-10-12 Gilead Sciences, Inc. Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
WO2017215590A1 (en) 2016-06-13 2017-12-21 I-Mab Anti-pd-l1 antibodies and uses thereof
WO2018026835A1 (en) 2016-08-04 2018-02-08 Gilead Sciences, Inc. Cobicistat for use in cancer treatments
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2018085069A1 (en) 2016-11-03 2018-05-11 Gilead Sciences, Inc. Combination of a bcl-2 inhibitor and a bromodomain inhibitor for treating cancer
WO2018097977A1 (en) 2016-11-22 2018-05-31 Gilead Sciences, Inc. Crystalline forms of a phosphate complex of a bet inhibitor
WO2018137598A1 (en) 2017-01-24 2018-08-02 I-Mab Anti-cd73 antibodies and uses thereof
EP4050030A1 (en) 2017-01-24 2022-08-31 I-Mab Biopharma US Limited Anti-cd73 antibodies and uses thereof
US10370381B2 (en) 2017-02-24 2019-08-06 Gilead Sciences, Inc. Inhibitors of bruton'S tyrosine kinase
US10314844B2 (en) 2017-02-24 2019-06-11 Gilead Sciences, Inc. Inhibitors of Bruton's tyrosine kinase
WO2018156901A1 (en) 2017-02-24 2018-08-30 Gilead Sciences, Inc. Inhibitors of bruton's tyrosine kinase
WO2018156895A1 (en) 2017-02-24 2018-08-30 Gilead Sciences, Inc. Inhibitors of bruton's tyrosine kinase
EP4026835A2 (en) 2017-04-20 2022-07-13 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2018195321A1 (en) 2017-04-20 2018-10-25 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
EP4227302A1 (en) 2018-02-13 2023-08-16 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019160882A1 (en) 2018-02-13 2019-08-22 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
US11149052B2 (en) 2018-04-06 2021-10-19 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′-cyclic dinucleotides
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
US11292812B2 (en) 2018-04-06 2022-04-05 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotides
WO2019204609A1 (en) 2018-04-19 2019-10-24 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
WO2019217780A1 (en) 2018-05-11 2019-11-14 Phosphorex, Inc. Microparticles and nanoparticles having negative surface charges
EP4029868A1 (en) 2018-05-14 2022-07-20 Gilead Sciences, Inc. Mcl-1 inhibitors
WO2019222112A1 (en) 2018-05-14 2019-11-21 Gilead Sciences, Inc. Mcl-1 inhibitors
WO2020014643A1 (en) 2018-07-13 2020-01-16 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
EP4234030A2 (en) 2018-07-13 2023-08-30 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2020072656A1 (en) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
WO2020086556A1 (en) 2018-10-24 2020-04-30 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
US11766447B2 (en) 2019-03-07 2023-09-26 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
WO2020263830A1 (en) 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
WO2021076908A1 (en) 2019-10-18 2021-04-22 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
WO2021087064A1 (en) 2019-10-31 2021-05-06 Forty Seven, Inc. Anti-cd47 and anti-cd20 based treatment of blood cancer
WO2021096860A1 (en) 2019-11-12 2021-05-20 Gilead Sciences, Inc. Mcl1 inhibitors
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
US11692038B2 (en) 2020-02-14 2023-07-04 Gilead Sciences, Inc. Antibodies that bind chemokine (C-C motif) receptor 8 (CCR8)
WO2021163064A2 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
WO2021222522A1 (en) 2020-05-01 2021-11-04 Gilead Sciences, Inc. Cd73 inhibiting 2,4-dioxopyrimidine compounds
WO2022221304A1 (en) 2021-04-14 2022-10-20 Gilead Sciences, Inc. CO-INHIBITION OF CD47/SIRPα BINDING AND NEDD8-ACTIVATING ENZYME E1 REGULATORY SUBUNIT FOR THE TREATMENT OF CANCER
WO2022245671A1 (en) 2021-05-18 2022-11-24 Gilead Sciences, Inc. Methods of using flt3l-fc fusion proteins
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023107954A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Antibodies targeting 5t4 and uses thereof
WO2023107956A1 (en) 2021-12-08 2023-06-15 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16 and 5t4
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
WO2023178181A1 (en) 2022-03-17 2023-09-21 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023183817A1 (en) 2022-03-24 2023-09-28 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023196784A1 (en) 2022-04-05 2023-10-12 Gilead Sciences, Inc. Combinations of antibody therapies for treating colorectal cancer
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
WO2024064668A1 (en) 2022-09-21 2024-03-28 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPα DISRUPTION ANTICANCER COMBINATION THERAPY

Similar Documents

Publication Publication Date Title
WO2013116562A1 (en) Compositions and methods of treating a disease with (s)-4 amino-6-((1-(5-chloro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)ethyl)amino)pyrimidine-5-carbonitrile
CN110382483A (en) Condensed N- heterocyclic compound and its application method
US8658668B2 (en) PI3 kinase/mTOR dual inhibitor
CN110036010A (en) The inhibitor of KRAS G12C mutain
CN110382482A (en) Condensed miscellaneous-Heterobicyclic compounds and its application method
US20130143902A1 (en) Compositions and methods of treating a proliferative disease with a quinazolinone derivative
EP1962843B1 (en) Use of parp-1 inhibitors
CN106456628A (en) Heterocyclic compounds for use in the treatment of pi3k-gamma mediated disorders
WO2010123931A1 (en) Methods of treatment for solid tumors
US11446301B2 (en) Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
US9464093B2 (en) Substituted imidazo[4',5':4,5]cyclopenta[1,2-e]pyrrolo[1,2-a]pyrazines and oxazolo[4',5':4,5]cyclopenta[1,2-e]pyrrolo[1,2-a]pyrazines for treating brain cancer
US20080269230A1 (en) Inhibitors of proton-gated cation channels and their use in the treatment of ischaemic disorders
AU775591B2 (en) Anxiety method
WO2017101777A1 (en) Pyrrolopyrimidine compound salt
CZ216092A3 (en) Application of xanthine derivative for treating secondary damage of neural cells and dysfunctions after injury of cranium and brain
US11434195B2 (en) Compositions and methods for treatment of central nervous system tumors
JP2001521895A (en) New metal complex
US11964982B1 (en) 5-(3-substituted phenyl)-pyrimido[4,5-d]pyrimidine-2,4,7(1H,3H,8H)-trione derivatives as anticancer agents
US11932649B1 (en) 5-(3-substituted phenyl)-pyrimido[4,5-d]pyrimidine-2,4,7(1H,3H,8H)-trione derivatives as anticancer agents
EA038527B1 (en) Method of treating a respiratory disease or condition, compositions and kits comprising a dual pi3k delta-gamma kinase inhibitor and a corticosteroid
CA3100923A1 (en) Pyrazolopyrimidine compounds as adenosine receptor antagonists
WO2022198315A1 (en) Methods of treating mental and/or mood disorders using 2-bromo-lsd alone or in combination with a mtor inhibitor
CN116113406A (en) GAS41 inhibitors and methods of use thereof
WO2004087120A2 (en) Use of quinazoline derivatives with src kinase inhibitory activity for the treatment of hypertension
WO2016003886A1 (en) 4-aminoquinoline compositions and methods for using same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13743414

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13743414

Country of ref document: EP

Kind code of ref document: A1