US20100202963A1 - Therapies for hematologic malignancies - Google Patents

Therapies for hematologic malignancies Download PDF

Info

Publication number
US20100202963A1
US20100202963A1 US12/618,612 US61861209A US2010202963A1 US 20100202963 A1 US20100202963 A1 US 20100202963A1 US 61861209 A US61861209 A US 61861209A US 2010202963 A1 US2010202963 A1 US 2010202963A1
Authority
US
United States
Prior art keywords
compound
cancer
cell
cells
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/618,612
Other languages
English (en)
Inventor
W. Michael Gallatin
Roger G. Ulrich
Neill A. Giese
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Calistoga LLC
Original Assignee
Calistoga Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Calistoga Pharmaceuticals Inc filed Critical Calistoga Pharmaceuticals Inc
Priority to US12/618,612 priority Critical patent/US20100202963A1/en
Assigned to CALISTOGA PHARMACEUTICALS, INC. reassignment CALISTOGA PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GIESE, NEILL A., GALLATIN, W. MICHAEL, ULRICH, ROGER G.
Publication of US20100202963A1 publication Critical patent/US20100202963A1/en
Assigned to GILEAD CALISTOGA LLC reassignment GILEAD CALISTOGA LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CALISTOGA PHARMACEUTICALS, INC.
Priority to US13/417,185 priority patent/US9492449B2/en
Priority to US13/762,238 priority patent/US9238070B2/en
Priority to US14/323,925 priority patent/US20140323439A1/en
Priority to US15/277,857 priority patent/US10154998B2/en
Priority to US16/189,435 priority patent/US20190083498A1/en
Priority to US16/989,685 priority patent/US20200368243A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • 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
    • 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
    • A61K31/4965Non-condensed pyrazines
    • 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
    • A61K31/33Heterocyclic 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
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • 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
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/745Polymers of hydrocarbons
    • A61K31/75Polymers of hydrocarbons of ethene

Definitions

  • the invention is in the field of therapeutics and medicinal chemistry.
  • the invention concerns uses of certain quinazoline derivatives for the treatment of hematologic malignancies and certain other conditions.
  • 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.
  • PI 3-kinase The initial purification and molecular cloning of PI 3-kinase revealed that it was a heterodimer consisting of p85 and p110 subunits.
  • Class I PI3Ks Four distinct Class I PI3Ks have been identified, designated PI3K ⁇ , ⁇ , ⁇ , and ⁇ , each consisting of a distinct 110 kDa catalytic subunit and a regulatory subunit. More specifically, three of the catalytic subunits, i.e., p110 ⁇ , p110 ⁇ and p110 ⁇ , each interact with the same regulatory subunit, p85; whereas p110 ⁇ interacts with a distinct regulatory subunit, p101. The patterns of expression of each of these PI3Ks in human cells and tissues are also distinct.
  • p110 ⁇ isoform of PI 3-kinase is described in Chantry et al., J Biol Chem, 272:19236-41 (1997). It was observed that the human p110 ⁇ isoform is expressed in a tissue-restricted fashion. It is expressed at high levels in lymphocytes and lymphoid tissues, suggesting that the protein might play a role in PI 3-kinase-mediated signaling in the immune system.
  • the p110 ⁇ isoform of PI3K may also play a role in PI3K-mediated signaling in certain cancers.
  • the present invention provides a class of quinazolinone type compounds and a method to use these compounds in the treatment of cancer, inflammatory, and autoimmune diseases.
  • cancers that are hematologic malignancies, such as leukemia and lymphoma are treated by the methods herein.
  • methods of using the quinazolinone compounds in combination with other therapeutic treatments in patients in need thereof are also provided.
  • the invention provides the use of a compound for the manufacture of a medicament for the treatment of a condition in a subject, wherein the condition is cancer or an autoimmune condition; wherein the compound is of formula A,
  • R is H, halo, or C1-C6 alkyl; R′ is C1-C6 alkyl; or a pharmaceutically acceptable salt thereof; and optionally a pharmaceutically acceptable excipient.
  • the compound is predominantly the S-enantiomer.
  • R is fluoro (F) and is attached to position 5 or 6 of the quinazolinyl ring.
  • R is H or F; and R′ is methyl, ethyl or propyl.
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-N-phenyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the autoimmune disease is allergic rhinitis, asthma, COPD, or rheumatoid arthritis.
  • the condition is cancer
  • the cancer is a hematological malignancy.
  • the hematological malignancy is leukemia.
  • the hematological malignancy is lymphoma.
  • the hematological malignancy is selected from the group consisting of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldenstrom's macroglobulinemia (WM), B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MM multiple myeloma
  • NHL non-Hodgkin's lymphoma
  • MCL mantle cell lymphoma
  • follicular lymphoma Waldenstrom's macroglobul
  • the cancer is acute lymphocytic leukemia (ALL).
  • ALL acute lymphocytic leukemia
  • the cancer is acute myeloid leukemia (AML).
  • the cancer is chronic lymphocytic leukemia (CLL).
  • CLL chronic lymphocytic leukemia
  • the cancer is multiple myeloma (MM).
  • the cancer is B-cell lymphoma.
  • the cancer is diffuse large B-cell lymphoma (DLBCL).
  • DLBCL diffuse large B-cell lymphoma
  • the cancer is B-cell or T-cell ALL.
  • the cancer is Hodgkin's lymphoma.
  • the cancer is breast, lung, colon, prostate or ovarian cancer.
  • the subject is refractory to chemotherapy treatment, or in relapse after treatment with chemotherapy.
  • the compound is prepared for administration with at least one additional therapeutic agent.
  • the additional therapeutic agent is a proteasome inhibitor.
  • the additional therapeutic agent is combined with the compound of Formula A.
  • the additional therapeutic agent is selected from the group consisting of bortezomib (Velcade®), carfilzomib (PR-171), PR-047, disulfiram, lactacystin, PS-519, eponemycin, epoxomycin, aclacinomycin, CEP-1612, MG-132, CVT 63417, PS-341, vinyl sulfone tripeptide inhibitors, ritonavir, PI-083, (+/ ⁇ ) 7 methylomuralide, ( ⁇ )-7-methylomuralide.
  • the additional therapeutic agent is bortezomib.
  • the compound is prepared for administration with at least a group of at least two agents, wherein said group of agents is selected from the groups consisting of a-q,
  • CHOP cyclophosphamide, doxorubicin, vincristine, prednisone
  • hyperCV AD hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine
  • FCM fludarabine, cyclophosphamide, mitoxantrone
  • R-FCM rituximab, fludarabine, cyclophosphamide, mitoxantrone
  • CVP cyclophosphamide, vincristine, prednisone
  • ICE iphosphamide, carboplatin, etoposide
  • FCR fludarabine, cyclophosphamide, rituximab
  • the compound of formula A is present in a pharmaceutical composition comprising the compound of formula A and at least one pharmaceutically acceptable excipient.
  • the invention provides the use of a compound for the manufacture of a medicament for the treatment of a condition in a subject, wherein the condition is selected from the group consisting of multiple myeloma, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), B-cell lymphoma, diffuse large B-cell lymphoma (DLBCL), B cell ALL, T cell ALL, Hodgkin's lymphoma, breast, and ovarian cancer, wherein the compound is a compound of formula I′′ or formula II′′:
  • the subject is refractory to chemotherapy treatment or in relapse after treatment with chemotherapy.
  • the subject has a cancer that constitutively expresses Akt phosphorylation activity.
  • the subject has a cancer with high p110 ⁇ activity and low p110 ⁇ activity.
  • the compound is used in combination with bortezomib.
  • the invention provides the use of a compound I′′ or H′′ in the manufacture of a medicament for treating a hematological cancer, wherein the medicament is prepared for administration with bortezomib or carfilzomib.
  • the compound maintains an average blood concentration above the EC50 level for PI3K ⁇ activation and below the level for EC50 PI3K ⁇ activation in basophils over a period of at least 12 hours from compound administration.
  • the compound maintains an average blood plasma concentration between 100 nM and 1100 nM over a period of at least 12 hours from compound administration.
  • the subject is resistant to standard chemotherapeutic treatments.
  • the subject has at least one enlarged lymph node.
  • the subject is refractory to at least two standard or experimental chemotherapy treatments had at least two prior chemotherapy treatments.
  • each chemotherapy treatment is selected from the group consisting of fludarabine, alkylating agents, rituximab, alemtuzumab, and the treatments a-q listed above.
  • FIG. 1 shows a graphical summary of multiple myeloma (MM) cell growth as a function of varying concentrations of cytokines IGF-1 and IL-6 in combination with compound I, using LB cells.
  • FIG. 2 shows a graphical summary of cell growth of multiple myeloma (MM) cells as a function of varying concentrations of compound I and the presence or absence of bone marrow stromal cells (BMSC) after 48 hours.
  • MM multiple myeloma
  • BMSC bone marrow stromal cells
  • FIG. 3 shows a graphical summary of apoptosis of Chronic Lymphocytic Leukemia (CLL) cells as a function of varying concentrations of compound of formula I.
  • CLL Chronic Lymphocytic Leukemia
  • FIG. 4 shows a summary chart of the effect of compound I on cell viability, reduction in Akt (Ser473) phosphorylation, and caspase 3 activation in several different Acute Lymphoblastic Leukemia (ALL) cell lines.
  • ALL Acute Lymphoblastic Leukemia
  • FIG. 5 shows a summary of the effect of compound I on the cell cycle of acute lymphoblastic leukemia (ALL) cell lines.
  • ALL acute lymphoblastic leukemia
  • FIG. 6 shows a graphical summary of the effect of varying concentration of compound I on cellular growth in breast cancer T47D and HS-578T cell lines at 48 hrs and 72 hrs.
  • FIG. 7 shows a graphical summary of the effect of varying concentrations of compound I on cellular growth of ovarian IGROV-1 and OVCAR-3 cell lines at 48 hrs and 72 hrs.
  • FIG. 8 shows a summary of the effect of compound I on Akt phosphorylation in many leukemia and lymphoma cell lines.
  • FIG. 9 shows SDS-PAGE images and displays of Akt and pAkt in various hematopoietic cancer cell lines as a function of the presence or absence of compound I, showing compound I inhibits Akt phosphorylation.
  • FIG. 10 shows graphical summaries of apoptotic and viable cell populations in breast cancer cell lines as a function of varying concentrations of compound formula I, demonstrating that the compound induces apoptosis.
  • FIG. 11 shows the concentration of compound I in the blood of a healthy human subject over 12 hours after oral administration of 50, 100 and 200 mg doses of said compound.
  • FIG. 12 shows the comparison of lesion areas in a human patient diagnosed with mantle cell lymphoma after 28 days (1 cycle) of treatment with compound I and lesion areas prior to treatment.
  • FIG. 13 shows the ALC (absolute lymphocyte count) in the blood of a patient over a period of 4 weeks after 28 days (1 cycle) of treatment with the compound of formula I.
  • FIG. 14 shows the concentration of compound I in the blood of patients with and without mantle cell lymphoma (MCL) over 6 hours after administration (50 mg BID) at day 28, compared to the concentration in the blood of a normal healthy volunteer at day 7 (D7) using the same dosing schedule or dosing with 100 mg BID of Compound I.
  • MCL mantle cell lymphoma
  • FIG. 15 shows PI3K isoform expression in a panel of lymphoma and leukemia cell lines.
  • FIG. 16A shows cell viability and apoptosis data in leukemia cell lines exposed to Compound I.
  • FIG. 16B the Annexin staining indicates an increase in apoptosis in the treated cells.
  • FIGS. 17A-D shows PAGE results of different PI3K isoform expression in CLL patient cells.
  • FIG. 18 shows the induction of (A) caspase 3 and (B) PARP cleavage in the presence of compound I.
  • FIG. 19 shows increased apoptosis of Chronic Lymphocytic Leukemia (CLL) cells from poor prognosis patients caused by exposure to compound I, demonstrating that compound I is effective in drug resistant patients.
  • CLL Chronic Lymphocytic Leukemia
  • FIG. 20 shows increased apoptosis of Chronic Lymphocytic Leukemia (CLL) cells from refractory/relapsed patients caused by exposure to the compound of formula I.
  • CLL Chronic Lymphocytic Leukemia
  • FIG. 21 shows the results of Phospho-Akt production in the absence or presence of 0.1, 1.0, 10 ⁇ M of compound I.
  • FIG. 22 shows flow cytometry results relating to PI3K signaling in basophils, demonstrating that (B) anti-FC ⁇ R1 or (C) fMLP increases CD63 expression compared to no stimulation (A).
  • FIG. 23 shows inhibition of PI3K inhibition by compound I in basophils, and demonstrates that Compound I is especially effective at inhibition of CD63 expression induced by a p110 ⁇ pathway, but also effective at micromolar concentration to inhibit expression induced by a p110 ⁇ pathway.
  • FIG. 24 shows pharmacokinetic data of (A) single dose administration of compound I at different dose amounts in healthy volunteers, and (B) a pharmacokinetic profile that maintains an effective dosage over a 12 hour period.
  • FIG. 25 shows the effects of various doses of compound I on (A) glucose and (B) insulin levels, exhibiting little off-target activity.
  • FIG. 26A shows the PI3K isoform expression in a panel of DLBCL cell lines.
  • FIG. 26 B shows an SDS-PAGE image of pAkt in DLBCL cell lines in the presence or absence of compound I.
  • FIG. 27 shows the effects of a 10 ⁇ M concentration of compound I on the phosphorylation of Akt and S6 in ALL cell lines in SDS-PAGE.
  • FIG. 28 shows a dose dependent reduction of phosphorylation of Akt, S6, and GSK-3 ⁇ after treatment with a series of compound I dilutions.
  • FIG. 29 shows dose dependent effects of compound I on ALL cell lines in the downregulation of cFLIP, cleavage of Caspase 3, and cleavage of PARP.
  • FIG. 30 shows expression of p110 ⁇ in A) MM cell lines and B) patient MM cells; and C) in MM.1S and LB cells.
  • FIG. 31A shows expression of p110 ⁇ from LB and INA-6 cells transfected with p110 ⁇ siRNA (Si) or control siRNA (mock).
  • FIG. 31B shows a graph of INA-6 cell growth after transfection with p110 ⁇ siRNA (Si) or control siRNA (mock).
  • FIG. 31C shows the % of viable cells cultured with or without compound I for 48 hours.
  • FIG. 31D shows the % of viable MM cells after being cultured with compound I at concentrations from 0 to 20 ⁇ M for 48 hours.
  • FIG. 31E shows the % of viable peripheral blood mononuclear cells from healthy donors after being cultured with compound I at various concentrations for 72 hours.
  • FIG. 31F shows immunoblotting results of lysates from INA-6 cells cultured with compound I (0-5 ⁇ M) for 120 hours.
  • FIG. 32 shows immunoblot AKT and ERK expression profiles after culturing of A) INA-6 cells with compound I or LY294002 for 12 hours; B) INA-6 and MM.1S cells with compound I at various concentrations for 6 hours; C) LB and INA-6 cells with compound I for 0-6 hours.
  • FIG. 33A shows fluorescent and transmission electron microscopic images of INA-6 and LB MM cells treated with compound I for 6 hours and LC3 accumulation; arrows indicate autophagosomes.
  • FIG. 33B shows fluorescence microscopy images of INA-6 cells treated with 5 ⁇ M of compound I or serum starvation for 6 hours.
  • FIG. 33C shows immunoblots of LC3 and beclin-1 protein levels from INA-6 cells treated with or without compound 1 and 3-MA (3-methyl adenine, a known inhibitor of autophagy).
  • FIG. 33D shows % growth of p110 ⁇ positive LB cells after treatment with up to 100 ⁇ M of 3-MA for 24 hours.
  • FIG. 34 shows the levels of growth inhibition of A) LB or B) INA-6 cells co-cultured with 0, 5, and 10 ⁇ M of compound I in the presence or absence of varying amounts of IL-6 or IGF-1; Legend: control media ( ⁇ ); compound I at 5.0 ⁇ M ( ⁇ ) or 10 ⁇ M ( ⁇ ).
  • FIGS. 34C and 34D show MM cell growth inhibition in the presence of BMSC.
  • control media
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • Compound I 2.5 ⁇ M
  • 5 ⁇ M
  • 10 ⁇ M
  • FIG. 34 E shows immunoblots of IL-6 in culture supernatants from BMSCs cultured with compound I or control media for 48 hours.
  • FIG. 34F shows immunoblots of AKT and ERK expression profiles in INA-6 cells treated with compound I cultured with our without BMSCs.
  • FIG. 34G shows % BMSC cell growth in two different patients after culturing with compound I for 48 hours.
  • FIG. 35A shows microscopic images of HuVECs (human umbilical vein endothelial cells) cultured with 0, 1 and 10 ⁇ M of compound I for 8 hours and microtubule formation assessed.
  • HuVECs human umbilical vein endothelial cells
  • FIG. 35B shows a bar chart summarizing endothelial cell tube formation in HuVEC cells treated with compound I.
  • FIG. 35 C shows a graph charting % cell growth of HuVECs as a function of the increasing culture concentration of compound I.
  • FIG. 35 D shows decreasing Akt and ERK expression of HuVEC cell lysates after being cultured with compound I for 8 hours.
  • FIG. 36A charts the tumor volume in SCID mice with human MM xenografts treated with 0, 10 mg/kg or 30 mg/kg of compound II as a function of time, showing strong in vivo activity on the human xenograft tumor
  • FIG. 36 B shows a photograph comparing the tumor from human MM xenografts on a mouse treated with compound II for 12 days to a control mouse.
  • FIG. 36C shows the survival rate of SCID mice with human MM xenografts treated with 0, 10, and 30 mg/kg compound II over time.
  • FIG. 36D shows images from immuno-histochemistric analysis of tumors harvested from a mouse treated with compound II in comparison to the control; wherein CD31 and P-AKT positive cells are dark brown.
  • FIG. 36E shows immunoblots detecting PDK-1 and AKT levels from tumor tissues harvested from mice treated with compound II in comparison to a control.
  • FIG. 36F shows a chart of sIL6R levels measured in mice treated with 0, 10 mg/kg or 30 mg/kg of compound II over a period of 4 weeks of treatment as determined by ELISA.
  • FIG. 37A show the % of viable LB or INA-6 MM cells after treatment with compound I with varying amounts of bortezomib (B); Legend: medium ( ⁇ ), compound I 1.25 ⁇ M ( ⁇ ), 2.5 ⁇ M ( ⁇ ), or 5.0 ⁇ M ( ⁇ ).
  • FIG. 37B shows immunoblots comparing levels of phosphorylation of AKT in INA-6 cells treated for 6 hours with compound I and/or bortezomib.
  • FIG. 38 shows (A) PI3K isoform expression in a panel of follicular lymphoma cell lines; (B) reduction in the expression of pAkt, Akt, pS6 and S6 after exposure to compound I; and (C) Increase in PARP and caspase-3 cleavage after exposure to compound I in a dose-dependent manner.
  • FIG. 39 shows (A) amounts of constitutive PI3K signaling in primary MCL cells in various amounts of compound I; (B) reduction in pAkt production in MCL cell lines containing a survival factor and varying amounts of compound I.
  • FIG. 40 show a computer tomography axillary image of a bulky lymphadenopathy in a patient with CLL (A) before treatment with compound I and (B) after 1 cycle of treatment with compound I.
  • the invention provides methods that relate to a novel therapeutic strategy for the treatment of cancer and inflammatory diseases.
  • the invention provides a method of treating cancer or an autoimmune disease in a subject comprising administering to said subject a compound of formula A
  • R is H, halo, or C1-C6 alkyl; R′ is C1-C6 alkyl; or a pharmaceutically acceptable salt thereof; and optionally a pharmaceutically acceptable excipient.
  • halo is F; and R′ is methyl, ethyl or propyl.
  • R is attached to position 5 of the quinazolinyl ring, having the structure
  • R is attached to position 6 of the quinazolinyl ring, having the structure
  • compound used herein, unless otherwise specified, refers to a compound of formula A, such as compound I, compound II, or an enantiomer, such as I′′ or II′′, or an enantiomeric mixture.
  • the compound of formula A is a compound of formula I. In another embodiment, the compound of formula A is a compound of formula II. In certain embodiments, the compound is a racemic mixture of R- and S-enantiomers. In certain embodiments, the compound is used as a mixture of enantiomers, and is often enriched with the S-enantiomer. In some embodiments, the compound is predominantly the S-enantiomer. In some embodiments, the compound of formula A, used in the methods described herein is at least 80% S-enantiomer. In certain embodiments, the compound is primarily composed of the S-enantiomer, wherein the compound comprises at least 66-95%, or 85-99% of the S-enantiomer.
  • the compound has an enantiomeric excess (e.e.) of at least 90% or at least 95% of S-enantiomer. In some embodiments the compound has an S-enantiomeric excess (e.e.) of at least 98% or at least 99%. In certain embodiments, the compound comprises at least 95% of the S-enantiomer. In the cellular and patient experiments provided in the Example section, the sample of compound I used was over 95% S-enantiomer.
  • the compound of formula I or II, used in the methods described herein is at least 80% S-enantiomer.
  • the compound of formula I or II is primarily composed of the S-enantiomer, wherein the compound comprises at least 66-95%, or 85-99% of the S-enantiomer.
  • the compound of formula I or II has an enantiomeric excess (e.e.) of at least 90% or at least 95% of S-enantiomer.
  • the compound of formula I or II has an S-enantiomeric excess (e.e.) of at least 98% or at least 99%.
  • the compound of formula I or II comprises at least 95% of the S-enantiomer. In the cellular and patient experiments provided in the Example section, the sample of compound I used was over 95% S-enantiomer.
  • the compound selectively inhibits PI3K p110 ⁇ compared to other PI3K isoforms.
  • the autoimmune disease is allergic rhinitis, asthma, COPD, or rheumatoid arthritis.
  • the cancer is a hematological malignancy and/or solid tumor.
  • the hematological malignancy is leukemia or lymphoma.
  • lymphoma is a mature (peripheral) B-cell neoplasm.
  • the mature B-cell neoplasm is selected from the group consisting of B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma; B-cell prolymphocytic leukemia; Lymphoplasmacytic lymphoma; Marginal zone lymphoma, such as Splenic marginal zone B-cell lymphoma (+/ ⁇ villous lymphocytes), Nodal marginal zone lymphoma (+/ ⁇ monocytoid B-cells), and Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT) type; Hairy cell leukemia; Plasma cell myeloma/plasmacytoma; Follicular lymphoma, follicle center; Mantle cell lymphoma; Diffuse large cell B-cell lymphoma (including Mediastinal large B-cell lympho
  • lymphoma is selected from the group consisting of multiple myeloma (MM) and non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldenstrom's macroglobulinemia (WM) or B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL).
  • MM multiple myeloma
  • NHL non-Hodgkin's lymphoma
  • MCL mantle cell lymphoma
  • follicular lymphoma follicular lymphoma
  • Waldenstrom's macroglobulinemia WM
  • B-cell lymphoma diffuse large B-cell lymphoma
  • leukemia is selected from the group consisting of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and small lymphocytic lymphoma (SLL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • Acute lymphocytic leukemia is also known as acute lymphoblastic leukemia and may be used interchangeably herein. Both terms describe a type of cancer that starts from the white blood cells, lymphocytes, in the bone marrow.
  • Non-Hodgkin's Lymphoma falls into one of two categories, aggressive NHL or indolent NHL. Aggressive NHL is fast growing and may lead to a patient's death relatively quickly. Untreated survival may be measured in months or even weeks.
  • Examples of aggressive NHL includes B-cell neoplasms, diffuse large B-cell lymphoma, T/NK cell neoplasms, anaplastic large cell lymphoma, peripheral T-cell lymphomas, precursor B-lymphoblastic leukemia/lymphoma, precursor T-lymphoblastic leukemia/lymphoma, Burkitt's lymphoma, Adult T-cell lymphoma/leukemia (HTLV1+), primary CNS lymphoma, mantle cell lymphoma, polymorphic post-transplantation lymphoproliferative disorder (PTLD), AIDS-related lymphoma, true histiocytic lymphoma, and blastic NK-cell lymphoma.
  • the most common type of aggressive NHL is diffuse large cell lymphoma.
  • Indolent NHL is slow growing and does not display obvious symptoms for most patients until the disease has progressed to an advanced stage. Untreated survival of patients with indolent NHL may be measured in years.
  • Non-limiting examples include follicular lymphoma, small lymphocytic lymphoma, marginal zone lymphoma (such as extranodal marginal zone lymphoma (also called mucosa associated lymphoid tissue—MALT lymphoma), nodal marginal zone B-cell lymphoma (monocytoid B-cell lymphoma), splenic marginal zone lymphoma), and lymphoplasmacytic lymphoma (Waldenstrom's macroglobulinemia).
  • follicular lymphoma small lymphocytic lymphoma
  • marginal zone lymphoma such as extranodal marginal zone lymphoma (also called mucosa associated lymphoid tissue—MALT lymphoma)
  • nodal marginal zone B-cell lymphoma nodal marginal zone B
  • histologic transformation may occur, e.g., indolent NHL in patients may convert to aggressive NHL.
  • the invention provides methods of treating a patient with aggressive NHL or indolent NHL.
  • the invention provides methods of treating a patient with a condition selected from the group consisting of mantle cell lymphoma (MCL), diffuse large B cell lymphoma (DLBCL), follicular lymphoma (FL), acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and small lymphocytic lymphoma (SLL), multiple myeloma (MM), and marginal zone lymphoma.
  • MCL mantle cell lymphoma
  • DLBCL diffuse large B cell lymphoma
  • FL follicular lymphoma
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MM multiple myeloma
  • marginal zone lymphoma MM
  • the methods of the invention are administered to patients with relapsed or refractory conditions.
  • the cancer is breast, lung, colon or prostate cancer.
  • the cancer or autoimmune disease is associated with abnormal PI3K activity compared to PI3K activity in a subject without cancer or without an autoimmune disease.
  • the preferred subject is refractory to chemotherapy treatment, or in relapse after treatment with chemotherapy.
  • the subject is a de novo patient.
  • the method comprises reducing the level of PI3K ⁇ activity in said patient.
  • the subject is a human subject.
  • Subjects that undergo treatment with known therapeutic agents may experience resistance to treatment.
  • bortezomib was FDA approved for relapsed/refractory, relapsed, and newly diagnosed MM, some patients do not respond and others acquire resistance to bortezomib.
  • the quinazolinone compound described herein synergistically augments efficacy of a known therapeutic agent.
  • the compounds described herein can augment any of the therapeutic agents described herein.
  • the compounds described herein synergistically augment proteasome inhibitors.
  • the subject is resistant to chemotherapeutic treatment.
  • the subject is resistant to proteasome inhibitors.
  • the subject is resistant bortezomib or carfilzomib.
  • the compounds described herein synergistically augment bortezomib-induced MM cytotoxicity.
  • the compounds discussed herein inhibit bortezomib-induced phosphorylation of AKT.
  • the methods described herein are used to overcome resistance to proteasome inhibitor treatment.
  • the invention provides a method to treat a subject that is resistant or has developed a resistance to therapeutic agents.
  • the synergistic effects between a compound of formula A and conventional therapies may be attributed to the ability of the compound of the invention to induce tumor cell mobilization into peripheral circulation. Inducing the peripheral circulation of the tumor cells increases the ability of conventional therapy to act upon and more effectively neutralize the tumor. This synergy has been demonstrated in CLL patients.
  • the method comprises administering in addition to a compound of formula A to a patient, a therapeutically effective amount of at least one additional therapeutic agent and/or a therapeutic procedure selected to treat said cancer or autoimmune disease in said patient.
  • “Therapeutic agent” may refer to one or more compounds, as used herein.
  • the therapeutic agent may be a standard or experimental chemotherapy drug.
  • the therapeutic agent may comprise a combination of more than one chemotherapy drug. Typical chemotherapy drug combinations are listed a-q herein.
  • a particular therapeutic agent may be chosen depending on the type of disease being treated. Non-limiting examples of conventional chemotherapeutic treatments for particular hematologic disease are described in later sections.
  • the invention provides a method to treat a hematopoietic cancer patient, e.g., a CLL patient, with bortezomib and a compound of formula A (e.g., formula I, II, I′′, or II′′), wherein the combination provides a synergistic effect.
  • a hematopoietic cancer patient e.g., a CLL patient
  • bortezomib e.g., formula I, II, I′′, or II′′
  • said therapeutic agent is selected from the following group consisting of bortezomib (Velcade®), carfilzomib (PR-171), PR-047, disulfiram, lactacystin, PS-519, eponemycin, epoxomycin, aclacinomycin, CEP-1612, MG-132, CVT-63417, PS-341, vinyl sulfone tripeptide inhibitors, ritonavir, PI-083, (+/ ⁇ )-7-methylomuralide, ( ⁇ )-7-methylomuralide, perifosine, rituximab, sildenafil citrate (Viagra®), CC-5103, thalidomide, epratuzumab (hLL2-anti-CD22 humanized antibody), simvastatin, enzastaurin, Campath-1H®, dexamethasone, DT PACE, oblimersen, antineoplaston A10
  • the therapeutic agent is preferably a proteasome inhibitor.
  • the methods comprise administering a compound with a proteasome inhibitor.
  • proteasome inhibitors include natural and synthetic compounds.
  • Non-limiting examples of proteasome inhibitors include bortezomib, ([(1R)-3-methyl-1-( ⁇ (2S)-3-phenyl-2-[(pyrazin-2-ylcarbonyl)amino]propanoyl ⁇ amino)butyl]boronic acid), which is marketed as ‘Velcade®’ by Millennium pharmaceuticals; carfilzomib (PR-171) and the oral analog, PR-047, both of which are developed by Proteolix, Inc.
  • proteasome inhibitors include disulfiram; lactacystin; synthetic compounds such as PS-519, eponemycin, epoxomycin, and aclacinomycin; calpain inhibitors, such as CEP-1612, MG-132, CVT-63417, PS-341; vinyl sulfone tripeptide inhibitors; ritonavir; PI-083; (+/ ⁇ )-7-methylomuralide; and ( ⁇ )-7-methylomuralide.
  • the compound of formula A is administered in combination with bortezomib or carfilzomib.
  • the compound of formula I is administered in combination with bortezomib or carfilzomib.
  • the compound of formula II is administered in combination with bortezomib or carfilzomib.
  • the invention provides a pharmaceutical composition comprising a compound of Formula I:
  • composition is enriched with the S-enantiomer.
  • the invention provides a pharmaceutical composition comprising a compound of Formula II:
  • composition is enriched with the S-enantiomer.
  • the invention provides a method of treating multiple myeloma (MM) in a patient comprising administering a combination of a compound of formula A and an additional therapeutic agent.
  • formula A is compound I or II.
  • formula A is compound I′′.
  • formula A is compound II′′.
  • the additional therapeutic agent is a proteasome inhibitor.
  • the additional therapeutic agent is bortezomib.
  • the method of treating multiple myeloma in a patient comprises administering compound I′′ with bortezomib.
  • the method of treating multiple myeloma in a patient comprises administering compound II′′ with bortezomib.
  • compound I′′ or II′′ has an enantiomeric excess of at least 60%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 70%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 80%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 90%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 95%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 98%.
  • compound I′′ or II′′ has an enantiomeric excess of at least 99%.
  • a combination of therapeutic agents is administered with a compound of Formula A, wherein said combination is selected from the group consisting of
  • CHOP cyclophosphamide, doxorubicin, vincristine, prednisone
  • hyperCV AD hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine
  • FCM fludarabine, cyclophosphamide, mitoxantrone
  • R-FCM rituximab, fludarabine, cyclophosphamide, mitoxantrone
  • CVP cyclophosphamide, vincristine, prednisone
  • ICE iphosphamide, carboplatin, etoposide
  • FCR fludarabine, cyclophosphamide, rituximab
  • the compound is used in combination with a therapeutic procedure.
  • the therapeutic procedure is selected from the group consisting of peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, total body irradiation, infusion of stem cells, bone marrow ablation with stem cell support, in vitro-treated peripheral blood stem cell transplantation, umbilical cord blood transplantation, immunoenzyme technique, immunohistochemistry staining method, pharmacological study, low-LET cobalt-60 gamma ray therapy, bleomycin, conventional surgery, radiation therapy, high-dose chemotherapy and nonmyeloablative allogeneic hematopoietic stem cell transplantation.
  • the method further comprises obtaining a biological sample from said patient; and analyzing said 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 or a combination thereof.
  • 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 or a combination thereof.
  • alkyl includes straight-chain, branched-chain and cyclic monovalent hydrocarbyl radicals, and combinations of these, which contain only C and H when they are unsubstituted. Examples include methyl, ethyl, isobutyl, cyclohexyl, cyclopentylethyl, and the like.
  • the total number of carbon atoms in each such group is sometimes described herein, e.g., when the group can contain up to ten carbon atoms it can be represented as 1-10C or as C1-C10 or C1-10.
  • Halo as used herein, includes fluoro, chloro, bromo and iodo. Fluoro and chloro are often preferred.
  • selective PI3K ⁇ inhibitor refers to a compound that inhibits the PI3K ⁇ or PI3 ⁇ isozyme, respectively, more effectively than at least one other isozymes of the PI3K family.
  • the selective inhibitor may also be active against other isozymes of PI3K, but requires higher concentrations to achieve the same degree of inhibition of the other isozymes.
  • Selective can also be used to describe a compound that inhibits a particular PI3-kinase more so than a comparable compound.
  • a “selective PI3K ⁇ inhibitor” compound is understood to be more selective for PI3K ⁇ than compounds conventionally and generically designated PI3K inhibitors, e.g., wortmannin or LY294002. Concomitantly, wortmannin and LY294002 are deemed “nonselective PI3K inhibitors.”
  • compounds of any type that selectively negatively regulate PI3K ⁇ expression or activity can be used as selective PI3K ⁇ inhibitors in the methods of the invention.
  • compounds of any type that selectively negatively regulate PI3K ⁇ expression or activity and that possess acceptable pharmacological properties can be used as selective PI3K ⁇ inhibitors in the therapeutic methods of the invention.
  • targeting p110 delta inhibition with a compound of the invention provides a novel approach for the treatment of hematological malignancies because this method inhibits constitutive signaling resulting in direct destruction of the tumor cell.
  • p110 delta inhibition represses microenvironmental signals which are crucial for tumor cell homing, survival and proliferation.
  • compounds of any type that selectively negatively regulate PI3K ⁇ expression or activity can be used as selective PI3K ⁇ inhibitors in the methods of the invention.
  • compounds of any type that selectively negatively regulate PI3K ⁇ expression or activity and that possess acceptable pharmacological properties can be used as selective PI3K ⁇ inhibitors in the therapeutic methods of the invention.
  • Treating refers to inhibiting a disorder, i.e., arresting its development; relieving the disorder, i.e., causing its regression; or ameliorating the disorder, i.e., reducing the severity of at least one of the symptoms associated with the disorder.
  • treating refers to preventing a disorder from occurring in an animal that can be predisposed to the disorder, but has not yet been diagnosed as having it.
  • disorder is intended to encompass medical disorders, diseases, conditions, syndromes, and the like, without limitation.
  • Autoimmune disease refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents.
  • the invention includes a method for suppressing a function of basophils and/or mast cells, and thereby enabling treatment of diseases or disorders characterized by excessive or undesirable basophil and/or mast cell activity.
  • a compound of the invention can be used that selectively inhibits the expression or activity of phosphatidylinositol 3-kinase delta (PI3K ⁇ ) in the basophils and/or mast cells.
  • the method employs a PI3K ⁇ inhibitor in an amount sufficient to inhibit stimulated histamine release by the basophils and/or mast cells.
  • PI3K ⁇ selective inhibitors can be of value in treating diseases characterized by histamine release, i.e., allergic disorders, including disorders such as chronic obstructive pulmonary disease (COPD), asthma, ARDS, emphysema, and related disorders.
  • allergic disorders including disorders such as chronic obstructive pulmonary disease (COPD), asthma, ARDS, emphysema, and related disorders.
  • COPD chronic obstructive pulmonary disease
  • asthma asthma
  • ARDS emphysema
  • emphysema emphysema
  • the present invention enables methods of treating such diseases as arthritic diseases, such as rheumatoid arthritis, psoriatic arthritis, monoarticular arthritis, osteoarthritis, gouty arthritis, spondylitis; Behçet disease; sepsis, septic shock, endotoxic shock, gram negative sepsis, gram positive sepsis, and toxic shock syndrome; multiple organ injury syndrome secondary to septicemia, trauma, or hemorrhage; ophthalmic disorders such as allergic conjunctivitis, vernal conjunctivitis, uveitis, and thyroid-associated opthalmopathy; eosinophilic granuloma; pulmonary or respiratory disorders such as asthma, chronic bronchitis, allergic rhinitis, ARDS, chronic pulmonary inflammatory disease (e.g., chronic obstructive pulmonary disease), silicosis, pulmonary sarcoidosis, pleurisy, alveolitis, vasculitis
  • the method can have utility in treating subjects who are or can be subject to reperfusion injury, i.e., injury resulting from situations in which a tissue or organ experiences a period of ischemia followed by reperfusion.
  • ischemia refers to localized tissue anemia due to obstruction of the inflow of arterial blood.
  • Transient ischemia followed by reperfusion characteristically results in neutrophil activation and transmigration through the endothelium of the blood vessels in the affected area. Accumulation of activated neutrophils in turn results in generation of reactive oxygen metabolites, which damage components of the involved tissue or organ.
  • reperfusion injury is commonly associated with conditions such as vascular stroke (including global and focal ischemia), hemorrhagic shock, myocardial ischemia or infarction, organ transplantation, and cerebral vasospasm.
  • vascular stroke including global and focal ischemia
  • hemorrhagic shock myocardial ischemia or infarction
  • organ transplantation organ transplantation
  • cerebral vasospasm cerebral vasospasm.
  • reperfusion injury occurs at the termination of cardiac bypass procedures or during cardiac arrest when the heart, once prevented from receiving blood, begins to reperfuse. It is expected that inhibition of PI3K ⁇ activity will result in reduced amounts of reperfusion injury in such situations.
  • the invention provides methods to treat a solid tumor.
  • the cancer is breast, lung, colon, or prostate cancer.
  • the invention provides methods to treat a solid tumor that is associated with abnormal or undesirable cellular signaling activity mediated by PI3K ⁇ .
  • a solid tumor is selected from the group consisting of pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, including metastatic breast cancer; prostate cancer, including androgen-dependent and androgen-independent prostate cancer; renal cancer, including, e.g., metastatic renal cell carcinoma; hepatocellular cancer; lung cancer, including, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma (BAC), and adenocarcinoma of the lung; ovarian cancer, including, e.g., progressive epithelial or primary peritoneal cancer; cervical cancer; gastric cancer; esophageal cancer; head and neck cancer, including, e.g., squamous cell carcinoma of the head and neck; melanoma; neuroendocrine cancer, including metastatic neuroendocrine tumors; brain tumors, including, e.g., glioma, anaplastic oligodendroglioma, adult glioblastom
  • B cell function is most affected and includes proliferation, differentiation, apoptosis, and response to B cell survival factors (BCR, CD40, IL-4, chemokines).
  • BCR BCR, CD40, IL-4, chemokines
  • the potential clinical indication includes cancer but clinical adverse events include hyperinsulinemia in cancer patients.
  • the advantage of a p110 ⁇ selective inhibitor which targets cells mediating inflammation and cancer cells, wherein potential clinical indication include cancer, rheumatoid arthritis, asthma, allergies and COPD, is that treatment is well tolerated, and side effects like hyperinsulinemia are avoided.
  • the invention provides a method to treat patients having insulin resistance, or type 2 diabetes, for cancer, rheumatoid arthritis, asthma, allergies, COPD, or other conditions treatable with the compounds of the invention.
  • the compounds of the invention are particularly advantageous over pan-PI3K inhibitors.
  • a compound of formula I or I′′ is preferred because it provides therapeutic benefits to treating hematologic malignancies without adversely affecting insulin signaling.
  • the invention relates to methods of inhibiting PI3K p110 ⁇ . In another embodiment, the invention relates to methods of inhibiting PI3K p110 ⁇ or p110 ⁇ .
  • the method described herein has little or no off target activity.
  • compound of formula I used in the method show little activity against over 300 protein kinases including those summarized in Table 3 of Example 16.
  • the method described herein has no or minimal hyperinsulinemia effects in cancer patients compared to methods comprising the administration of pan-PI3K inhibitors.
  • the method described herein is useful in targeting cells mediating Akt phosphorylation, because the compounds of Formula A inhibit Akt phosphorylation. Suitable patients for treatment with the compounds of the invention can thus be selected, in one embodiment, by selecting a patient exhibiting elevated Akt phosphorylation associated with a hematopoietic cancer such as lymphoma, leukemia or multiple myeloma.
  • the methods herein avoid off-target liabilities and are characterized by negative results in receptor gram screens, having no hERG inhibition and no significant P450 inhibition.
  • Another advantage of the inventive method is the absence of adverse cardiovascular, respiratory, or central nervous system effects as demonstrated in safety pharmacology studies.
  • a 28-day toxicity study in rats and dogs demonstrated a high therapeutic index, e.g., a NOAEL (no observable adverse effect level)>>10 ⁇ M.
  • NOAEL no observable adverse effect level
  • Adverse effects are defined as any effects that result in functional impairment and/or pathological lesions that may affect the performance of the whole organism or that reduce an organism's ability to respond to an additional challenge.
  • the inventive methods are non-genotoxic in a standard battery of tests.
  • Another advantage of the invention is that compound selectivity for one or two PI3K isoforms results in an improved safety profile over compounds having pan-PI3K inhibition.
  • compound I has a favorable pharmacokinetic profile with good target coverage, and no adverse effects on glucose or insulin levels, and is well tolerated at doses above commonly used therapeutic doses by normal healthy volunteers.
  • Another advantage of the invention includes the ability to treat a wide range of hematological malignancies as demonstrated by the examples herein.
  • the methods of the invention are directed towards treating a cancer or an autoimmune disease.
  • the cancer is a hematological malignancy.
  • the hematological malignancy is selected from the group consisting of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and non-Hodgkin lymphoma (NHL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • MM multiple myeloma
  • NHL non-Hodgkin lymphoma
  • the non-Hodgkin lymphoma is selected from the group consisting of large diffuse B-cell lymphoma (LDBCL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM) and lymphoplasmacytic lymphoma.
  • LLBCL large diffuse B-cell lymphoma
  • MCL mantle cell lymphoma
  • WM Waldenstrom's macroglobulinemia
  • lymphoplasmacytic lymphoma is selected from the group consisting of large diffuse B-cell lymphoma (LDBCL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM) and lymphoplasmacytic lymphoma.
  • PI3K is implicated in many hematological malignancies and preclinical proof of concept relating to treatment with compound I has been established.
  • the table below summarizes particular hematological malignancies and the method of action on the primary patient cell or disease cell line.
  • a Chronic Lymphocytic Leukemia Primary patient cells Induces apoptosis Blocks survival factors
  • AML Primary patient cells Blocks PI3K signaling Inhibits proliferation
  • Acute Lymphocytic Leukemia (ALL) Cell Lines Blocks PI3K signaling Induces apoptosis Non-Hodgkin's Lymphomas (NHL) Cell Lines (MCL, DLBCL, FL) Blocks PI3K signaling Induces apoptosis Multiple Myeloma (MM) Primary patient cells P110 ⁇ overexpressed in 24/24 samples Induces apoptosis
  • FIG. 15 demonstrates that p110 ⁇ is prevalent in most lymphoma cell lines, while p110 ⁇ is not generally observed.
  • FIG. 16A shows that cell cultures from six different leukemia cell lines were sensitive to Compound I, and were strongly affected by 5-10 micromolar concentrations of this compound.
  • FIGS. 8 and 9 support compound I as reducing Akt(Ser473) production in several cell lines.
  • Example 3 shows dose-dependent cytotoxicity for compound I ( FIG. 3 ), in CLL cells, including cells taken from poor prognosis patients ( FIG. 19 ), and cells from patients shown to be resistant to other CLL treatments ( FIG. 20 ).
  • Example 13 and FIG. 13 demonstrate that compound I administered to a CLL patient at a rate of 50 mg BID for a 28-day cycle provides a significant therapeutic effect. An ALC concentration percent decrease in lymphocytes is observed.
  • the invention provides methods for treating CLL patients with drug-resistant CLL using compounds of Formula A.
  • Example 17 suggests that a fibroblast cell line relying mainly on p110 ⁇ for signaling was not sensitive to Compound I.
  • patient selection can include excluding patients having a cancer that relies mainly on p110 ⁇ for signaling.
  • the compounds of Formula A are also useful to treat lymphoma, including both B-cell and T-cell lymphomas.
  • Data in FIG. 4 demonstrates that six different ALL cell lines were sensitive to Compound I, which caused a significant reduction in cell viability in all six cell lines.
  • FIG. 12 and Example 12 demonstrate that mantle cell lymphoma patients treated with 50 mg BID of Compound I for 28 days experienced on average a 44% decrease in tumor burden.
  • FIG. 14 demonstrates that an MCL patient at the end of the 28 day cycle experienced similar plasma levels of Compound I following administration of a 50 mg dose to that observed in a normal healthy volunteer (NHV); thus the compound does not build up excessively over the course of a cycle of treatment, nor does the patient become tolerant by increased metabolism over the course of a treatment cycle.
  • NMV normal healthy volunteer
  • the compounds of Formula A, or Formula I are useful to treat hematopoietic cancers that constitutively express Akt phosphorylation activity.
  • Example 8 and FIGS. 8 and 9 list cancer cell lines that demonstrate constitutive Akt phosphorylation, including B-cell lymphomas, T-cell lymphomas, ALL, malignant histiocytosis, DLBCL and AML. Exposure of the cell to compound I results in the reduction of Akt phosphorylation. See also Example 19, which shows that constitutive Akt phosphorylation was inhibited by Compound I in 13 of 13 cell lines.
  • the cancer is a solid tumor.
  • the cancer is breast, ovarian, lung, colon, or prostate cancer.
  • FIG. 6 shows that Compound I reduces cellular proliferation of two breast cancer cell lines
  • FIG. 10 illustrates cytotoxicity to three different breast cancer cell lines.
  • FIG. 7 demonstrates that Compound I is cytotoxic to two ovarian cancer cell lines.
  • a compound of Formula A that expresses good activity (e.g., IC 50 less than about 1 ⁇ M, and preferably less than about 250 nM—see Example 15) against p110 ⁇ , since solid tumors often utilize this isozyme rather than or more than p110 ⁇ .
  • a compound of formula A that has an IC 50 less than about 250 nM is preferred for treatment of a solid tumor; compound I, I′′, II, or II′′ is suitable for this use, as demonstrated herein.
  • the subject for treatments described herein as one who has been diagnosed with at least one of the conditions described herein as treatable by the use of a compound of Formula A.
  • the subject has been diagnosed with a cancer named herein, and has proven refractory to treatment with at least one conventional chemotherapeutic agent.
  • treatments such as proteasome inhibitors, autologous stem cell transplant, CHOP regimens, rituximab, fludarabine, alemtuzumab, conventional anticancer nucleoside analogues and alkylating agents frequently respond to the methods of treatment described herein.
  • the treatments of the invention are directed to patients who have received one or more than one such treatment.
  • the autoimmune disease is allergic rhinitis, asthma, chronic obstructive pulmonary disease (COPD), or rheumatoid arthritis.
  • COPD chronic obstructive pulmonary disease
  • the methods of the invention are directed to B-cell, or B lymphocyte, related diseases.
  • B-cells play a role in the pathogenesis of autoimmune diseases.
  • the compounds of Formula A are suitable for treating a variety of subjects having the conditions described herein, especially hematological cancers in humans.
  • the subject selected for treatment of a hematological malignancy that is a subject experiencing relapse after other treatments or is refractory to other treatments.
  • the subject is selected for treatment of a hematological malignancy that is resistant to other cancer drugs.
  • the subject is selected for treatment of a hematological malignancy that exhibits a high level of p110 ⁇ activity.
  • the subject is selected for treatment of a hematological malignancy that exhibits a relatively low level of p110 ⁇ activity.
  • the subject is selected for treatment of a hematological malignancy that constitutively expresses Akt phosphorylation activity.
  • the method described herein comprises administering to a subject a compound of formula A described herein, in combination with a therapy used to treat cancer or an autoimmune disease.
  • “Therapy” or “treatment”, as used herein, is a treatment of cancer or an autoimmune disease by any well-known conventional or experimental form of treatment used to treat cancer or an autoimmune disease that does not include the use of a compound of formula A.
  • the combination of a compound of formula A with a conventional or experimental therapy used to treat cancer or an autoimmune disease provides beneficial and/or desirable treatment results superior to results obtained by treatment without the combination.
  • therapies used to treat cancer or an autoimmune disease are well-known to a person having ordinary skill in the art and are described in the literature. Therapies include, but are not limited to, chemotherapy, combinations of chemotherapy, biological therapies, immunotherapy, radioimmunotherapy, and the use of monoclonal antibodies, and vaccines.
  • the combination method provides for a compound of formula A administered simultaneously with or during the period of administration of the therapy. In some of the foregoing embodiments the compound of formula A is administered simultaneously with the other chemotherapeutic treatment. In certain embodiments, the combination method provides for a compound of formula A administered prior to or after the administration of the therapy.
  • the subject is refractory to at least one standard or experimental chemotherapy. In some of the foregoing embodiments, the subject is refractory to at least two standard or experimental chemotherapies. In some of the foregoing embodiments, the subject is refractory to at least three standard or experimental chemotherapies. In some of the foregoing embodiments, the subject is refractory to at least four standard or experimental chemotherapies.
  • the subject is refractory to at least one standard or experimental chemotherapy selected from the group consisting of fludarabine, rituximab, alkylating agents, alemtuzumab and the chemotherapy treatments a-q listed above.
  • the subject is refractory to at least two standard or experimental chemotherapies selected from the group consisting of fludarabine, rituximab, alkylating agents, alemtuzumab and the chemotherapy treatments a-q listed above.
  • the subject is refractory to at least three standard or experimental chemotherapies selected from the group consisting of fludarabine, rituximab, alkylating agents, alemtuzumab and the chemotherapy treatments a-q listed above.
  • the subject is refractory to at least four standard or experimental chemotherapies selected from the group consisting of fludarabine, rituximab, alkylating agents, alemtuzumab and the chemotherapy treatments a-q listed above.
  • Non-limiting examples of experimental or standard therapy are described below.
  • treatment of certain lymphomas is reviewed in Cheson, B. D., Leonard, J. P., “Monoclonal Antibody Therapy for B-Cell Non-Hodgkin's Lymphoma” The New England Journal of Medicine 2008, 359(6), p. 613-626; and Wierda, W. G., “Current and Investigational Therapies for Patients with CLL” Hematology 2006, p. 285-294. Lymphoma incidence patterns in the United States is profiled in Morton, L. M., et al. “Lymphoma Incidence Patterns by WHO Subtype in the United States, 1992-2001” Blood 2006, 107(1), p. 265-276.
  • Treatment of non-Hodgkin's lymphomas, especially of B cell origin include, but are not limited to use of monoclonal antibodies, standard chemotherapy approaches (e.g., CHOP, CVP, FCM, MCP, and the like), radioimmunotherapy, and combinations thereof, especially integration of an antibody therapy with chemotherapy.
  • standard chemotherapy approaches e.g., CHOP, CVP, FCM, MCP, and the like
  • Non-limiting examples of unconjugated monoclonal antibodies for Non-Hodgkin's lymphoma/B-cell cancers include rituximab, alemtuzumab, human or humanized anti-CD20 antibodies, lumiliximab, anti-TRAIL, bevacizumab, galiximab, epratuzumab, SGN-40, and anti-CD74.
  • Non-limiting examples of experimental antibody agents used in treatment of Non-Hodgkin's lymphoma/B-cell cancers include ofatumumab, ha20, PRO131921, alemtuzumab, galiximab, SGN-40, CHIR-12.12, epratuzumab, lumiliximab, apolizumab, milatuzumab, and bevacizumab. Any of the monoclonal antibodies can be combined with rituximab, fludarabine, or a chemotherapy agent/regimen.
  • Non-limiting examples of standard regimens of chemotherapy for Non-Hodgkin's lymphoma/B-cell cancers include CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), FCM (fludarabine, cyclophosphamide, mitoxantrone), CVP (cyclophosphamide, vincristine and prednisone), MCP (mitoxantrone, chlorambucil, and prednisolone), R-CHOP (rituximab plus CHOP), R-FCM (rituximab plus FCM), R-CVP (rituximab plus CVP), and R-MCP (R-MCP).
  • CHOP cyclophosphamide, doxorubicin, vincristine, prednisone
  • FCM fludarabine, cyclophosphamide, mitoxantrone
  • CVP cyclophosphamide, vincri
  • Non-limiting examples of radioimmunotherapy for Non-Hodgkin's lymphoma/B-cell cancers include yttrium-90-labeled ibritumomab tiuxetan, and iodine-131-labeled tositumomab. These therapeutic agents are approved for use in subjects with relapsed or refractory follicular or low-grade lymphoma.
  • Therapeutic treatments for mantle cell lymphoma include combination chemotherapies such as CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), hyperCV AD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine) and FCM (fludarabine, cyclophosphamide, mitoxantrone).
  • these regimens can be supplemented with the monoclonal antibody rituximab (Rituxan) to form combination therapies R-CHOP, hyperCV AD-R, and R-FCM.
  • Other approaches include combining any of the abovementioned therapies with stem cell transplantation or treatment with ICE (iphosphamide, carboplatin and etoposide).
  • Another approach to treating mantle cell lymphoma includes immunotherapy such as using monoclonal antibodies like Rituximab (Rituxan).
  • Rituximab is also effective against other indolent B-cell cancers, including marginal-zone lymphoma, WM, CLL and small lymphocytic lymphoma.
  • a combination of Rituximab and chemotherapy agents is especially effective.
  • a modified approach is radioimmunotherapy, wherein a monoclonal antibody is combined with a radioisotope particle, such as Iodine-131 tositumomab (Bexxar®) and Yttrium-90 ibritumomab tiuxetan (Zevalin®).
  • Bexxar® is used in sequential treatment with CHOP.
  • Another immunotherapy example includes using cancer vaccines, which is based upon the genetic makeup of an individual patient's tumor.
  • a lymphoma vaccine example is GTOP-99 (MyVax®).
  • Another approach to treating mantle cell lymphoma includes autologous stem cell transplantation coupled with high-dose chemotherapy.
  • Another approach to treating mantle cell lymphoma includes administering proteasome inhibitors, such as Velcade® (bortezomib or PS-341), or antiangiogenesis agents, such as thalidomide, especially in combination with Rituxan.
  • proteasome inhibitors such as Velcade® (bortezomib or PS-341)
  • antiangiogenesis agents such as thalidomide
  • Another treatment approach is administering drugs that lead to the degradation of Bcl-2 protein and increase cancer cell sensitivity to chemotherapy, such as oblimersen (Genasense) in combination with other chemotherapeutic agents.
  • Another treatment approach includes administering mTOR inhibitors, which can lead to inhibition of cell growth and even cell death; a non-limiting example is Temsirolimus (CCI-779), and Temsirolimus in combination with Rituxan®, Velcade® or other chemotherapeutic agents.
  • Non-limiting examples include Flavopiridol, PD0332991, R-roscovitine (Selicilib, CYC202), Styryl sulphones, Obatoclax (GX15-070), TRAIL, Anti-TRAIL DR4 and DR5 antibodies, Temsirolimus (CCI-779), Everolimus (RAD001), BMS-345541, Curcumin, Vorinostat (SAHA), Thalidomide, lenalidomide (Revlimid®, CC-5013), and Geldanamycin (17-AAG).
  • Flavopiridol PD0332991, R-roscovitine (Selicilib, CYC202), Styryl sulphones, Obatoclax (GX15-070), TRAIL, Anti-TRAIL DR4 and DR5 antibodies, Temsirolimus (CCI-779), Everolimus (RAD001), BMS-345541, Curcumin, Vorinostat (SAHA
  • Non-limiting examples of other therapeutic agents used to treat Waldenstrom's Macroglobulinemia include perifosine, bortezomib (Velcade®), rituximab, sildenafil citrate (Viagra®), CC-5103, thalidomide, epratuzumab (hLL2-anti-CD22 humanized antibody), simvastatin, enzastaurin, campath-1H, dexamethasone, DT PACE, oblimersen, antineoplaston A10, antineoplaston AS2-1, alemtuzumab, beta alethine, cyclophosphamide, doxorubicin hydrochloride, prednisone, vincristine sulfate, fludarabine, filgrastim, melphalan, recombinant interferon alfa, carmustine, cisplatin, cyclophosphamide, cytarabine, e
  • Non-limiting examples of other therapeutic agents used to treat diffuse large B-cell lymphoma (DLBCL) drug therapies include cyclophosphamide, doxorubicin, vincristine, prednisone, anti-CD20 monoclonal antibodies, etoposide, bleomycin, many of the agents listed for Waldenstrom's, and any combination thereof, such as ICE and R-ICE.
  • Non-limiting examples of therapeutic procedures used to treat Waldenstrom's Macroglobulinemia include peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, total body irradiation, infusion of stem cells, bone marrow ablation with stem cell support, in vitro-treated peripheral blood stem cell transplantation, umbilical cord blood transplantation, immunoenzyme technique, pharmacological study, low-LET cobalt-60 gamma ray therapy, bleomycin, conventional surgery, radiation therapy, and nonmyeloablative allogeneic hematopoietic stem cell transplantation.
  • Non-limiting examples of other therapeutic agents used to treat Chronic Lymphocytic Leukemia include Chlorambucil (Leukeran), Cyclophosphamide (Cyloxan, Endoxan, Endoxana, Cyclostin), Fludarabine (Fludara), Pentstatin (Nipent), Cladribine (Leustarin), Doxorubicin (Adriamycin®, Adriblastine), Vincristine (Oncovin), Prednisone, Prednisolone, Alemtuzumab (Campath, MabCampath), many of the agents listed for Waldenstrom's, and combination chemotherapy and chemoimmunotherapy, including the common combination regimen: CVP (cyclophosphamide, vincristine, prednisone); R-CVP (rituximab-CVP); ICE (iphosphamide, carboplatin, etoposide); R
  • the method comprises administering in addition to a compound of I or II to said patient, a therapeutically effective amount of at least one therapeutic agent and/or therapeutic procedure selected to treat said cancer or autoimmune disease in said patient.
  • the method comprises administering in addition to a compound of I or II to said patient, a therapeutically effective amount of a combination of therapeutic agents selected from the group consisting of a) CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone); b) R-CHOP (rituximab-CHOP); c) hyperCV AD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate, cytarabine); d) R-hyperCV AD (rituximab-hyperCV AD); e) FCM (fludarabine, cyclophosphamide, mitoxantrone); 1) R-FCM (
  • the compounds of the invention may be formulated for administration to animal subject using commonly understood formulation techniques well known in the art.
  • Formulations which are suitable for particular modes of administration and for the compounds of formula A may be found in Remington's Pharmaceutical Sciences , latest edition, Mack Publishing Company, Easton, Pa.
  • the compounds of the invention may be prepared in the form of prodrugs, i.e., protected forms which release the compounds of the invention 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 of the present invention 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.
  • the present invention also provides pharmaceutical compositions that comprise a compound of formula A and a biocompatible pharmaceutical carrier, adjuvant, or vehicle.
  • the composition can include the compound of Formula A 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.
  • the formulation can include stabilizing materials, such as polyols (e.g., sucrose) and/or surfactants (e.g., nonionic surfactants), and the like.
  • 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 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.
  • 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 compound of Formula A 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).
  • compositions comprising the compound of Formula A 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 dragée 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, potato, etc.
  • 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;
  • 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;
  • 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 (dosage) of active compound
  • 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.
  • Dragée 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 compound. Salts tend to be more soluble in aqueous or other protonic 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.
  • compositions of structural formula (A) that contain basic moieties can form pharmaceutically acceptable acid addition salts with suitable acids.
  • suitable acids for example, Berge, et al., describe pharmaceutically acceptable salts in detail in J Pharm Sci, 66:1 (1977).
  • the salts can be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting a free base function 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-tol
  • Basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates; long chain alkyl halides such as decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides; arylalkyl halides such as benzyl and phenethyl bromides; and others. Products having modified solubility or dispersibility are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • compositions comprising a compound of the invention formulated in a pharmaceutical acceptable carrier can be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • an article of manufacture such as a container comprising a dosage form of a compound of the invention and a label containing instructions for use of the compound.
  • Kits are also contemplated under the invention.
  • 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.
  • conditions indicated on the label can include treatment of inflammatory disorders, cancer, etc.
  • compositions comprising a compound of formula 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 compound of formula A is administered before, during, or after administration of chemotherapy, radiotherapy, and/or surgery.
  • the formulation and route of administration chosen 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 therapeutic index of the compound of formula A can be enhanced by modifying or derivatizing the compounds for targeted delivery to cancer cells expressing a marker that identifies the cells as such.
  • the compounds can be linked to an antibody that recognizes a marker that is selective or specific for cancer cells, so that the compounds are brought into the vicinity of the cells to exert their effects locally, as previously described (see for example, Pietersz, et al., Immunol Rev, 129:57 (1992); Trail, et al., Science, 261:212 (1993); and Rowlinson-Busza, et al., Curr Opin Oncol, 4:1142 (1992)).
  • Tumor-directed delivery of these compounds enhances the therapeutic benefit by, inter alia, minimizing potential nonspecific toxicities that can result from radiation treatment or chemotherapy.
  • the compound of formula A and radioisotopes or chemotherapeutic agents can be conjugated to the same anti-tumor antibody.
  • 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. Then, dosage can be formulated in animal models to achieve a desirable circulating concentration range that modulates expression or activity of a particular PI3K isoform or combination of isoforms. As human studies are conducted, further information will emerge regarding the appropriate dosage levels and duration of treatment for various diseases and conditions.
  • LFT liver function tests
  • levels such as alanine transaminase, aspartate transaminase, alkaline phosphatase, bilirubin, and gamma glutamyl transpeptidase, that are outside the normal range can signal possible liver toxicity.
  • Dosing of the therapeutic compound can be adjusted to avoid or reduce elevated liver function test values and subsequent potential for liver toxicity. For instance, a subject may be administered escalating doses of a compound.
  • the subject begins to develop elevated LFT levels outside a normal range, signaling potential liver toxicity at that dosage.
  • the dosage may be reduced to an amount such that LFT levels are reduced to an acceptable range as judged by the treating physician, e.g. a level that is in the range normal for the subject being treated, or within about 25% to 50% of normal. Therefore, liver function tests can be used to titrate the administration dosage of a compound.
  • 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 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 ED 50 with little or no toxicity.
  • Dosage may be limited by treatment-related toxicity symptoms. Such symptoms besides elevated liver function tests include anemia, vision blurring, diarrhea, vomiting, fatigue, mucositis, peripheral edema, pyrexia, peripheral neuropathy, pleural effusion, night sweats, and orthopnea, or a combination thereof. At a certain dose amount, if the subject develops intolerable levels of such symptoms, the dosage may be reduced such that the adverse event is eliminated and no longer adverse or reduced to an acceptable level as judged by a treating physician.
  • the concentration of compound in the blood is between 40-3,000 ng/mL over a 12 hour period from the time of administration. In another particular embodiment, the concentration of compound in the blood is between 75-2,000 ng/mL over a 12 hour period from the time of administration. In another particular embodiment, the concentration of compound in the blood is between 500-2,000 ng/mL over a 12 hour period from the time of administration.
  • the concentration of compound in the blood is between 40-3,000 ng/mL over a 12 hour period from the time of administration, wherein the compound is a formula of I, I′′, II, or II′′ and is orally administered in an amount of about 50 mg, 100 mg, or 200 mg.
  • the concentration of compound in the blood is between 40-3,000 ng/mL over a 12 hour period from the time of administration, wherein the compound is a formula of I and is orally administered in an amount of about 50 mg, 100 mg, or 200 mg.
  • the concentration of compound in the blood is between 40-3,000 ng/mL over a 12 hour period from the time of administration, wherein the compound is a formula of II and is orally administered in an amount of about 50 mg, 100 mg, or 200 mg.
  • the maximum concentration in the blood plasma is achieved within two hours of administration.
  • the dosage of the compound of Formula I or II is selected to produce a plasma concentration of drug of about 10 nM or higher over a period of 8 to 12 hours, on average, and to provide a peak plasma concentration of about 500 nM or higher, preferably about 1000 nM or higher. In certain embodiments, the dosage of the compound of Formula I or II is selected to produce a plasma concentration of drug of about 100 nM or higher over a period of 8 to 12 hours, on average, and to provide a peak plasma concentration of about 500 nM or higher, preferably about 1000 nM or higher.
  • the dosage of the compound of Formula I or II is selected to produce a plasma concentration of drug of about 200 nM or higher over a period of 8 to 12 hours, on average, and to provide a peak plasma concentration of about 500 nM or higher, preferably about 1000 nM or higher.
  • the dosage of the compound of formula I or II is selected to produce a plasma concentration wherein the trough concentration of the compound is in the range where a therapeutic effect, such as apoptosis of cancer cells, is observed. In certain embodiments, the dosage of the compound of formula I or II is selected to produce a trough plasma concentration at or higher than the EC 50 PI3K ⁇ isoform activation in blood plasma. In certain embodiments, the dosage of the compound of formula I or II is selected to produce an trough blood concentration above the EC 50 level for PI3K ⁇ activation and below the level for EC 50 PI3K ⁇ activation in a cell during a period of at least 12 hours from compound administration.
  • the dosage of the compound selected provides a trough plasma concentration of the compound between 60 nM and 1100 nM during a period of 8-12 hours from compound administration.
  • a dosage can be selected to produce an trough blood concentration above the EC 50 level for PI3K ⁇ basophil activation and below the EC 50 level for PI3K- ⁇ , - ⁇ or - ⁇ basophil activation.
  • the EC 50 values for the PI3K isoform activation or inhibition in vivo can be determined by a person having ordinary skill in the art.
  • the upper range of the trough concentration of the drug may exceed and is not limited by the EC 50 value of the PI3K- ⁇ , - ⁇ , or - ⁇ isoform in blood plasma.
  • the blood concentration range of the drug is at a level which is therapeutically beneficial in treating the hematologic malignancy, while minimizing undesirable side effects.
  • the compounds can exhibit sufficient activity on p110 ⁇ to be clinically useful, i.e., to be effective on a cancer that relies upon p110 ⁇ for signaling, because a plasma level above the effective dosage for inhibition of p110 ⁇ can be achieved while still being selective relative to other isoforms, particularly the alpha isoform.
  • the dosage of the compound is selected to produce a blood concentration effective for selectively inhibiting p110 ⁇ and p110 ⁇ .
  • the dosage of the compound provides a trough blood plasma concentration between 65 nM and 1100 nM during a period of 8 to 12 hours from compound administration. In some foregoing embodiments, the period is at least 12 hours from compound administration.
  • the compound is administered in a therapeutically effective amount.
  • the compound is administered at a dose of 20-500 mg/day. In a particular embodiment, the compound is administered at a dose of 50-250 mg/day.
  • the compound is administered at a dose of 25 to 150 mg per dose, and two doses are administered per day (e.g., BID dosing with 25 to 150 mg doses).
  • a subject is treated with 50 mg to 100 mg of a compound of formula A twice per day.
  • the method comprises administering to said patient an initial daily dose of 20-500 mg of the compound and increasing said dose by increments until clinical efficacy is achieved. Increments of about 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.
  • the method comprises continuing to treat said patient by administering the same dose of the compound at which clinical efficacy is achieved or reducing said dose by increments to a level at which efficacy can be maintained.
  • the method comprises administering to said patient an initial daily dose of 20-500 mg of the compound and increasing said dose to a total dosage of 50-400 mg per day over at least 6 days.
  • the dosage can be further increased to about 750 mg/day.
  • the compound is administered at least twice daily.
  • the compound is administered orally, intravenously or by inhalation.
  • the compound is administered orally.
  • it is administered orally at a dosage of about 50 mg BID or at a dosage of about 100 mg BID.
  • 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 PI3K ⁇ 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 or disorder in a subject.
  • Suitable dosage ranges for the compounds of formula A vary according to these considerations, but in general, the compounds are administered in the range of 10.0 ⁇ g/kg-15 mg/kg of body weight; 1.0 ⁇ g/kg-10 mg/kg of body weight, or 0.5 mg/kg-5 mg/kg of body weight.
  • the dosage range is from 700 ⁇ g-1050 mg; 70 ⁇ g-700 mg; or 35 mg-350 mg per dose, and two or more doses may be administered per day. Dosages may be higher when the compounds are administered orally or transdermally as compared to, for example, i.v. administration.
  • the reduced toxicity of a compound of formula A permits the therapeutic administration of relatively high doses.
  • oral administration of up to 750 mg/day of a compound of the invention is suitable.
  • a compound of formula A is administered at a dose of 50 mg BID.
  • a compound of formula A is administered at a dose of 100 mg BID.
  • a compound of formula A is administered at a dose of 200 mg BID.
  • a compound of formula A is administered at a dose of 350 mg BID.
  • a dosage of about 50-350 mg per dose, administered orally once or preferably twice per day, is often suitable for treatment of leukemias, lymphomas and multiple myeloma.
  • oral administration of up to 750 mg/day of compound I′′ or II′′ is suitable.
  • a compound of formula I′′ or II′′ is administered at a dose of 50 mg BID.
  • a compound of formula I′′ or II′′ is administered at a dose of 100 mg BID.
  • a compound of formula I′′ or II′′ is administered at a dose of 200 mg BID.
  • a compound of formula I′′ or II′′ is administered at a dose of 350 mg BID.
  • a dosage of about 50-350 mg per dose of a compound of formula I′′ or II′′, administered orally once or preferably twice per day, is often suitable.
  • the compounds may be administered as a single bolus dose, a dose over time, as in i.v. or transdermal administration, or in multiple dosages.
  • Dosing may be continued for at least seven days. In some embodiments, daily dosing is continued for about 28 days. In some embodiments, dosing is continued for about 28 days and is then discontinued for at least 7 days. In some embodiments, a complete cycle is continuous daily dosing for 28 days. Evaluation of a clinical response in the patient can be measured after each cycle. The clinical results can be used to make a decision to increase, decrease, discontinue or maintain the dosage.
  • 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 any infection. Additional factors that can be taken into account include time and frequency of administration, drug combinations, reaction sensitivities, and tolerance/response to therapy.
  • the frequency of dosing will depend on the pharmacokinetic parameters of the compound of Formula A 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 compound.
  • Short-acting pharmaceutical compositions i.e., short half-life
  • Long acting pharmaceutical compositions might be administered every 3 to 4 days, every week, or once every two weeks.
  • Pumps such as subcutaneous, intraperitoneal, or subdural pumps, can be preferred for continuous infusion.
  • Subjects that will respond favorably to the method of the invention include medical and veterinary subjects generally, including human patients. Among other subjects for whom the methods of the invention is useful are cats, dogs, large animals, avians such as chickens, and the like. In general, any subject who would benefit from a compound of formula A is appropriate for administration of the invention method.
  • the patient has a cytogenetic characteristic of del(17p) or del(11q).
  • the patient has a lymphadenopathy.
  • the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy in a patient.
  • the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy after one cycle of treatment. In some foregoing embodiments, the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 10% after one cycle of treatment. In some foregoing embodiments, the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 25% after one cycle of treatment. In some foregoing embodiments, the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 30% after one cycle of treatment.
  • the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 40% after one cycle of treatment. In some foregoing embodiments, the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 50% after one cycle of treatment. In some foregoing embodiments, the use of compound I, I′′, II, or II′′ reduces the size of a lymphadenopathy by at least 75% after one cycle of treatment.
  • the invention provides a method of treating a condition, comprising administering a compound of formula I, II or a pharmaceutically acceptable salt thereof and one or more therapeutic agents to a subject in need of such treatment, wherein the condition is a cancer or an autoimmune condition.
  • the therapeutic agent is a proteasome inhibitor.
  • the therapeutic agent is bortezomib.
  • the condition is a hematologic malignancy.
  • the condition is selected from the group consisting of multiple myeloma, acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, B-cell lymphoma, diffuse large B-cell lymphoma, B-cell ALL, T-cell ALL and Hodgkin's lymphoma.
  • the compound is substantially comprised of the S-enantiomer.
  • the compound comprises at least 95% of the S-enantiomer.
  • the administration of said compound and therapeutic agent provides a synergistic benefit superior to results obtained without the combination of the compound and therapeutic agent.
  • This example demonstrates the compound of formula I inhibits the cellular growth stimulatory effects of cytokines (IGF-1 and IL-6) in multiple myeloma (MM) cells.
  • LB cells Myelomonocytic myeloma cell line
  • the inhibitory effect of the compound of formula I on MM cell growth was assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrasodium bromide (MTT; Chemicon International) dye absorbance.
  • FIG. 1 Exposure of 0.625 ⁇ M-2.5 ⁇ M of Compound I inhibits MM cell growth even in the presence of cell growth stimulatory cytokines.
  • BMSCs Bone Marrow Stromal Cells
  • LB cells were cultured with control media, and with the compound of formula I for 48 hours, in the presence or absence of BMSCs.
  • Cell proliferation was assessed using [ 3 H]-thymidine uptake assay. All data represent mean ( ⁇ SD) of triplicate experiment. A summary of the results is shown in FIG. 2 .
  • LB cell growth is reduced after exposure to 0.625 ⁇ M-10 ⁇ M of compound I even in the presence of BMSC.
  • CLL chronic lymphocytic leukemia
  • Peripheral blood was obtained from patients with B-CLL through the CLL Research Consortium from Ohio State University.
  • Primary CD19-positive cells were isolated using Rosette-Sep (StemCell Technologies).
  • Cells were maintained in RPMI 1640 (Invitrogen) supplemented with 10% heat-inactivated fetal bovine serum, 2 mmol/L L-glutamine, and penicillin (100 units/mL)/streptomycin (100 ⁇ g/mL; Invitrogen) at 37° C., 5% CO 2 , and high humidity.
  • Compound I induced apoptosis was also seen to be effective in CLL cells from refractory/relapsed patients as shown in FIG. 20 .
  • This example demonstrates the compound of formula I results in a reduction of Akt phosphorylation and a decrease in cellular proliferation accompanied by cell death in both T-ALL and B-ALL (Acute Lymphoblastic Leukemia) leukemic cell lines.
  • Viability assays of cell lines were performed using the AlamarBlue assay (Invitrogen). Cells (1 ⁇ 10 6 per well) in a volume of 100 ⁇ L were placed in a 96-well flat-bottom plate and the compound of formula I (100 ⁇ L per well at 2 ⁇ final concentration) or medium alone was added to the plates. All were performed in quadruplicate. Cells were incubated for fixed times (48 hours). After the incubation, 10 ⁇ L AlamarBlue® was added to each well.
  • This example demonstrates treatment of the acute lymphoblastic leukemia (ALL) cell line CCRF-SB with the compound of formula I results in G0/G1 cell cycle arrest.
  • the average percentage of cells in G 0 -G 1 , S, and G 2 -M phases is calculated in the table below the histographs. Results are shown in FIG. 5 .
  • This example demonstrates the compound of formula I inhibits proliferation of breast cancer cell lines.
  • T47D and HS-578T cell lines were grown in the presence of serum plus the indicated concentrations of the compound of formula I.
  • Proliferation was measured in triplicate wells by AlamarBlue® assay (Invitrogen) 96-well plates. Results of proliferation assays are expressed as the mean cellular percentage values and shown in FIG. 6 .
  • This example demonstrates the compound of formula I inhibits proliferation of ovarian cancer cell lines.
  • IGROV-1 and OVCAR-3 cell lines were grown in the presence of serum plus the indicated concentrations of the compound of formula I. Proliferation was measured in triplicate wells by AlamarBlue assay (Invitrogen) 96-well plates. Results of proliferation assays are expressed as the mean cellular percentage values and are shown in FIG. 7 .
  • This example demonstrates the compound of formula I reduces constitutive Akt phosphorylation in hematopoietic tumor cell lines that exhibited constitutive Akt phosphorylation.
  • a large panel of leukemia and lymphoma cell lines was assessed for constitutive Akt phosphorylation. These cell lines represent B-lymphoma, T-lymphoma, ALL, Malignant histiocytosis, DLBCL and AML.
  • Cell lines that demonstrated serum independent Akt phosphorylation were treated with the compound of formula I for 2 hours. Thereafter, cell were lysed, size-fractioned and immunoblotted with antibodies directed against phospho-Akt(Ser473). Results are shown in FIG. 8 . Reduction in Akt(Ser473) was achieved for all cell lines after exposure to compound I.
  • This example demonstrates the compound of formula I induces apoptosis in breast cancer cell lines.
  • HS-578T, T47D, and MCF7 cells were treated with the compound of formula I or corresponding DMSO concentrations for 24 h.
  • the percentage of apoptotic cells was determined by Annexin V-FITC/7AAD staining. Bottom left, viable cells (Annexin V-FITC/PI negative); bottom right, early apoptotic cells (Annexin V-FITC positive only); top right, mid-late apoptotic cells (Annexin V-FITC/7AAD double-positive); and top left, late apoptotic/necrotic (7AAD positive only). Percentages of cells in each quadrant are indicated except for the bottom left quadrant (viable cells).
  • FIG. 10 One experiment representative of three different experiments that gave similar results is shown in FIG. 10 .
  • This example provides data relating to the concentration of the compound of formula I in the blood of a healthy human subject on day 7.
  • the concentration was monitored over a period of 12 hours, after oral administration of 50, 100, or 200 mg BID of the compound of formula I on day 7 of the study.
  • FIG. 11 follows the plasma concentration of the drug over a period of 12 hours from administration. The maximum concentration of drug is achieved within two hours for all doses.
  • Administration of 50, 100 or 200 mg BID of said compound results in a concentration level that exceeds the PI3K ⁇ EC 50 concentration in basophil for at least 12 hours.
  • the mean trough concentration was higher than the EC50 for PI3K ⁇ and the mean peak concentration was lower than the EC50 for PI3K ⁇ as determined in the whole blood basophil activation assay, FIG. 24B .
  • This example demonstrates the concentration range of compound I administered at 50 mg BID is at a level that is above the ED 50 PI3K ⁇ basophil activation level but lower than the minimum ED 50 PI3K ⁇ basophil level activation level in whole blood for at least 12 hours.
  • Table 1 below, provides an overview of the subjects in the study, wherein either a singe dose (SD) or multiple dose (MD) of the compound of formula I is administered to a subject at varying amounts.
  • SD singe dose
  • MD multiple dose
  • n refer to the number of subjects in each group.
  • This example provides data relating to the area of lesions of a patient with mantle cell lymphoma after 1 cycle of treatment (28 days) with the compound of formula I.
  • the area of 6 lesions was measured prior to treatment and after a cycle of treatment.
  • the response to 28 days of oral administration of 50 mg BID of the compound of formula I results in a decrease of lesion area compared to area prior to treatment and represents a 44% decrease in tumor burden.
  • the results are summarized in a bar graph found in FIG. 12 .
  • This example provides data relating to the concentration of absolute lymphocyte count (ALC) in the blood of a patient with CLL after 1 cycle (28 days) of treatment with oral administration of the compound of formula I.
  • ALC absolute lymphocyte count
  • the blood ALC concentration was measured over a period of 4 weeks after completion of one cycle of treatment.
  • a 55% decrease in lymphocytosis and a 38% decrease in lymphadenopathy as a result of treatment were observed.
  • a marked decrease in ALC concentration is observed between week 1 and week 2, FIG. 13 .
  • This example provides data comparing the concentration of the compound of formula I in a lymphoma patient to normal healthy volunteers.
  • the concentration of the compound in the blood was measured over a period of 6 hours after administration.
  • the concentration of 50 and 100 mg oral administration in normal healthy volunteers on day 7 of administration was also observed.
  • the results are summarized in FIG. 14 .
  • the compound does not build up excessively over the course of a cycle of treatment, nor does the patient become tolerant by increased metabolism over the course of the treatment cycle.
  • This example shows the IC 50 profile of compound I across classes of kinases as summarized in Table 2. While especially active on p110 ⁇ , Compound I was also active on p110 ⁇ and even active enough to be therapeutically useful at non-toxic doses against p110 ⁇ , due to the demonstrated high NOAEL level of the compound; while exhibiting little activity on Class II-V phosphoinositide kinases. Thus while being delta-selective, the compounds can exhibit sufficient activity on p110 ⁇ to be clinically useful, i.e., to be effective on a cancer that relies upon p110 ⁇ for signaling, because a plasma level above the effective dosage for inhibition of p110 ⁇ can be achieved while still being selective relative to other isoforms, particularly the alpha isoform.
  • Class I PI3Ks Class II PI3K, Class III PI3K, Class IV PI3K, Phosphoinositide IC 50 (nM) IC 50 (nM) IC 50 (nM) IC 50 (nM) kinases
  • Compound P110 ⁇ p110 ⁇ p110 ⁇ p110 ⁇ CIIbeta hVPS34 DNA-PK mTOR PIP5K ⁇ PIP5K ⁇ I 435 128 1 14 >10 3 978 6,729 >10 3 >10 3 >10 3 NVP-BEZ-235 19 293 63 267 3 6 1 2 ND* ND Novartis InvitroGen Adapta assay *ND not determined
  • Swiss-3T3 fibroblasts and RAW-264 were seeded on a 96-well tissue culture plate and allowed to reach at least 90% confluency.
  • Cells were starved and treated with either vehicle or serial dilutions of compound I for 2 hrs and stimulated with PDGF or C5a respectively.
  • Akt phosphorylation and total AKT was detected by ELISA.
  • Purified B-cells were treated with either vehicle or serial dilutions of compound I for 30 minutes at room temperature before the addition of purified goat anti-human IgM. Results are expressed as relative [ 3 H] thymidine incorporation induced by IgM crosslinking.
  • PI3K p110 ⁇ promotes proliferation and survival in a wide range of leukemia and lymphoma cell lines.
  • the cell types investigated are MCL, DLBCL, AML, ALL, and CML.
  • FIG. 15 Proteins from 10 6 cells were separated by SDS-PAGE and analyzed by Western blot using antibodies specific for the ⁇ , ⁇ , ⁇ and ⁇ isoforms. Purified recombinant p110 proteins were used as controls. Anti-actin antibodies were used to assess equal loading of the samples. p110 ⁇ was consistently expressed at a high level while other p110 isoforms were highly variable. PI3K p110 ⁇ is known to be uniformly expressed in patient AML cells as discussed by Sujobert, et al., Blood 2005 106(3), 1063-1066.
  • Example 19 shows compound I inhibition of p110 ⁇ blocks PI3K signaling in leukemia and lymphoma cell lines with constitutive pathway activation.
  • PI3K pathway is frequently deregulated in leukemia and lymphoma cell lines. 48% of cell lines, or 13 out of 27, were found to have constitutive p-AKT. In addition, PI3K pathway activation is dependent on p110 ⁇ . Compound I was found to inhibit constitutive AKT phosphorylation in 13 out of 13 cell lines.
  • FIG. 9 PAGE results of FIG. 9 demonstrates that constitutive AKT phosphorylation was inhibited by the presence of compound I in each of 11 cell lines, including B-cell and T-cell lymphomas.
  • Cells were incubated for 2 hrs with 10 ⁇ M compound I.
  • Cell lysates were run on SDS-PAGE and transferred onto PDVF membrane and probed with appropriate antibodies.
  • Compound I was found to inhibit constitutive AKT phosphorylation in 11 out of 11 cell lines.
  • Additional cell line data for T-ALL and B-ALL cell lines is shown in FIG. 27 .
  • Compound I Inhibits Proliferation and Apoptosis in Leukemia Cell Lines
  • Example 20 demonstrates that compound I inhibits proliferation and induces apoptosis in leukemia cell lines.
  • FIGS. 16A-B show that treatment with compound I for 24 hours reduces cellular viability in a dose dependent manner.
  • Proliferation assays (AlamarBlue®) on ALL cell lines grown in the presence of 10% FBS serum and measurements were taken at 24 hrs. Proliferation was measured in triplicate wells in 96-well plates. The inhibition of PI3K signaling by compound I resulted in a block of cell cycle progression, and/or cell death. In each of six leukemia cell lines, viability was reduced by 40-50% with 10 micromolar concentrations of Compound I, FIG. 16A .
  • This example demonstrates PI3K p110 ⁇ and p110 ⁇ isoform expression in patient CLL cells.
  • PI3K mediated signaling pathways have been implicated in CLL. These pathways have a role in cell proliferation, prevention of apoptosis and cell migration. Efforts were made to determine PI3K isoform expression in patient CLL cells.
  • the PAGE images of FIG. 17A-D compare the expression of p110 ⁇ , p110 ⁇ , p110 ⁇ , and p110 ⁇ in CLL cells of patients A-E.
  • p110 ⁇ and p110 ⁇ is expressed in each patient compared to the other PI3K isoforms.
  • FIGS. 18A-B show results of caspase 3 and PARP (Poly(ADP) Ribose Polymerase) cleavage in the presence of 1, 10 ⁇ M of compound I or 25 ⁇ M of LY294002.
  • FIG. 21 shows the results of Phospho-Akt production in the absence or presence of 0.1, 1.0, 10 ⁇ M of Compound I. This provides evidence that compound I reduces phospho-Akt production in patient AML cells.
  • This example demonstrates a whole-blood assay for measurement of PI3K signaling in basophils using flow cytometry by the induction of CD63 surface expression.
  • PI3K signaling is monitored by CD63 surface expression.
  • p110 ⁇ mediates FC ⁇ R1 signaling
  • p110 ⁇ mediates fMLP receptor signaling.
  • the flow cytometry analysis of PI3K mediated CD63 expression on basophils comprises the following sequential steps:
  • Basophil stimulation fMLP or Anti-FC ⁇ R1 Mab
  • FIG. 22A-C compares the results of A) no stimulation, B) stimulation with Anti-FC ⁇ R1, or C) stimulation with fMLP.
  • FIG. 23 shows that Compound I is especially active where p110 ⁇ mediated signaling is most important, but is also relatively active where p110 ⁇ is utilized: it achieved 50% reduction in SD63 expression at ⁇ 1 ⁇ M for the p110 ⁇ test, and ca. 10 ⁇ M for the p110 ⁇ test.
  • Basophil activation was measured in human whole blood using the Flow2 CAST® kit. Whole blood samples were treated with either vehicle or serial dilutions of compound I prior to activation of basophils either with anti-Fc ⁇ RI mAb or fMLP. Cells were stained with the combination of anti-human CD63-FITC and anti-human CCR3-PE mAbs. The percent CD63 positive cells within the gated basophil population were determined in different treatment groups and normalized to the vehicle control.
  • This example provides evidence of the reduction in size of a bulky lymphadenopathy in a CLL patient with a del[17p].
  • a patient with del(17p) had an axillary lymphadenopathy, which was imaged by computed tomography (CT) to provide a baseline measurement of 5.9 cm ⁇ 4.1 cm, FIG. 40A .
  • CT computed tomography
  • the lymphadenopathy was reduced to a dimension of 3.8 ⁇ 1.8 cm, FIG. 40B .
  • a cycle treatment was 28 days of continuous oral dosing at either 200 mg BID or 350 mg BID of compound I.
  • This example demonstrates that treatment with compound I has little or no effect on glucose and insulin levels.
  • Compound I was administered at 50-200 mg amounts BID to a subject over a period of up to 10 days. Blood glucose and insulin concentrations were measured over time and compared to placebo results as shown in FIGS. 25A-B .
  • p110 ⁇ inhibitor compound I and compound II were provided by Calistoga Pharmaceuticals, (Seattle, Wash.). The sample of compound I and II used was over 95% the S enantiomer.
  • Compound I was dissolved in Dimethyl sulphoxide at 10 mM and stored at ⁇ 20° C. for in vitro study.
  • Compound II was dissolved in 1% carboxyl methylcellulose (CMC)/0.5% Tween 80 and stored at 4° C. for in vivo study.
  • Recombinant human P110 ⁇ , ⁇ , ⁇ , and ⁇ were reconstituted with sterile phosphate-buffered saline (PBS) containing 0.1% BSA.
  • bortezomib was provided by Millennium Pharmaceuticals (Cambridge, Mass.). 3-Methyladenine was purchased from Sigma-Aldrich (St. Louis, Mo.).
  • MM.1S and resistant human MM cell lines were kindly provided by Dr. Steven Rosen (Northwestern University, Chicago, Ill.). H929, RPMI8226, and U266 human MM cell lines were obtained from American Type Culture Collection (Manassas, Va.). Melphalan-resistant RPMI-LR5 and Doxorubicin (Dox)-resistant RPMI-Dox40 cell lines were kindly provided by Dr. William Dalton (Lee Moffitt Cancer Center, Tampa, Fla.). OPM1 plasma cell leukemia cells were provided by Dr. Edward Thompson (University of Texas Medical Branch, Galveston). IL-6-dependent human MM cell line INA-6 was provided by Dr.
  • LB human MM cell line was established in the laboratory. Phenotypic analysis revealed no cytogenetic abnormalities. Phenotypic analysis is shown in table 6. CD expression profile of LB cell line, defined by flow-cytometric analysis.
  • MM cell lines were cultured in RPMI1640 medium.
  • Bone marrow stromal cells were cultured in Dulbecco's modification of Eagle's medium (DMEM) (Sigma) containing 15% fetal bovine serum, 2 mM L-glutamine (Life Technologies), 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin (Life Technologies).
  • DMEM Dulbecco's modification of Eagle's medium
  • PBMNCs peripheral blood mononuclear cells
  • BM mononuclear cells were separated using Ficoll-PaqueTM density sedimentation, and plasma cells were purified (>95% CD138+) by positive selection with anti-CD138 magnetic activated cell separation micro beads (Miltenyi Biotec, Auburn, Calif.). Tumor cells were also purified from the BM of MM patients using the RosetteSep negative selection system (StemCell Technologies, Vancouver, BC, Canada).
  • the growth inhibitory effect of compound I on growth of MM cell lines, PBMCs, and BMSCs was assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetra-sodium bromide (MTT; Chemicon International, Temecula, Calif.) dye absorbance.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetra-sodium bromide
  • MM cells (2 ⁇ 104 cells/well) were cultured for 48 h in BMSC coated 96-well plates (Costar, Cambridge, Mass.), in the presence or absence of drug. DNA synthesis was measured by [3H]-thymidine (Perkin-Elmer, Boston, Mass.) uptake, with [3H]-thymidine (0.5 ⁇ Ci/well) added during the last 8 h of 48 h cultures. All experiments were performed in quadruplicate.
  • INA-6 cells and LB cells were transiently transfected with siRNA ON-TARGET plus SMART pool P110 ⁇ or nonspecific control duplex (Dharmacon Lafayette, Co) using Cell Line Nucleofector Kit V (Amaxa BIosystems Gaitherburg, Md.).
  • Viable MM cells (2.5 ⁇ 104) were pelleted on glass slides by centrifugation at 500 rpm for 5 minutes using a cytospin system (Thermo Shandon, Pittsburgh, Pa.). Cells were fixed in cold absolute acetone and methanol for 10 min. Following fixation, cells were washed in phosphate-buffered saline (PBS) and then blocked for 60 min with 5% FBS in PBS. Slides were then incubated with anti-CD138 antibody (Santa Cruz Biotechnology, Santa Cruz, Calif.) at 4° C. for 24 h, washed in PBS, incubated with goat anti-mouse IgG for 1 h at 4° C., and analyzed using Nikon E800 fluorescence microscopy.
  • PBS phosphate-buffered saline
  • vital staining was performed for 15 min with acridine orange at a final concentration of 1 ⁇ g/ml. Samples were examined under a fluorescence microscope.
  • HUVEC and endothelial growth media were obtained from Lonza (Walkersville, Md., USA). HUVEC were cultured with compound I on polymerized matrix gel at 37° C. After 8 h, tube formation was evaluated using Leika DM IL microscopy (Leica Microsystems, Wetzlar, Germany) and analyzed with IM50 software (Leica Microsystems Imaging Solutions, Cambridge, UK). HUVEC cell migration and rearrangement was visualized, and the number of branching points counted.
  • MM cells were cultured with or without compound I; harvested; washed; and lysed using radioimmuno precipitation assay (RIPA) buffer, 2 mM Na 3 VO 4 , 5 m M NaF, 1 mM phenylmethylsulfonyl fluoride (5 mg/ml).
  • RIPA radioimmuno precipitation assay
  • Cytokine secretion by human BMSCs cocultured with MM cells was assessed by ELISA.
  • BMSCs were cultured in 96-well plates with varying concentrations of compound I, with or without INA-6 cells. After 48 h, supernatants were harvested and stored at ⁇ 80° C. Cytokines were measured using Duo set ELISA Development Kits (R&D Systems, Minneapolis, Minn.). All measurements were carried out in triplicate.
  • cytokine levels in culture supernatants were assessed using Proteome Profiler Antibody Arrays Panel A (R&D Systems, Minneapolis, Minn.), Supernatants from co-cultures with BMSCs were incubated for 4 hours with membranes arrayed with Abs against 37 cytokines, according to manufacturer's instructions.
  • mice 48-54 days old were purchased from Charles River Laboratories (Wilmington, Mass.). All animal studies were conducted according to protocols approved by the Animal Ethics Committee of the Dana-Farber Cancer Institute. Mice were inoculated subcutaneously in the right flank with 3 ⁇ 106 LB cells in 100 ⁇ L RPMI-1640. When tumors were palpable, mice were assigned into the treatment groups receiving 10 mg/kg or 30 mg/kg gavages twice daily; and 7 mice in the control group receiving vehicle alone. Caliper measurements of the longest perpendicular tumor diameters were performed every alternate day to estimate the tumor volume using the following formula representing the 3D volume of an ellipse: 4/3 ⁇ (width/2)2 ⁇ (length/2).
  • FIGS. 30A and 30B This example demonstrates that p110 ⁇ is highly expressed in patient MM cells.
  • Abs was used against recombinant human PI3K/p110 ⁇ , ⁇ , ⁇ , and ⁇ proteins with specific immunoreactivity against these isoforms.
  • the expression of p110 ⁇ in 11 MM cell lines (MM.1S, OPM1, OPM2, RPMI8226, DOX40, LR5, MM.1R, U266, INA-6, H929, and LB), as well as 24 patient MM samples were evaluated and immunoblots shown in FIGS. 30A and 30B .
  • FIG. 30A shows expression of p110- ⁇ , - ⁇ , - ⁇ , and - ⁇ in MM cell lines detected by immunoblotting using specific antibodies.
  • Anti- ⁇ -Tubulin MAb served as a loading control.
  • p110 ⁇ in patient MM cells was detected by immunoblotting using anti-P110 ⁇ Ab ( FIG. 30B ).
  • Anti-GAPDH MAb served as a loading control.
  • INA-6 and LB cells strongly expressed p110 ⁇ , whereas MM.1S, OPM1, MM.1R, Dox40, U266 or H929 lacked p110 ⁇ expression ( FIG. 30A ).
  • FIG. 30C Human recombinant P110- ⁇ , - ⁇ , - ⁇ , - ⁇ proteins in SDS sample buffer were heated for 3 min prior to loading on gel. (10-20 ⁇ g per lane.) Recombinant human P110- ⁇ , - ⁇ , - ⁇ , - ⁇ proteins were detected by Immunoblot analysis. Levels of P110 ⁇ were measured in MM1S and LB cells using P110 ⁇ specific FITC conjugated secondary antibodies. P110 ⁇ stained green, and nucleic acids (DAPI) stained blue.
  • DAPI nucleic acids
  • compound I has selective cytotoxicity against cells with p110 ⁇ . Specifically, compound I potently induced cytotoxicity in p110 ⁇ positive MM cells as well as in primary patient MM cells without cytotoxicity in peripheral blood mononuclear cells from healthy donors, suggesting a favorable therapeutic index.
  • the growth inhibitory effect of p110 ⁇ knockdown in MM cells was evaluated.
  • LB and INA-6 cells were transfected with P110 ⁇ siRNA (Si) or control siRNA (Mock). After 24 h, expression of P110 ⁇ was determined by western blot analysis, see FIG. 31A .
  • INA-6 cells were transfected with p110 ⁇ siRNA or control siRNA, and then cultured for 72 hours. Cell growth was assessed by MTT assay, see FIG. 31B . Data indicates mean ⁇ SD of triplicate cultures, expressed as fold of control. Transfection with p110 ⁇ siRNA, but not mock siRNA, down-regulated p110 ⁇ and inhibited MM cell growth at 72 h ( FIGS. 31A and 31B ).
  • the growth inhibitory effect of p110 ⁇ specific small molecule inhibitor compound I in MM cell lines, PBMCs, and patient MM cells was evaluated.
  • FIG. 31C Compound I induced cytotoxicity against LB and INA-6 MM cells (p110 ⁇ -positive) in a dose- and time-dependent fashion; in contrast, minimal cytotoxicity was noted in p110 ⁇ -negative cell lines ( FIG. 31C ).
  • the legend for FIG. 31C LB ( ⁇ ), INA-6 ( ⁇ ), RPMI 8226 ( ⁇ ), OPM2 ( ⁇ ), H929 ( ⁇ ), U266 ( ⁇ ), RPMI-LR5 ( ⁇ ) and OPM1 ( ⁇ ) MM cells were cultured with or without compound I for 48 h.
  • compound I also induced cytotoxicity against patient MM cells ( FIG. 31D ), without cytotoxicity in PBMCs from 4 healthy volunteers at concentrations up to 20 ⁇ M ( FIG. 31E ).
  • Patients MM cells isolated from BM by negative selection were cultured with compound I for 48 h.
  • Peripheral blood mononuclear cells isolated from healthy donors were cultured with compound I for 72 h.
  • Data represent mean ⁇ SD viability, assessed by MTT assay of triplicate cultures, expressed as percentage of untreated controls.
  • INA-6 cells were cultured with compound I (0-5 ⁇ M) for 120 h. Total cell lysates were subjected to immunoblotting using anti-caspase-3, -8, -9, PARP, and ⁇ -tubulin Abs. FL indicates full-length protein, and CL indicates cleaved protein. Significantly increased cleavage of caspase-8, caspase-9, caspase-3, and PARP was observed in INA-6 MM cells treated with compound I for 120 h ( FIG. 31F ). These results indicate that cytotoxicity triggered by compound I is mediated, at least in part, via caspase-dependent (both intrinsic and extrinsic) apoptosis.
  • PI3K serine/threonine protein kinase AKT, which is activated by phosphorylation of Thr308 in the activation loop of the kinase domain and Ser473 in the C-terminal tail. Phosphorylation of both sites requires an interaction between the N-terminal pleckstrin homology domain of AKT and membrane phosphoinositide generated by PI3K. It was shown that compound I inhibits both domains, suggesting that P110 ⁇ is the predominant isoform responsible for PI3K signaling in MM cell lines.
  • INA-6 cells were cultured with Compound I or LY294002 for 12 h, FIG. 32A .
  • Actin Ab was used as a loading control.
  • INA-6 and MM.1S cells were cultured with Compound I (0, 0.25, 1.0, 5.0 ⁇ M) for 6 hours, FIG. 32B .
  • LB and INA-6 cells were cultured with compound I for 0-6 hours, FIG. 32C .
  • Whole cell lysates were subjected to immunoblotting using AKT, P-AKT (Ser473 and Thr308), ERK1/2, P-ERK1/2, P-PDK1, and P-FKRHL antibodies.
  • ⁇ -tubulin is used as a loading control.
  • Compound I significantly blocked phosphorylation of AKT and ERK1/2 in p110 ⁇ positive INA-6 cells ( FIG. 32A ), but did not affect phosphorylation of AKT or ERK in MM.1S cells with low expression of P110 ⁇ ( FIG. 32B ).
  • Compound I also significantly inhibited phosphorylation of upstream PDK-1 and downstream FKHRL in INA-6 and LB MM cells in a time- and dose-dependent fashion ( FIG. 32C ), further confirming inhibition of a both PI3K/AKT and ERK pathways in these cells.
  • AKT regulates autophagy, thus investigation of compound I in inducing autophagy in LB and INA-6 MM cells was carried out.
  • INA-6 and LB MM cells were treated with 5 ⁇ M Compound I for 6 h.
  • Compound I treatment induced LC3 accumulation in LB and INA-6 cells, evidenced by fluorescence microscopy or transmission electron microscopy. Autophagosome formation was defined by the accumulation of LC3; arrows indicate autophagosomes, FIG. 33A .
  • INA-6 cells were treated with 5 ⁇ M Compound I or serum starvation for 6 h, stained with 1 ⁇ g/mL acridine orange for 15 min, and analyzed by fluorescence microscopy, FIG. 33B .
  • LC3 and beclin-1 protein levels were determined by western blotting using LC3 and beclin-1 antibodies of lysates from INA-6 cells treated with Compound I, with or without 3-MA, FIG. 33C .
  • GAPDH served as a loading control.
  • Immunofluorescence analysis showed markedly increased LC 3 staining in INA-6 and LB cells triggered by compound I (5 ⁇ M, 6 h) treatment ( FIG. 33A ). Electron microscopic analysis also showed increased autophagic vacuoles (arrows) in MM cells treated with compound I. Since autophagy is characterized as acidic vesicular organelle (AVO) development, acridine orange staining was carried out. As shown in FIG. 33B , vital staining with acridine orange revealed development of AVOs in compound I-treated LB and INA-6 cells. Moreover, markedly increased LC3-II and Beclin 1 protein were detected in INA-6 MM cells after 6 h treatment with compound I, which was blocked by 3-MA autophagic inhibitor ( FIG. 33C ).
  • AVO acidic vesicular organelle
  • FIG. 33D P110 ⁇ positive LB cells ( ⁇ ) were treated with 3-MA (0-100 ⁇ M) for 24 h. Data represent means ( ⁇ SD) of triplicate cultures.
  • LC3-II a hallmark of autophagy, is induced by compound I treatment in p110 ⁇ positive MM cell lines.
  • compound I treatment resulted in a marked increase in autophagy, evidenced by the presence of autophagic vacuoles in the cytoplasm, formation of AVOs, membrane association of microtubule-associated protein I of LC3 with autophagosomes, and a marked induction of LC3-II protein.
  • Electron microscopic analysis confirmed that compound I induced autophagosomes.
  • LC3-II was expressed through LC3-I conversion.
  • autophagy induced by compound I was suppressed by 3-MA, a specific inhibitor of autophagy.
  • Compound I Inhibits Cell Growth in the Presence of BMSC
  • This example demonstrates the ability of compound Ito inhibit paracrine MM cell growth with BMSCs.
  • IL-6 and IGF-1 induces growth and anti-apoptosis in MM cells
  • compound I was examined in overcoming the effects of these cytokines in INA-6 and LB MM cells.
  • LB and INA-6 cells were cultured for 48 h with control media ( ⁇ ); or with compound I at 5.0 ⁇ M ( ⁇ ) or 10 ⁇ M ( ⁇ ), in the presence or absence of IL-6 (1 and 10 ng/ml), FIG. 34A , or IGF-1 (10 and 100 ng/mL), FIG. 34B .
  • DNA synthesis was determined by measuring [3H]-thymidine incorporation during the last 8 h of 72 h cultures. Data represent means ( ⁇ SD) of triplicate cultures. Neither IL-6 nor IGF-1 protected against the growth inhibition induced by compound I ( FIGS. 34A and 34B ).
  • the BM microenvironment confers proliferation and drug-resistance in MM, thus MM cell growth inhibitory effect of compound I in the presence of BMSCs was examined.
  • LB and INA-6 MM cells were cultured for 48 h with control media ( ⁇ ), and with 2.5 ⁇ M ( ), 5 ⁇ M ( ⁇ ), and 10 ⁇ M ( ⁇ ) of Compound I, in the presence or absence of BMSCs, FIG. 34C .
  • DNA synthesis was determined by [3H]-thymidine incorporation. Data represent means ( ⁇ SD) of triplicate cultures.
  • IL-6 in culture supernatants from BMSCs treated with compound I (0-2.5 ⁇ M) was measured by ELISA, FIG. 34D . Error bars indicate SD ( ⁇ ).
  • BMSCs were cultured with 1.0 ⁇ M compound I or control media for 48 h; cytokines in culture supernatants were detected using cytokine arrays, FIG. 34E .
  • INA-6 cells cultured with or without BMSCs were treated with compound for 48 h.
  • Total cell lysates were subjected to immunoblotting using indicated antibodies, FIG. 34F .
  • Actin was used as a loading control.
  • BMSCs from 2 different patients were cultured with compound I (0-20 ⁇ M) for 48 h.
  • Cell viability was assessed by MTT assay, FIG. 34G . Values represent mean ⁇ SD of triplicate cultures.
  • This example demonstrates the ability of Compound Ito inhibit HuVEC tubule formation.
  • PI3K specifically p110 isoform
  • Endothelial cells are an essential regulator of angiogenesis for tumor growth.
  • Both Akt and ERK pathways are associated with endothelial cell growth and regulation of angiogenesis; and importantly, endothelial cells express p110 ⁇ .
  • This example also demonstrates that compound I blocks in vitro capillary-like tube formation, associated with down regulation of Akt phosphorylation.
  • HuVECs were treated with 0, 1.0, or 10 ⁇ M of compound I for 8 h, and tube formation by endothelial cells was evaluated ( FIG. 35A ).
  • HuVEC cells were plated on Matrigel-coated surfaces and allowed to form tubules for 8 h, in the presence or absence of Compound I. Endothelial cell tube formation was measured by microscopic analysis, FIG. 35B . *P ⁇ 0.005.
  • HuVECs were cultured with Compound I (0-20 ⁇ M) 48 h, and viability was assessed by MIT assay, FIG. 35C . Data shown are mean ⁇ SE of triplicate wells from a representative experiment. Thus, compound I inhibited capillary-like tube formation in a dose-dependent fashion (p ⁇ 0.05) ( FIG. 35B ), without associated cytotoxicity ( FIG. 35C ).
  • HuVECs were cultured with compound I (0-200 ⁇ M) for 8 h, and cell lysates were analyzed by immunoblotting using the indicated antibodies, FIG. 35D . Actin was used as a loading control.
  • This example demonstrates the ability of compound II to inhibit human MM cell growth in vivo.
  • the in vivo efficacy of P110 ⁇ inhibitor was evaluated in a xenograft model in which SCID mice are injected subcutaneously with human MM cells.
  • mice injected with 5 ⁇ 10 6 LB cells were treated orally twice a day with control vehicle ( ⁇ ), and compound II 10 mg/kg ( ⁇ ) or 30 mg/kg ( ⁇ ).
  • Mean tumor volume was calculated as in Materials and Methods, FIG. 36A . Error bars represent SD ( ⁇ ).
  • FIG. 36B Representative whole-body images from a mouse treated for 12 d with control vehicle (top panel) or Compound II (30 mg/kg) (bottom panel), FIG. 36B .
  • CD31 and P-AKT positive cells are dark brown, FIG. 36D .
  • mice were treated with Compound II 10 mg/kg (- -), 30 mg/kg ( . . . ) or Control vehicle ( ⁇ ). Survival was evaluated from the first day of treatment until sacrifice using Kaplan-Meier curves, FIG. 36C .
  • Tumor tissues were harvested from mice treated with control vehicle or Compound II (30 mg/kg). Protein levels of phosphorylated of PDK-1 and AKT (Ser473) were determined by western blotting of cell lysates, FIG. 36E . Actin was used as a loading control.
  • INA-6 cells engrafted in human bone chips in SCID mice was monitored by serial serum measurements of shuIL-6R. Mice were treated with Compound II 10 mg/kg ( ⁇ ), 30 mg/kg ( ⁇ ) or control vehicle ( ⁇ ), and shuIL-6R levels were determined weekly by ELISA, FIG. 36F . Error bars indicate SD ( ⁇ ).
  • OS Overall Survival
  • FIG. 36D immunohistochemical
  • FIG. 36E immunoblot analysis confirmed that compound II treatment (30 mg/kg) significantly inhibited p-Akt and p-PDK-1, as well as significantly decreased CD31 positive cells and microvessel density (p ⁇ 0.01) ( FIG. 36D ). This suggests that compound II can inhibit angiogenesis in vivo via suppression of the Akt pathway.
  • This example demonstrates the effect of Compound I in combination with bortezomib to mediate synergistic MM cytotoxicity.
  • LB and INA-6 MM cells were cultured with medium ( ⁇ ) and with compound I, 1.25 ⁇ M ( ⁇ ), 2.5 ⁇ M ( ) or 5.0 ⁇ M ( ⁇ ), in the presence or absence of bortezomib (0-5 nM). Cytotoxicity was assessed by MTT assay; data represent the mean ⁇ SD of quadruplicate-cultures, FIG. 37A .
  • INA-6 cells were treated with Compound I (5 ⁇ M) and/or bortezomib (5 nM) for 6 h.
  • Phosphorylation of AKT was determined by western blotting of cell lysates using phospho-AKT (ser473) antibody, FIG. 37B .
  • Actin served as a loading control.
  • Compound I enhances cytotoxicity of bortezomib.
  • Increasing concentrations of compound I (1.5-5.0 ⁇ M) added to bortezomib (2.5, 5.0 nM) triggered synergistic cytotoxicity in LB and INA-6 MM cells ( FIG. 37A and Table 7).
  • induction of phospho-Akt by bortezomib treatment was inhibited in the presence of compound I ( FIG. 37B ).
  • FIG. 38A This example provides evidence that compound I blocks PI3K signaling and induces apoptosis in follicular lymphoma cells.
  • P110 ⁇ is expressed in FL cell lines as shown in FIG. 38A . Certain cell lines show reduction in the production of pAkt, Akt, pS6 and S6 when the cell is exposed to compound I, FIG. 38B . Cleavage of PARP and Caspase-3 is observed after exposure to compound I in a dose dependent fashion after 24 hours at 0.1 ⁇ M and 0.5 ⁇ M, FIG. 38C .
  • FIG. 39A shows a significant reduction of pAkt in MCL lines exposed to different survival factors in the presence of compound I.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Rheumatology (AREA)
  • Endocrinology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Otolaryngology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US12/618,612 2008-11-13 2009-11-13 Therapies for hematologic malignancies Abandoned US20100202963A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US12/618,612 US20100202963A1 (en) 2008-11-13 2009-11-13 Therapies for hematologic malignancies
US13/417,185 US9492449B2 (en) 2008-11-13 2012-03-09 Therapies for hematologic malignancies
US13/762,238 US9238070B2 (en) 2008-11-13 2013-02-07 Therapies for hematologic malignancies
US14/323,925 US20140323439A1 (en) 2008-11-13 2014-07-03 Therapies for hematologic malignancies
US15/277,857 US10154998B2 (en) 2008-11-13 2016-09-27 Therapies for hematologic malignancies
US16/189,435 US20190083498A1 (en) 2008-11-13 2018-11-13 Therapies for hematologic malignancies
US16/989,685 US20200368243A1 (en) 2008-11-13 2020-08-10 Therapies for hematologic malignancies

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US11443408P 2008-11-13 2008-11-13
US14284509P 2009-01-06 2009-01-06
US15505709P 2009-02-24 2009-02-24
US18076809P 2009-05-22 2009-05-22
US23127809P 2009-08-04 2009-08-04
US24519609P 2009-09-23 2009-09-23
US12/618,612 US20100202963A1 (en) 2008-11-13 2009-11-13 Therapies for hematologic malignancies

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US13/417,185 Continuation-In-Part US9492449B2 (en) 2008-11-13 2012-03-09 Therapies for hematologic malignancies
US13/762,238 Continuation US9238070B2 (en) 2008-11-13 2013-02-07 Therapies for hematologic malignancies
US15/277,857 Continuation US10154998B2 (en) 2008-11-13 2016-09-27 Therapies for hematologic malignancies

Publications (1)

Publication Number Publication Date
US20100202963A1 true US20100202963A1 (en) 2010-08-12

Family

ID=41604034

Family Applications (5)

Application Number Title Priority Date Filing Date
US12/618,612 Abandoned US20100202963A1 (en) 2008-11-13 2009-11-13 Therapies for hematologic malignancies
US13/762,238 Active 2030-05-27 US9238070B2 (en) 2008-11-13 2013-02-07 Therapies for hematologic malignancies
US14/323,925 Abandoned US20140323439A1 (en) 2008-11-13 2014-07-03 Therapies for hematologic malignancies
US16/189,435 Abandoned US20190083498A1 (en) 2008-11-13 2018-11-13 Therapies for hematologic malignancies
US16/989,685 Abandoned US20200368243A1 (en) 2008-11-13 2020-08-10 Therapies for hematologic malignancies

Family Applications After (4)

Application Number Title Priority Date Filing Date
US13/762,238 Active 2030-05-27 US9238070B2 (en) 2008-11-13 2013-02-07 Therapies for hematologic malignancies
US14/323,925 Abandoned US20140323439A1 (en) 2008-11-13 2014-07-03 Therapies for hematologic malignancies
US16/189,435 Abandoned US20190083498A1 (en) 2008-11-13 2018-11-13 Therapies for hematologic malignancies
US16/989,685 Abandoned US20200368243A1 (en) 2008-11-13 2020-08-10 Therapies for hematologic malignancies

Country Status (13)

Country Link
US (5) US20100202963A1 (de)
EP (2) EP2355828B1 (de)
JP (3) JP5794919B2 (de)
KR (3) KR101664511B1 (de)
CN (2) CN102271683B (de)
AU (6) AU2009313878B2 (de)
CA (3) CA2743642C (de)
ES (1) ES2674719T3 (de)
IL (1) IL212851A0 (de)
NZ (3) NZ631024A (de)
PT (1) PT2355828T (de)
SG (2) SG10201703911XA (de)
WO (1) WO2010057048A1 (de)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060079538A1 (en) * 2004-05-13 2006-04-13 Dennis Hallahan Methods for inhibiting angiogenesis
US20080275067A1 (en) * 2004-05-13 2008-11-06 Icos Corporation Quinazolinones as Inhibitors of Human Phosphatidylinositol 3-Kinase Delta
US20100029693A1 (en) * 2003-08-14 2010-02-04 Jason Douangpanya Novel pi3k delta inhibitors and methods of use thereof
US20100152211A1 (en) * 2000-04-25 2010-06-17 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US20110044942A1 (en) * 2009-04-20 2011-02-24 Puri Kamal D Methods of treatment for solid tumors
WO2012064671A1 (en) * 2010-11-08 2012-05-18 The Ohio State University Research Foundation Compositions and methods for increasing drug efficacy in cancer
WO2012121953A1 (en) * 2011-03-08 2012-09-13 The Trustees Of Columbia University In The City Of New York Methods and pharmaceutical compositions for treating lymphoid malignancy
US8440677B2 (en) 2009-03-24 2013-05-14 Gilead Calistoga Llc Atropisomers of 2-purinyl-3-tolyl-quinazolinone derivatives and methods of use
WO2013134288A1 (en) * 2012-03-05 2013-09-12 Gilead Calistoga Llc Polymorphic forms of (s)-2-(1-(9h-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3h)-one
US8691829B2 (en) 2009-07-21 2014-04-08 Gilead Calistoga Llc Treatment of liver disorders with PI3K inhibitors
US20150105383A1 (en) * 2013-10-10 2015-04-16 Acetylon Pharmaceuticals, Inc. HDAC Inhibitors, Alone Or In Combination With PI3K Inhibitors, For Treating Non-Hodgkin's Lymphoma
US9056852B2 (en) 2011-03-28 2015-06-16 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US20150320754A1 (en) * 2014-04-16 2015-11-12 Infinity Pharmaceuticals, Inc. Combination therapies
US9238070B2 (en) 2008-11-13 2016-01-19 Gilead Calistoga Llc Therapies for hematologic malignancies
CN105682658A (zh) * 2013-05-30 2016-06-15 无限药品公司 使用pi3激酶亚型调节剂的癌症疗法
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
US9567337B2 (en) 2013-12-20 2017-02-14 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
US9708327B2 (en) 2013-12-20 2017-07-18 Gilead Calistoga Llc Polymorphic forms of a hydrochloride salt of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US9822131B2 (en) 2008-01-04 2017-11-21 Intellikine Llc Certain chemical entities, compositions and methods
USRE46621E1 (en) 2011-01-10 2017-12-05 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
AU2017201779B2 (en) * 2012-03-05 2018-10-18 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US10208066B2 (en) 2014-09-24 2019-02-19 Hutchison Medipharma Limited Imidazopyridazine compounds and their use
US11021467B2 (en) 2014-06-13 2021-06-01 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies
US11304953B2 (en) 2017-05-23 2022-04-19 Mei Pharma, Inc. Combination therapy
US11351176B2 (en) 2017-08-14 2022-06-07 Mei Pharma, Inc. Combination therapy
US11433065B2 (en) 2008-01-04 2022-09-06 Intellikine Llc Certain chemical entities, compositions and methods

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ597050A (en) 2009-06-29 2014-02-28 Incyte Corp Pyrimidinones as pi3k inhibitors
US8759359B2 (en) 2009-12-18 2014-06-24 Incyte Corporation Substituted heteroaryl fused derivatives as PI3K inhibitors
EP2558463A1 (de) 2010-04-14 2013-02-20 Incyte Corporation Kondensierte derivate als i3-hemmer
CA3154024C (en) 2010-06-03 2024-02-27 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (btk) in the treatment of relapsed or refractory follicular lymphoma
EP2579878A1 (de) * 2010-06-11 2013-04-17 Gilead Calistoga LLC Verfahren zur behandlung hämatologischer störungen mit chinazolinonverbindungen bei ausgewählten patienten
WO2011163195A1 (en) * 2010-06-21 2011-12-29 Incyte Corporation Fused pyrrole derivatives as pi3k inhibitors
DE102010039631A1 (de) * 2010-08-22 2012-02-23 Ulrich Schubert Pharmazeutische Zusammensetzungen mit immunmodulatorischen Eigenschaften
CA2818163A1 (en) * 2010-11-17 2012-05-24 Niiki Pharma Inc. Method of treating hematological cancers
AR084366A1 (es) 2010-12-20 2013-05-08 Incyte Corp N-(1-(fenil sustituido)etil)-9h-purin-6-aminas como inhibidores de pi3k
SG10201601909TA (en) * 2011-03-11 2016-04-28 Gilead Calistoga Llc Combination Therapies For Hematologic Malignancies
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
WO2012135009A1 (en) 2011-03-25 2012-10-04 Incyte Corporation Pyrimidine-4,6-diamine derivatives as pi3k inhibitors
BR122019020716B1 (pt) 2011-09-02 2021-02-17 Incyte Holdings Corporation heterociclilaminas como inibidores de pi3k e composição farmacêutica que as compreende
WO2013082540A1 (en) * 2011-12-02 2013-06-06 Gilead Calistoga Llc Compositions and methods of treating a proliferative disease with a quinazolinone derivative
EA029473B1 (ru) 2011-12-15 2018-03-30 Новартис Аг Применение ингибиторов pi3k для лечения острой и церебральной малярии
AR090548A1 (es) 2012-04-02 2014-11-19 Incyte Corp Azaheterociclobencilaminas biciclicas como inhibidores de pi3k
US8828998B2 (en) 2012-06-25 2014-09-09 Infinity Pharmaceuticals, Inc. Treatment of lupus, fibrotic conditions, and inflammatory myopathies and other disorders using PI3 kinase inhibitors
HUE034591T2 (en) 2012-07-04 2018-02-28 Rhizen Pharmaceuticals S A Selective PI3K delta inhibitors
MX2015001081A (es) 2012-07-24 2015-10-14 Pharmacyclics Inc Mutaciones asociadas a resistencia a inhibidores de la tirosina cinasa de bruton (btk).
CN105102000B (zh) * 2012-11-01 2021-10-22 无限药品公司 使用pi3激酶亚型调节剂的癌症疗法
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
PL2968555T3 (pl) * 2013-03-13 2020-11-16 Sanofi Kompozycje zawierające przeciwciała anty-cd38 i karfilzomib
KR102240356B1 (ko) * 2013-04-17 2021-04-14 시그날 파마소티칼 엘엘씨 Tor 키나제 억제제와 5-치환된 퀴나졸리논 화합물을 포함하는 암 치료용 조합 요법
UY35675A (es) 2013-07-24 2015-02-27 Novartis Ag Derivados sustituidos de quinazolin-4-ona
WO2015014851A1 (en) * 2013-07-30 2015-02-05 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch for the treatment of hematological neoplasms
ES2880999T3 (es) 2014-05-27 2021-11-26 Rhizen Pharmaceuticals S A Sal de tosilato cristalina de un inhibidor selectivo de PI3K delta para uso en formulaciones farmacéuticas
US10077277B2 (en) 2014-06-11 2018-09-18 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
MD3262046T2 (ro) 2015-02-27 2021-03-31 Incyte Corp Săruri de inhibitori ai PI3K și procedee pentru prepararea lor
WO2016183063A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Crystalline forms of a pi3k inhibitor
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
WO2017214140A1 (en) * 2016-06-06 2017-12-14 University Of Iowa Research Foundation Compositions and methods for cancer therapy
KR101932146B1 (ko) * 2016-07-14 2018-12-24 주식회사 바이오웨이 Pi3k를 억제하는 신규한 퀴나졸리논 유도체 및 이를 포함하는 약학적 조성물
WO2018017928A1 (en) * 2016-07-22 2018-01-25 Seattle Genetics, Inc. Combination therapy using a cd19-adc and rchp
TW201825465A (zh) 2016-09-23 2018-07-16 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
TW201813963A (zh) 2016-09-23 2018-04-16 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
TW201815787A (zh) 2016-09-23 2018-05-01 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
EP3630079A4 (de) * 2017-05-31 2021-02-24 The Children's Medical Center Corporation Targeting von lysin-demethylasen (kdm) als therapeutische strategie für diffuses grosses b-zell-lymphom
SG11201912403SA (en) 2017-06-22 2020-01-30 Celgene Corp Treatment of hepatocellular carcinoma characterized by hepatitis b virus infection
AU2018309739B2 (en) * 2017-07-31 2022-01-27 The Trustees Of Columbia University In The City Of New York Compounds, Compositions, and Methods for Treating T-cell Acute Lymphoblastic Leukemia
CN111658636B (zh) * 2020-06-12 2021-04-13 桂林医学院 穿心莲内酯与地塞米松联用在制备复方抗急性淋巴细胞白血病药物中的应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060106038A1 (en) * 2004-05-25 2006-05-18 Icos Corporation Methods for treating and/or preventing aberrant proliferation of hematopoietic cells

Family Cites Families (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1053063A (de) 1963-05-18
US3691016A (en) 1970-04-17 1972-09-12 Monsanto Co Process for the preparation of insoluble enzymes
DE2027645A1 (de) 1970-06-05 1971-12-09 Byk Gulden Lomberg Chemische Fa bnk GmbH, 7750 Konstanz Piperazinylalkyl chinazolon (4) den vate, Verfahren zu deren Herstellung und sie enthaltende Arzneimittel
NL7204972A (de) 1971-04-21 1972-10-24
US3897432A (en) 1971-04-21 1975-07-29 Merck & Co Inc Substituted benzimidazole derivatives
CA1023287A (en) 1972-12-08 1977-12-27 Boehringer Mannheim G.M.B.H. Process for the preparation of carrier-bound proteins
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4195128A (en) 1976-05-03 1980-03-25 Bayer Aktiengesellschaft Polymeric carrier bound ligands
DE2644265C2 (de) 1976-09-30 1983-02-10 Bayer Ag, 5090 Leverkusen Chinazoline
US4330440A (en) 1977-02-08 1982-05-18 Development Finance Corporation Of New Zealand Activated matrix and method of activation
CA1093991A (en) 1977-02-17 1981-01-20 Hideo Hirohara Enzyme immobilization with pullulan gel
US4183931A (en) 1977-09-08 1980-01-15 Research Corporation 2-Ketoalkyl-4(3H)-quinazolinones
US4229537A (en) 1978-02-09 1980-10-21 New York University Preparation of trichloro-s-triazine activated supports for coupling ligands
DE2812635A1 (de) 1978-03-22 1979-09-27 Bayer Ag Heterocyclische verbindungen
JPS55118918U (de) 1979-01-19 1980-08-22
JPS6124020Y2 (de) 1979-01-19 1986-07-18
JPS562322Y2 (de) 1979-03-09 1981-01-20
US4289872A (en) 1979-04-06 1981-09-15 Allied Corporation Macromolecular highly branched homogeneous compound based on lysine units
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
DE3675588D1 (de) 1985-06-19 1990-12-20 Ajinomoto Kk Haemoglobin, das an ein poly(alkenylenoxid) gebunden ist.
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
USRE35862E (en) 1986-08-18 1998-07-28 Emisphere Technologies, Inc. Delivery systems for pharmacological agents encapsulated with proteinoids
US4925673A (en) 1986-08-18 1990-05-15 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents encapsulated with proteinoids
US6696250B1 (en) 1986-12-03 2004-02-24 Competitive Technologies, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5229490A (en) 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
US5225347A (en) 1989-09-25 1993-07-06 Innovir Laboratories, Inc. Therapeutic ribozyme compositions and expression vectors
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
FR2675803B1 (fr) 1991-04-25 1996-09-06 Genset Sa Oligonucleotides fermes, antisens et sens et leurs applications.
ATE135583T1 (de) 1991-06-18 1996-04-15 American Home Prod Verwendung von rapamycin zur behandlung von t- zellen lymphom/leukämie bei erwachsenen
EP0636156B1 (de) 1992-04-14 1997-07-30 Cornell Research Foundation, Inc. Makromoleküle auf basis von dendritischen polymeren und verfahren zur herstellung
US5658780A (en) 1992-12-07 1997-08-19 Ribozyme Pharmaceuticals, Inc. Rel a targeted ribozymes
GB9301000D0 (en) 1993-01-20 1993-03-10 Glaxo Group Ltd Chemical compounds
US5378725A (en) 1993-07-19 1995-01-03 The Arizona Board Of Regents Inhibition of phosphatidylinositol 3-kinase with wortmannin and analogs thereof
GB9404485D0 (en) 1994-03-09 1994-04-20 Cancer Res Campaign Tech Benzamide analogues
FI951367A (fi) 1994-03-28 1995-09-29 Japan Energy Corp Puriinijohdannaiset ja tulehdustautien tukahduttajat (suppressantit)
US5480906A (en) 1994-07-01 1996-01-02 Eli Lilly And Company Stereochemical Wortmannin derivatives
WO1996004923A1 (en) 1994-08-12 1996-02-22 Pro-Neuron, Inc. Methods for treating sepsis or inflammatory diseases with oxypurine nucleosides
US5561138A (en) 1994-12-13 1996-10-01 American Home Products Corporation Method of treating anemia
US6043062A (en) 1995-02-17 2000-03-28 The Regents Of The University Of California Constitutively active phosphatidylinositol 3-kinase and uses thereof
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
US5948664A (en) 1996-02-29 1999-09-07 The Regents Of The University Of California PI 3-kinase polypeptides
EP0904107B1 (de) 1996-03-18 2004-10-20 Board Of Regents, The University Of Texas System Immunglobulinähnliche domäne mit erhöhten halbwertszeiten
SK284108B6 (sk) 1996-05-15 2004-09-08 Pfizer Inc. 2,3-Disubstituované 4(3H)-chinazolinóny, ich použitie a farmaceutické kompozície s ich obsahom
GB9611460D0 (en) 1996-06-01 1996-08-07 Ludwig Inst Cancer Res Novel lipid kinase
US5858753A (en) 1996-11-25 1999-01-12 Icos Corporation Lipid kinase
TW528755B (en) 1996-12-24 2003-04-21 Glaxo Group Ltd 2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
ES2199366T3 (es) 1996-12-31 2004-02-16 Dr. Reddy's Laboratories Ltd. Compuestos heterociclicos, procedimiento para su preparacion y composiciones farmaceuticas que los contienen y su uso en el tratamiento de diabetes y enfermedades relacionadas.
GB9702701D0 (en) 1997-02-01 1997-04-02 Univ Newcastle Ventures Ltd Quinazolinone compounds
ATE267817T1 (de) 1997-02-28 2004-06-15 Pfizer Prod Inc Atropisomere von 3-aryl-4(3h)-chinazolinonen und ihre verwendung als ampa-rezeptor
ES2245015T3 (es) 1997-06-09 2005-12-16 Pfizer Products Inc. Quinazolin-4-onas como antagonistas de ampa.
AU8280798A (en) 1997-07-03 1999-01-25 Thomas Jefferson University An improved method for design and selection of efficacious antisense oligonucleotides
IL125950A0 (en) 1997-09-05 1999-04-11 Pfizer Prod Inc Methods of administering ampa receptor antagonists to treat dyskinesias associated with dopamine agonist therapy
US20010051654A1 (en) * 1997-09-25 2001-12-13 Elliott Peter J. Treatment of inflammatory and autoimmune diseases
CA2219867A1 (en) * 1997-10-31 1999-04-30 Jiangping Wu The use of proteasome inhibitors for treating cancer, inflammation, autoimmune disease, graft rejection and septic shock
WO1999034804A1 (en) 1998-01-08 1999-07-15 The University Of Virginia Patent Foundation A2a adenosine receptor agonists
DE69828445D1 (de) 1998-04-23 2005-02-03 Reddys Lab Ltd Dr Heterozyklische verbindungen,und deren verwendung in arzneimittel,verfahren zu deren herstellung und diese enthaltende pharmazeutische zusammenstellungen
US6048970A (en) 1998-05-22 2000-04-11 Incyte Pharmaceuticals, Inc. Prostate growth-associated membrane proteins
US6100090A (en) 1999-06-25 2000-08-08 Isis Pharmaceuticals Inc. Antisense inhibition of PI3K p85 expression
EP1949911A3 (de) * 1998-11-09 2008-08-06 Biogen Idec, Inc. Behandlung von chronischer lymphozitärer Leukämie (CLL) unter Verwendung chimärer Anti-CD20-Antikörper
US6046049A (en) 1999-07-19 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of PI3 kinase p110 delta expression
ES2263501T3 (es) 1999-10-27 2006-12-16 Cytokinetics, Inc. Procedimiento y composiciones que utilizan quinazolinonas.
EP1257537B1 (de) 2000-01-24 2007-05-30 AstraZeneca AB Durch einen morpholinrest substituierte therapeutische verbindungen
JP2003522174A (ja) 2000-02-07 2003-07-22 ブリストル−マイヤーズ スクイブ カンパニー サイクリン依存性キナーゼの3−アミノピラゾール阻害剤
WO2001082346A1 (fr) 2000-04-24 2001-11-01 Beijing Normal University Procede de fabrication d'un materiau en silicium sur isolant (soi)
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
DE60144277D1 (de) 2000-04-25 2011-05-05 Icos Corp Hemmer der menschlichen phosphatidyl-inositol-3-kinase delta
AU2003247483A1 (en) 2002-05-30 2003-12-31 The Children's Hospital Of Philadelphia Methods for treatment of acute lymphocytic leukemia
US20040092561A1 (en) 2002-11-07 2004-05-13 Thomas Ruckle Azolidinone-vinyl fused -benzene derivatives
JP4782564B2 (ja) 2002-07-10 2011-09-28 メルク セローノ ソシエテ アノニム アゾリジノン−ビニル縮合−ベンゼン誘導体
GB0217777D0 (en) 2002-07-31 2002-09-11 Novartis Ag Organic compounds
US20040023390A1 (en) 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
CA2400254A1 (en) 2002-09-19 2004-03-19 University Health Network Compositions and methods for treating heart disease
PT2042504E (pt) 2002-09-30 2011-09-07 Bayer Schering Pharma Ag Derivados da azolepirimidina fundida
WO2004052373A1 (en) 2002-12-06 2004-06-24 Warner-Lambert Company Llc Benzoxazin-3-ones and derivatives thereof as inhibitors of pi3k
AU2003303231A1 (en) 2002-12-20 2004-07-14 Warner-Lambert Company Llc Benzoxazines and derivatives thereof as inhibitors of pi3ks
SG160211A1 (en) 2003-04-03 2010-04-29 Semafore Pharmaceuticals Inc Pi-3 kinase inhibitor prodrugs
MXPA05013210A (es) 2003-06-05 2006-03-09 Warner Lambert Co Benzo[b]tiofenos cicloalquilsulfanil-sustituidos como agentes terapeuticos.
JP2006526608A (ja) 2003-06-05 2006-11-24 ワーナー−ランバート カンパニー リミティド ライアビリティー カンパニー Pi3k活性を有する治療薬としてのテトラゾールベンゾフランカルボキシアミド
EP1644360A1 (de) 2003-06-05 2006-04-12 Warner-Lambert Company LLC 3-substituierte indole und ihre derivate als therapeutische mittel
JP2006526614A (ja) 2003-06-05 2006-11-24 ワーナー−ランバート カンパニー リミティド ライアビリティー カンパニー 治療用剤としての3−アリールスルファニル及び3−ヘテロアリールスルファニル置換ベンゾ[b]チオフェン
US20040259926A1 (en) 2003-06-05 2004-12-23 Bruendl Michelle M. 3-Aryloxy and 3-heteroaryloxy substituted benzo[b]thiophenes as therapeutic agents
JP2006526606A (ja) 2003-06-05 2006-11-24 ワーナー−ランバート カンパニー リミティド ライアビリティー カンパニー 治療薬としてのシクロアルキル及びヘテロシクロアルキル置換されるベンゾチオフェン
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
CA2552664A1 (en) 2004-01-08 2005-07-28 Michigan State University Methods for treating and preventing hypertension and hypertension-related disorders
US20100009929A1 (en) * 2004-03-29 2010-01-14 Cheng Jin Q Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
EP1750714A1 (de) 2004-05-13 2007-02-14 Vanderbilt University Phosphoinositid 3-kinase delta selektive hemmer zur hemmung der angiogenese
CN101031569B (zh) 2004-05-13 2011-06-22 艾科斯有限公司 作为人磷脂酰肌醇3-激酶δ抑制剂的喹唑啉酮
WO2005120511A1 (en) * 2004-06-04 2005-12-22 Icos Corporation Methods for treating mast cell disorders
US20060084691A1 (en) * 2004-10-18 2006-04-20 Bilal Piperdi Combined treatment with bortezomib and an epidermal growth factor receptor kinase inhibitor
US20080287469A1 (en) 2005-02-17 2008-11-20 Diacovo Thomas G Phosphoinositide 3-Kinase Inhibitors for Inhibiting Leukocyte Accumulation
CN100441222C (zh) 2005-12-09 2008-12-10 中国科学院广州生物医药与健康研究院 化合物ps-341在制备治疗急性髓性白血病药物中的应用
AR057227A1 (es) * 2005-12-09 2007-11-21 Centocor Inc Metodo para usar antagonistas de il6 con inhibidores del proteasoma
AU2008209555A1 (en) * 2007-01-23 2008-07-31 Gloucester Pharmaceuticals, Inc. Combination therapy comprising romidepsin and I.A. bortezomib
WO2009052467A1 (en) 2007-10-19 2009-04-23 Board Of Regents Of The University Of Texas System Methods of identifying pi-3-kinase inhibitor resistance
US20090131512A1 (en) 2007-10-31 2009-05-21 Dynavax Technologies Corp. Inhibition of type I in IFN production
AU2009313878B2 (en) * 2008-11-13 2016-01-07 Gilead Calistoga Llc Therapies for hematologic malignancies
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
CA2745280A1 (en) 2008-12-04 2010-06-10 The United States Of America, As Represented By The Secretary, Departmen T Of Health And Human Services Phosphatidylinositol-3-kinase p110 delta-targeted drugs in the treatment of cns disorders
WO2010071944A1 (en) 2008-12-24 2010-07-01 Prana Biotechnology Limited Quinazolinone compounds
BRPI1012333A2 (pt) 2009-03-24 2016-03-29 Gilead Calistoga Llc atropisômeros de derivados de 2-purinil-3-tolil-quinazolinonas e métodos de uso
US8546409B2 (en) 2009-04-20 2013-10-01 Gilead Calistoga Llc Methods of treatment for solid tumors
WO2011032277A1 (en) 2009-09-18 2011-03-24 Boehringer Ingelheim International Gmbh Quinazolinone derivatives as viral polymerase inhibitors
SG10201601909TA (en) 2011-03-11 2016-04-28 Gilead Calistoga Llc Combination Therapies For Hematologic Malignancies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060106038A1 (en) * 2004-05-25 2006-05-18 Icos Corporation Methods for treating and/or preventing aberrant proliferation of hematopoietic cells

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Byrd et al. Blood, Published online 2004, Vol. 105, pgs. 49-53. *
Gokbuget et al. Hematology, 2006, Vol. 2006, No. 1, pgs. 133-141. *
Kurcharsky, D. DocGuide.com, June 2005, pgs 1-2. *

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10695349B2 (en) 2000-04-25 2020-06-30 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US9487772B2 (en) 2000-04-25 2016-11-08 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US20100152211A1 (en) * 2000-04-25 2010-06-17 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US20100168139A1 (en) * 2000-04-25 2010-07-01 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US8653077B2 (en) 2000-04-25 2014-02-18 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US8637533B2 (en) 2000-04-25 2014-01-28 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US8623881B2 (en) 2000-04-25 2014-01-07 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US10010550B2 (en) 2000-04-25 2018-07-03 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US8492389B2 (en) 2000-04-25 2013-07-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US10398695B2 (en) 2000-04-25 2019-09-03 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US20100029693A1 (en) * 2003-08-14 2010-02-04 Jason Douangpanya Novel pi3k delta inhibitors and methods of use thereof
US10336756B2 (en) 2004-05-13 2019-07-02 Icos Corporation (S)-2-(1-aminopropyl)-5-fluoro-3-phenylquinazolin-4(3H)-one precursor of a quinazolinone as inhibitor of human phosphatidylinositol 3-kinase delta
US20100256167A1 (en) * 2004-05-13 2010-10-07 Fowler Kerry W Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
US10906907B2 (en) 2004-05-13 2021-02-02 Icos Corporation Tert-butyl (s)-(1-(5-fluoro-4-oxo-3-phenyl-3,4-dihydroquinazolin-2-yl)propyl)carbamate precursor of a quinazolinone inhibitor of human phosphatidylinositol 3-kinase delta and a process for preparing thereof
US9149477B2 (en) 2004-05-13 2015-10-06 Icos Corporation 5-fluoro-3-phenyl-2-[1-(9h-purin-6-ylamino)propyl]-3h-quinazolin-4-one as an inhibitor of human phosphatidylinositol 3-kinase delta
US7932260B2 (en) 2004-05-13 2011-04-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
USRE44599E1 (en) 2004-05-13 2013-11-12 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
US8586597B2 (en) 2004-05-13 2013-11-19 Icos Corporation 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)ethyl]-3H-quinazolin-4-one as an inhibitor of human phosphatidylinositol 3-kinase delta
USRE44638E1 (en) 2004-05-13 2013-12-10 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
US20080275067A1 (en) * 2004-05-13 2008-11-06 Icos Corporation Quinazolinones as Inhibitors of Human Phosphatidylinositol 3-Kinase Delta
US8207153B2 (en) 2004-05-13 2012-06-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
US20100256168A1 (en) * 2004-05-13 2010-10-07 Fowler Kerry W Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
US8993583B2 (en) 2004-05-13 2015-03-31 Icos Corporation 5-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)propyl]-3H-quinazolin-4-one and 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)ethyl]-3H-quinazolin-4-one as inhibitors of human phosphatidylinositol 3-kinase delta
US8779131B2 (en) 2004-05-13 2014-07-15 Icos Corporation 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)-ethyl]-3H-quinazolin-4-one as an inhibitor of human phosphatidylinositol 3-kinase delta
US20060079538A1 (en) * 2004-05-13 2006-04-13 Dennis Hallahan Methods for inhibiting angiogenesis
US8980901B2 (en) 2004-05-13 2015-03-17 Icos Corporation 5-fluoro-3-phenyl-2[1-(9H-purin-6-ylamino)propyl]-3H-quinazolin-4-one and 6-fluoro-3-phenyl-2-[1-(9H-purin-6-ylamino)ethyl]-3H-quinazolin-4-one as inhibitors of human phosphatidylinositol 3-kinase delta
US9822131B2 (en) 2008-01-04 2017-11-21 Intellikine Llc Certain chemical entities, compositions and methods
US11433065B2 (en) 2008-01-04 2022-09-06 Intellikine Llc Certain chemical entities, compositions and methods
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
US9238070B2 (en) 2008-11-13 2016-01-19 Gilead Calistoga Llc Therapies for hematologic malignancies
US10154998B2 (en) 2008-11-13 2018-12-18 Gilead Calistoga Llc Therapies for hematologic malignancies
US8440677B2 (en) 2009-03-24 2013-05-14 Gilead Calistoga Llc Atropisomers of 2-purinyl-3-tolyl-quinazolinone derivatives and methods of use
US20110044942A1 (en) * 2009-04-20 2011-02-24 Puri Kamal D Methods of treatment for solid tumors
US8546409B2 (en) 2009-04-20 2013-10-01 Gilead Calistoga Llc Methods of treatment for solid tumors
US8691829B2 (en) 2009-07-21 2014-04-08 Gilead Calistoga Llc Treatment of liver disorders with PI3K inhibitors
WO2012064671A1 (en) * 2010-11-08 2012-05-18 The Ohio State University Research Foundation Compositions and methods for increasing drug efficacy in cancer
US9840505B2 (en) 2011-01-10 2017-12-12 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1 (2H)-one and methods of use thereof
US11312718B2 (en) 2011-01-10 2022-04-26 Infinity Pharmaceuticals, Inc. Formulations of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one
US10550122B2 (en) 2011-01-10 2020-02-04 Infinity Pharmaceuticals, Inc. Solid forms of (S)-3-(1-(9H-purin-6-ylamino)ethyl)-8-chloro-2-phenylisoquinolin-1(2H)-one and methods of use thereof
USRE46621E1 (en) 2011-01-10 2017-12-05 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
US20190209570A1 (en) * 2011-03-08 2019-07-11 The Trustees Of Columbia University In The City Of New York Methods and Pharmaceutical Compositions for Treating Lymphoid Malignancy
WO2012121953A1 (en) * 2011-03-08 2012-09-13 The Trustees Of Columbia University In The City Of New York Methods and pharmaceutical compositions for treating lymphoid malignancy
US10064868B2 (en) 2011-03-28 2018-09-04 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US11400097B2 (en) 2011-03-28 2022-08-02 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US9056852B2 (en) 2011-03-28 2015-06-16 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US10603324B2 (en) 2011-03-28 2020-03-31 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US10335415B2 (en) 2011-03-28 2019-07-02 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
EA025407B1 (ru) * 2012-03-05 2016-12-30 Джилид Калистога Ллс Полиморфная форма i (s)-2-(1-(9h-пурин-6-иламино)пропил)-5-фтор-3-фенилхиназолин-4(3h)-она
AU2017201779B2 (en) * 2012-03-05 2018-10-18 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
WO2013134288A1 (en) * 2012-03-05 2013-09-12 Gilead Calistoga Llc Polymorphic forms of (s)-2-(1-(9h-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3h)-one
US9469643B2 (en) 2012-03-05 2016-10-18 Gilead Calistoga, LLC. Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
AU2013203620B2 (en) * 2012-03-05 2015-11-19 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US8865730B2 (en) 2012-03-05 2014-10-21 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US10730879B2 (en) 2012-03-05 2020-08-04 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
AU2019200160B2 (en) * 2012-03-05 2020-01-16 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
CN105682658A (zh) * 2013-05-30 2016-06-15 无限药品公司 使用pi3激酶亚型调节剂的癌症疗法
US20150105383A1 (en) * 2013-10-10 2015-04-16 Acetylon Pharmaceuticals, Inc. HDAC Inhibitors, Alone Or In Combination With PI3K Inhibitors, For Treating Non-Hodgkin's Lymphoma
US10414737B2 (en) 2013-12-20 2019-09-17 Gilead Sciences, Inc. Process methods for phosphatidylinositol 3-kinase inhibitors
US9708327B2 (en) 2013-12-20 2017-07-18 Gilead Calistoga Llc Polymorphic forms of a hydrochloride salt of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US10059677B2 (en) 2013-12-20 2018-08-28 Gilead Calistoga Llc Process for preparing phosphatidylinositol 3-kinase inhibitors and intermediates thereof
US10047060B2 (en) 2013-12-20 2018-08-14 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
US10954199B2 (en) 2013-12-20 2021-03-23 Gilead Sciences, Inc. Process methods for phosphatidylinositol 3-kinase inhibitors
US10442805B2 (en) 2013-12-20 2019-10-15 Gilead Calistoga Llc Polymorphic forms of a hydrochloride salt of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US9567337B2 (en) 2013-12-20 2017-02-14 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
US11944631B2 (en) 2014-04-16 2024-04-02 Infinity Pharmaceuticals, Inc. Combination therapies
US20150320754A1 (en) * 2014-04-16 2015-11-12 Infinity Pharmaceuticals, Inc. Combination therapies
US11110096B2 (en) 2014-04-16 2021-09-07 Infinity Pharmaceuticals, Inc. Combination therapies
US11021467B2 (en) 2014-06-13 2021-06-01 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10208066B2 (en) 2014-09-24 2019-02-19 Hutchison Medipharma Limited Imidazopyridazine compounds and their use
US10611777B2 (en) 2014-09-24 2020-04-07 Hutchison Medipharma Limited Imidazopyridazine compounds and their use
US11147818B2 (en) 2016-06-24 2021-10-19 Infinity Pharmaceuticals, Inc. Combination therapies
US11304953B2 (en) 2017-05-23 2022-04-19 Mei Pharma, Inc. Combination therapy
US11351176B2 (en) 2017-08-14 2022-06-07 Mei Pharma, Inc. Combination therapy

Also Published As

Publication number Publication date
NZ611764A (en) 2015-01-30
NZ592880A (en) 2013-06-28
EP2355828A1 (de) 2011-08-17
AU2017200837A1 (en) 2017-03-02
NZ631024A (en) 2016-04-29
AU2016201634B2 (en) 2016-12-01
AU2018247326A1 (en) 2018-11-08
AU2009313878B2 (en) 2016-01-07
AU2009313878A1 (en) 2010-05-20
AU2019213337A1 (en) 2019-08-29
SG195655A1 (en) 2013-12-30
US20200368243A1 (en) 2020-11-26
CN102271683A (zh) 2011-12-07
CA2743642A1 (en) 2010-05-20
KR20160091440A (ko) 2016-08-02
JP2015155436A (ja) 2015-08-27
JP5794919B2 (ja) 2015-10-14
US9238070B2 (en) 2016-01-19
CA3092449A1 (en) 2010-05-20
EP2355828B1 (de) 2018-05-30
US20140323439A1 (en) 2014-10-30
ES2674719T3 (es) 2018-07-03
EP3427739A1 (de) 2019-01-16
AU2019213337C1 (en) 2021-11-25
AU2018204264C1 (en) 2020-01-23
AU2019213337B2 (en) 2021-03-11
JP2012508775A (ja) 2012-04-12
WO2010057048A1 (en) 2010-05-20
AU2018247326B2 (en) 2020-04-09
KR20110105771A (ko) 2011-09-27
PT2355828T (pt) 2018-07-02
AU2017200837B2 (en) 2018-07-12
CA2975473A1 (en) 2010-05-20
IL212851A0 (en) 2011-07-31
KR101664511B1 (ko) 2016-10-11
US20190083498A1 (en) 2019-03-21
US20140066386A1 (en) 2014-03-06
JP2017008088A (ja) 2017-01-12
AU2018204264A1 (en) 2018-07-05
CN102271683B (zh) 2014-07-09
CA2743642C (en) 2017-09-19
SG10201703911XA (en) 2017-06-29
AU2018204264B2 (en) 2019-05-16
AU2016201634A1 (en) 2016-04-07
CN104042618B (zh) 2018-02-16
CN104042618A (zh) 2014-09-17
CA2975473C (en) 2021-01-19
KR20170060179A (ko) 2017-05-31

Similar Documents

Publication Publication Date Title
AU2019213337B2 (en) Therapies for hematologic malignancies
US20170119771A1 (en) Combination therapies for hematologic malignancies
JP2012508775A5 (de)
US10154998B2 (en) Therapies for hematologic malignancies
OA16585A (en) Combination therapies for hematologic malignancies.

Legal Events

Date Code Title Description
AS Assignment

Owner name: CALISTOGA PHARMACEUTICALS, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GALLATIN, W. MICHAEL;ULRICH, ROGER G.;GIESE, NEILL A.;SIGNING DATES FROM 20100317 TO 20100318;REEL/FRAME:024297/0242

AS Assignment

Owner name: GILEAD CALISTOGA LLC, CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:CALISTOGA PHARMACEUTICALS, INC.;REEL/FRAME:027596/0107

Effective date: 20110526

STCB Information on status: application discontinuation

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