WO2016048982A2 - COMBINATION TREATMENT WITH PI3Kα INHIBITORS AND TAXANES - Google Patents

COMBINATION TREATMENT WITH PI3Kα INHIBITORS AND TAXANES Download PDF

Info

Publication number
WO2016048982A2
WO2016048982A2 PCT/US2015/051387 US2015051387W WO2016048982A2 WO 2016048982 A2 WO2016048982 A2 WO 2016048982A2 US 2015051387 W US2015051387 W US 2015051387W WO 2016048982 A2 WO2016048982 A2 WO 2016048982A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
administered
taxane
cancer
pi3ka
Prior art date
Application number
PCT/US2015/051387
Other languages
French (fr)
Inventor
Rachael L. BRAKE
Natalia Iartchouk
Chirag Patel
Yaping Shou
Original Assignee
Millennium 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 Millennium Pharmaceuticals, Inc. filed Critical Millennium Pharmaceuticals, Inc.
Publication of WO2016048982A2 publication Critical patent/WO2016048982A2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • 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
    • A61P35/00Antineoplastic agents

Definitions

  • the phosphoinositide 3-kinases are members of a unique and conserved family of intracellular lipid kinases that phosphorylate the 3'-OH group on phosphatidylinositols or phosphoinositides.
  • the PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation ( atso et al., 2001).
  • the class I PI3Ks (pi 10a, pi 10 ⁇ , pi 106, and pi ⁇ ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate phosphatidylinositol-3,4,5-trisphosphate (PIP 3 ), which engages downstream effectors such as those in the Akt/PDKl pathway, mTOR, the Tec family kinases, and the Rho family GTPases.
  • PIP 3 phosphatidylinositol-3,4,5-trisphosphate
  • the class II and ⁇ PI3-Ks play a key role in intracellular trafficking through the synthesis of PI(3)P and PI(3,4)P2.
  • the PIKKs are protein kinases that control cell growth (mTORCl) or monitor genomic integrity (ATM, ATR, DNA-P , and hSmg-1).
  • mTORCl control cell growth
  • ATM genomic integrity
  • ATR DNA-P
  • hSmg-1 monitor genomic integrity
  • the production of PIP 3 initiates potent growth and survival signals.
  • the PI3K pathway is activated by direct genetic mutation.
  • PI3K signaling pathway plays a pivotal role in cell proliferation and differentiation, inhibition of this pathway has been shown to be beneficial in hyperproliferative diseases.
  • the alpha (a) isoform of PI3 has been implicated, for example, in a variety of human cancers.
  • Angiogenesis has been shown to selectively require the a isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453;662-6). Mutations in the gene coding for PI3Koc or mutations which lead to upregulation of PI3Ka are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers.
  • mutations in the gene coding for PI3Kot are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K mutations, targeting of this pathway may provide valuable therapeutic opportunities. While other PI3K isoforms such as ⁇ 3 ⁇ or PI3K5 are expressed primarily in hematopoietic cells, PI3 a, along with ⁇ 3 ⁇ , is expressed constitutively.
  • Taxanes are diterpenes produced from the plants of the genus Taxus (yews). Taxanes are anticancer agents that interfere with cell growth by disrupting microtubule function. Examples of taxanes include, but are not limited to, paclitaxel, docetaxel, cabazitaxel, ortataxel, larotaxel, tesetaxel, 10-deacetyltaxol and cephalomannine.
  • Docetaxel (2R, 3S)— N-carboxy-3- phenylisoserine, N-tert-butyl ester, 13-ester with 5 ⁇ -20- ⁇ -1,2 ⁇ ,4,7 ⁇ ,10 ⁇ ,13 ⁇ - hexahydroxytax-l-l-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®.
  • Docetaxel is indicated for the treatment of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric
  • adenocarcinoma adenocarcinoma
  • squamous cell carcinoma of head and neck cancer adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
  • the present invention provides compositions and methods for treating a variety of neoplastic diseases. Furthermore, the present invention also provides compositions and methods for treating a variety of diseases mediated by PI3K-kinase a.
  • the invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a
  • the invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a therapeutically effective amount of a combination of a PI3K- kinase a (PI3Ka) inhibitor and a taxane, wherein the taxane is not paclitaxel, and wherein the PI3 a inhibitor is a compound of the following formula:
  • W 1 is CR 3 ;
  • R 1 is hydrogen
  • R 2 hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and R 3 is amido of formula -C(0)N(R) 2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R) 2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
  • the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
  • the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
  • the neoplastic condition is a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
  • the neoplastic condition is gastric cancer.
  • the cancer is gastrointestinal cancer.
  • the cancer is selected from gastric cancer and gastroesophageal adenocarcinoma.
  • the cancer is gastric cancer.
  • the cancer is gastroesophageal adenocarcinoma.
  • the neoplastic condition is non-small cell lung cancer.
  • the non-small cell lung cancer is squamous non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer. In some embodiments, the subject has a PIK3CA mutation and/or amplification.
  • the invention provides a method of enhancing apoptosis in cancer cells comprising administering to the cells simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane.
  • the invention provides for a method of enhancing apoptosis in cancer cells comprising aciministering to the cells simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3Ka) inhibitor and a taxane, wherein the PI3 a inhibitor is a compound of the following formula:
  • W 1 is CR 3 ;
  • R 1 is hydrogen
  • R 2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
  • R 3 is amido of formula -C(0)N(R)2 or-NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R) 2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
  • the administration takes place in vivo. In some embodiments, the administration takes place in vivo.
  • the invention provides a method of suppressing tumor regrowth in a subject in need thereof comprising administering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PD a) inhibitor and a taxane.
  • the invention provides a method of method of suppressing tumor regrowth in a subject in need thereof comprising adrninistering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the PDKa inhibitor is a compound of the following formula:
  • W 1 is CR 3 ;
  • R 1 is hydrogen
  • R 2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
  • R 3 is amido of formula -C(0)N(R) 2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R) 2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
  • the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the PI3 a inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the PD a inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the PI3Ka inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the taxane.
  • the taxane and the PI3Ka inhibitor are administered
  • the taxane and the PI3Ka inhibitor are administered sequentially, wherein the taxane and the PDKa inhibitor are introduced at two different time points.
  • the taxane is administered to the subject prior to administration of the PI3 a inhibitor.
  • the taxane is administered to the subject about 1, 5, 8, 10, 12, 24, 36 or 72 hours prior to administration of the PD a inhibitor.
  • taxane is administered to the subject about 12 to about 96 hours prior to administration of the PDKcc inhibitor.
  • the taxane is administered to the subject about 12 to about 72 hours prior to administration of the PI3Ka inhibitor.
  • taxane is administered to the subject about 12 to about 36 hours prior to administration of the PDKa inhibitor.
  • the taxane is administered to the subject about 24 hours prior to administration of the PI3Ka inhibitor.
  • the invention provides for a pharmaceutical regimen for the treatment of a disorder mediated by PI3 -kinase a (PDKa), wherein the regimen comprises simultaneous or sequential administration of at least one taxane and at least one PDKa inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel.
  • PDKa PI3 -kinase a
  • the invention provides for a pharmaceutical regimen for the treatment of a disorder mediated by PD-kinase a (PDKa), wherein the regimen comprises simultaneous or sequential administration of at least one taxane and at least one PI3K.cc inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel; and wherein the PI3Ka inhibitor is a compound of the following formula:
  • W 1 is CR 3 ;
  • R 1 is hydrogen
  • R 2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamide, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
  • R 3 is amido of formula -C(0)N(R) 2 or-NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R) 2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
  • the disorder is a cancer selected from the group consisting of non- small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
  • the disorder is a cancer selected from the group consisting of non- small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
  • the disorder is non-small cell lung cancer.
  • the disorder is a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
  • the disorder is gastric cancer.
  • the disorder is squamous non-small cell lung cancer.
  • the disorder is non-squamous non-small cell lung cancer.
  • the subject has a PIK3CA mutation and/or amplification [0019]
  • the regimen comprises at least one cycle wherein the taxane is administered before the PI3Ka inhibitor and wherein the taxane and the PI3Ka inhibitor are not administered on the same day.
  • the cycle comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 25, 30, 40 or more days.
  • the cycle comprises least 5 to 15 days.
  • the cycle consists of 7 days.
  • the cycle consists of 21 days.
  • the PI3Ka inhibitor is administered nine times in a 21 -day cycle.
  • the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17, and 18.
  • the PI3Kct inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18, and 20.
  • the PI3Kct inhibitor is administered according to an intermittent regimen. In some embodiments, the PI3K.cc inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission.
  • the PI3Koc inhibitor is administered for at least 1 day, followed by an intermission in which the PI3Ka inhibitor is not administered for at least 1 day for at least one cycle. In some embodiments, the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3Ka inhibitor is not administered for at least 1 day. In some embodiments, the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1 , 2, 3, 4, 5, or 6 consecutive days. In some embodiments, the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days. In some
  • the PI3Koc inhibitor is administered for 7, 8, 9, 10, 11, 12, 13, or 14 consecutive days, followed by an intermission where the PI3Ka inhibitor is not administered for at least 1, 2, 3, 4, 5, 6, or 7 days.
  • the PBKoc inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3K ⁇ x inhibitor is not administered for at least 3, 4, or 5 consecutive days.
  • the PI3Ka inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In other embodiments, the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for 3 consecutive days of a 7-day cycle followed by an intermission of at least one day. In some embodiments, the PI3Kct inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 4 consecutive days followed by an intermission of 3 consecutive days for at least one 7-day cycle.
  • the PI3Kot inhibitor is administered for 5 consecutive days followed by an intermission of 2 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Kct inhibitor is administered for 6 consecutive days followed by an intermission of 1 day for at least one 7-day cycle.
  • the PI3Ka inhibitor is administered for at least 1 day for at least one 7- day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 2 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 3 days for at least one 7-day cycle. In some embodiments, the PI3Kct inhibitor is adrriinistered for at least 4 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 5 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 6 days for at least one 7-day cycle.
  • the PI3Koc inhibitor is administered every other day. In some embodiments, the PI3Ka inhibitor is administered for three non-consecutive days in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle. In some embodiments, the PI3 a inhibitor is administered on alternate days in a 7-day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered at least 3 times on alternate days within a 7-day cycle. In some embodiments, the PI3Koc inhibitor is administered at least 4 times on alternate days within a 7-day cycle.
  • the taxane is administered on Day 1 and the PI3Ka inhibitor is adrriinistered on the Days 2, 3 and 4 of a 7-day cycle. In some embodiments, the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 3, 4, and 5 of a 7-day cycle. In some embodiments, the taxane is admimstered on Day 1 and the ⁇ 3 ⁇ inhibitor is administered on Days 4, 5 and 6 of a 7-day cycle. In some embodiments, the taxane is administered on Day land the PI3Ka inhibitor is administered on Days 5, 6, and 7 of a 7-day cycle.
  • the taxane is administered on the Day 1 and the PI3Ka inhibitor is administered on Days 2, 4, and 6 of a 7-day cycle. In some embodiments, the taxane is admimstered on Day 1 and the PI3Ka inhibitor is administered on Days 3, 5, and 7 of a 7- day cycle.
  • the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17 and 18 of a 21 -day cycle as shown in Figure 8A. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is adrriinistered on Days 3, 4, 5, 10, 11, 12, 17, 18 and 19 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is adniinistered on Days 4, 5, 6, 11, 12, 13, 18, 19 and 20 of a 21 -day cycle. In some
  • the taxane is administered on Days 1 and 8 and the PDKct inhibitor is administered on Days 5, 6, 7, 12, 13, 14, 19, 20 and 21 of a 21 -day cycle.
  • the taxane is administered on Days 1 and 8 and the PDKot inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18 and 20 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 5, 7, 10, 12, 14, 17, 19 and 21 of a 21 -day cycle.
  • the PI3Ka inhibitor is administered at least once a day in each of the days that the PI3 a inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered once daily in each of the days that the ⁇ 3 ⁇ inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered twice daily in each of the days that the PI3Koc inhibitor is administered to the subject.
  • the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 6300 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 900 mg to about 3600 mg of the PI3Koc inhibitor in a 7-day cycle.
  • the PI3 ct inhibitor is administered to a subject within a range of about an amount of 900 mg to about 6300 mg of the PDKct inhibitor in a 7-day cycle.
  • the amount of PI3Ka inhibitor administered in a 7-day cycle is about 300 mg.
  • the amount of PI3Ka inhibitor administered in a 7-day cycle is about 900 mg.
  • the amount of PDKcc inhibitor administered in a 7-day cycle is about 1800 mg.
  • the amount of PI3Ka inhibitor administered in a 7-day cycle is about 2700 mg.
  • the amount of PI3Ka inhibitor administered in a 7-day cycle is about 3600 mg.
  • the amount of PDKct inhibitor administered in a 7-day cycle is about 4500 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 5400 mg. In some embodiments, the amount of PDKct inhibitor administered in a 7-day cycle is about 6300 mg.
  • a dose of the PI3Ka inhibitor is about 100 to about 1200 mg. In some embodiments, a dose of the PDKct inhibitor is about 100 to about 2100 mg. In some embodiments, a dose of the PDKct inhibitor is about 300 to about 1200 mg. In some embodiments, a dose of the PI3 a inhibitor is about 300 to about 2100 mg. In some embodiments,
  • a dose of the P13 a inhibitor is about 100 mg, about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, or about 1200 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, or about 1200 mg. In some embodiments a dose of the PI3 a inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, or about 1200 mg. In some embodiments a dose of the PI3 a inhibitor is about 100 mg, about 300 mg, about 600 mg, about
  • a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3 a inhibitor is about 100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 600 mg.
  • a dose of the PI3 a inhibitor is about 900 mg. In some embodiments, a dose of the PI3 a inhibitor is about 1200 mg. In some embodiments, a dose of the PDKa inhibitor is about 1500 mg. In some embodiments, a dose of the PI3 a inhibitor is about 1800 mg. In some embodiments, a dose of the PI3 a inhibitor is about 2100 mg.
  • the taxane is administered to a subject at a dose of about 25 mg/m 2 to about 100 mg m 2 in a single administration. In some embodiments, the taxane is administered at a dose of about 25 mg/m 2 , 26 mg/m 2 , 27 mg/m 2 , 28 mg m 2 , 29 mg m 2 , 30 mg/m 2 , 31 mg m 2 , 32 mg m 2 , 33 mg/m 2 , 34 mg/m 2 , 35 mg/m 2 , 36 mg/m 2 , 37 mg/m 2 , 38 mg/m 2 ,
  • the taxane is administered at a dose of about 36 mg/m 2 in a single administration. In some embodiments, the taxane is administered at a dose of about 75 mg/m 2 in a single administration.
  • the taxane is docetaxel. In some embodiments, the taxane is docetaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring. In a further embodiment, R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 6-membered ring. In a further embodiment, R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring. In some embodiments, R 2 is amino. In a further embodiment, R 2 is Nt .
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring and R 2 is amino.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 6-membered ring and R 2 is amino.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring and R 2 is NH 2 .
  • the PO a inhibitor is a compound with the following structure:
  • Figure 1 shows the effect of docetaxel on the phosphorylation of AKT, S6 and 4EBP1 in NCI-H1048 (PIK3CA-mutated) cells.
  • Figure 2A shows the effect of docetaxel and compound A on the in vitro
  • FIG. 2B shows the effect of docetaxel and compound A on the in vitro phosphorylation of S6 240/244 in NCI-H1048 cells.
  • Figure 3A shows a Western blot depicting the effects of compound A and docetaxel on the in vitro apoptosis of NCI-H1048 cells.
  • Figure 3B shows quantified cleaved PARP.
  • Figure 4 shows the effect of docetaxel on the phosphorylation of AKT, S6 and 4EBP1 in Hs746T gastric xenograft tumor samples.
  • Figures 5A and 5B show the increased apoptosis observed in vivo in NCI-H1048 when pre-dosing docetaxel in combination with compound A.
  • Figure 4A shows quantified Cleaved lamin A
  • Figure 5B shows quantified cleaved PARP.
  • Figures 6A and 6B show the effect that pre-dosing docetaxel in combination with compound A has on in vivo anti-tumor activity compared to simultaneous combination dosing in NCI-H1048 cells.
  • Figure 6A shows the effect of pre-dosing docetaxel at a low clinically relevant dose.
  • Figure 6B shows the effect of pre-dosing docetaxel at 10 mg kg.
  • Figure 7 shows the effect that pre-dosing docetaxel in combination with compound A has on enhanced tumor delay on NCI-H1048 cells (compound A: 140 mg kg QD3; docetaxel: 10 mg kg QW).
  • Figure 8 shows the effect that pre-dosing docetaxel for 24h in combination with compound A has on in vivo antitumor activity compared to simultaneous combination dosing in GA0098 xenografts
  • Figure 9 shows the administration of a combination of a taxane and a PI3Ka inhibitor wherein the PI3Kot inhibitor is administered according to an intermittent regimen for a 7-day cycle.
  • Figures 10A and 10B show the aclministration of a combination of a taxane and a PI3Kcc inhibitor wherein the PI3Ka inhibitor is administered according to an intermittent regimen for a 21 -day cycle.
  • Treatment refers to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions may be aclministered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • neoplastic condition or “neoplastic disorder” refers to the presence of cells possessing abnormal growth characteristics, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, perturbed oncogenic signaling, and certain characteristic morphological features. This includes but is not limited to the growth of: (1) benign or malignant cells (e.g., tumor cells) that correlates with
  • tyrosine or serine/threonine kinase overexpression of a tyrosine or serine/threonine kinase; (2) benign or malignant cells (e.g., tumor cells) that correlates with abnormally high level of tyrosine or serine/threonine kinase activity or lipid kinase activity.
  • exemplary tyrosine kinases implicated in a neoplastic condition include but are not limited to receptor tyrosine kinases such as epidermal growth factor receptors (EGF receptor), platelet derived growth factor (PDGF) receptors, and cytosolic tyrosine kinases such as src and abl kinase.
  • Non-limiting serine/threonine kinases implicated in neoplastic condition include but are not limited to raf, mek, mTor, and akt.
  • Exemplary lipid kinases include but are not limited to PI3 kinases such as ⁇ , ⁇ , PI3K5, and ⁇ 3 ⁇ .
  • the term "effective amount” or “therapeutically effective amount” refers to that amount of an inhibitor described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or down regulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • a "sub-therapeutic amount" of an agent or therapy is an amount less than the effective amount for that agent or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a result desired by the physician, due to, for example, synergy in the resulting efficacious effects, or reduced side effects.
  • a "synergistically effective" therapeutic amount, “synergistically effective” amount of an agent or therapy is an amount which, when combined with an effective or sub-therapeutic amount of another agent or therapy, produces a greater effect than when either of the two agents are used alone.
  • a synergistically effective therapeutic amount of an agent or therapy produces a greater effect when used in combination than the additive effects of each of the two agents or therapies when used alone.
  • the term "greater effect” encompasses not only a reduction in symptoms of the disorder to be treated, but also an improved side effect profile, improved tolerability, improved patient compliance, improved efficacy, or any other improved clinical outcome.
  • agent or “biologically active agent” refers to a biological
  • Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound.
  • Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures.
  • various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • agonist refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term “agonist” is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g., bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
  • dose refers to the amount of a particular pharmaceutically active ingredient used in a single/one-time administration.
  • dose refers to the amount of a particular pharmaceutically active ingredient used in a single/one-time administration.
  • dose may be administered at a "dose” of 300 mg each time, and an amount of 900 mg over a course of a 7-day cycle during which the PI3 a inhibitor is administered on Day 2, Day 4 and Day 6 only.
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms “antagonist” and “inhibitors” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
  • a preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
  • PI3Kcc inhibitor refers to a PI3Ka inhibitor that interacts with and reduces activity of PI3Ka kinase.
  • the PI3 a inhibitor can be (6-(2-aminobenzo[d]oxazol- 5-yl)-midazo[l,2-a]pyridin-3-yl)(mo holino)methanone and its pharmaceutically acceptable salts, prodrugs, or radioactive isomers.
  • compound A is (6-(2- ammobenzo[d]oxazol-5-yl)imidazo[l,2-a]pyridin-3-yl)(mo holino)methanone and has the following structure:
  • an "anti-neoplastic”, “anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition.
  • One class of anticancer agents comprises chemotherapeutic agents.
  • “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • cell proliferation refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
  • co-administration encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time.
  • Coadministration includes simultaneous a ⁇ lministration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • Co-administered agents may be in the same formulation.
  • Co-administered agents may also be in different formulations.
  • a "therapeutic effect,” as used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • salts refers to salts derived from a variety of organic and inorganic counter ions well known in the art.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids.
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as
  • the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, and sodium, calcium and magnesium salts.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • Signal transduction is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response.
  • a modulator of a signal transduction pathway refers to a compound that modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway.
  • a modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
  • Subject refers to an animal, such as a mammal, for example a human.
  • the methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications.
  • the subject is a mammal, and in some embodiments, the subject is human.
  • w vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject assay.
  • In vitro assays encompass cell-based assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • PI3K Phosphoinositide-3-kinase
  • PI phosphatidylinositol
  • connection of compound name moieties are at the rightmost recited moiety. That is, the substituent name starts with a terminal moiety, continues with any linking moieties, and ends with the linking moiety.
  • heteroarylthio C alkyl has a heteroaryl group connected through a thio sulfur to a Ci ⁇ alkyl radical that connects to the chemical species bearing the substituent.
  • the terminal group is a C3. 8 cycloalkyl group attached to a linking C MO alkyl moiety which is attached to an element L, which is itself connected to the chemical species bearing the substituent.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., Ci-Cio alkyl).
  • a numerical range such as “1 to 10” refers to each integer in the given range; e.g., "1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term "alkyl” where no numerical range is designated.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like.
  • the alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), n-propyl, 1-methylethyl (iso-propyl), «-butyl, M-pentyl,
  • an alkyl group is optionally substituted by one or more of substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
  • halo or halogen refers to fluoro, chloro, bromo, or iodo.
  • haloalkyl refers to an alkyl group substituted with one or more halo groups, for example chloromethyl, 2-bromoethyl, 3-iodopropyl, trifluoromethyl, perfluoropropyl, 8- chlorononyl, and the like.
  • Acyl refers to the groups (alkyl)-C(O)-, (aryl)-C(O)-, (heteroaryl)-C(O)-,
  • it is a Ci-Cio acyl radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocycloalkyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e. three other ring or chain atoms plus carbonyl. If the R radical is heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms.
  • R of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , SR a , -OC(0)-R ⁇ -N(R a ) 2 , -C(0)R a , -C(0)OR a , -OC(0)N(R a ) 2 , -C(0)N(R a ) 2 , -N(R a )C(0)OR a , -N(R a )C(0)R a , -N(R a )C(0)OR a , -N(R a )C(0)R a
  • heterocycloalkylalkyl heteroaryl or heteroarylalkyl.
  • Cycloalkyl refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (i.e., C3-C10 cycloalkyl). Whenever it appears herein, a numerical range such as “3 to 10" refers to each integer in the given range; e.g., "3 to 10 carbon atoms” means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C 3 -C 8 cycloalkyl radical.
  • cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbomyl, and the like.
  • a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a ,SR a , -OC(0)-R a , -N(R a ) 2 , -C(0)R ⁇ -C(0)OR a , -OC(0)N(R a ) 2 , -C(0)N(R a ) 2 , -N(R a )C(0)OR a , -N(R a )C(0)R a , -N(R a )C(0)OR a , -N(R a )C(0)R a
  • heterocycloalkylalkyl heteroaryl or heteroarylalkyl.
  • heteroatom or "ring heteroatom” is meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
  • Heteroalkyl includes optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a numerical range may be given, e.g., C1-C4 heteroalkyl which refers to the chain length in total, which in this example is 1 to 4 atoms long.
  • a -CH2OCH2CH3 radical is referred to as a "C4" heteroalkyl, which includes the heteroatom center in the atom chain length description. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl chain.
  • a heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , SR a ,
  • each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • alkene refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond
  • an "alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • the alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
  • alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (i.e., C 2 -Cio alkenyl).
  • a numerical range such as “2 to 10” refers to each integer in the given range; e.g., "2 to 10 carbon atoms” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.
  • an alkenyl comprises two to eight carbon atoms.
  • an alkenyl comprises two to five carbon atoms (e.g., C2-C5 alkenyl).
  • the alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-l-enyl (i.e., allyl), but-l-enyl, pent-l-enyl, penta-l,4-dienyl, and the like.
  • an alkenyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
  • heterocycloalkylalkyl heteroaryl or heteroarylalkyl.
  • cycloalkenyl refers to a cyclic aliphatic 3 to 8 membered ring structure, optionally substituted with alkyl, hydroxy and halo, having 1 or 2 ethylenic bonds such as methylcyclopropenyl, trifluoromethylcyclopropenyl, cyclopentenyl, cyclohexenyl, 1,4-cyclohexadienyl, and the like.
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (i.e., C 2 -Cio alkynyl).
  • a numerical range such as “2 to 10” refers to each integer in the given range; e.g., "2 to 10 carbon atoms” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms.
  • an alkynyl comprises two to eight carbon atoms.
  • an alkynyl has two to five carbon atoms (e.g., C2-C5 alkynyl).
  • the alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -OR a , SR a , -OC(0)-R a , -N(R a ) 2 , -C(0)R a , -C(0)OR a , -OC(0)N(R a ) 2 , -C(0)N(R a ) 2 ,
  • R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • Amino refers to a -N(R a ) 2 radical group, where each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
  • R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
  • R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroaryl
  • -N(R a ) 2 is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
  • Amide or “amido” refers to a chemical moiety with formula -C(0)N(R) 2 or -
  • NHC(0)R where R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), each of which moiety may itself be optionally substituted. In some embodiments it is a C1-C4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical.
  • the R 2' of - N(R) 2 of the amide may optionally be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring.
  • an amido group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl.
  • An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified.
  • Aromatic or "aryl” refers to an aromatic radical with six to ten ring atoms (e.g., C 6 -Cio aromatic or Ce-Cio aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl).
  • Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals.
  • Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene” to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
  • a numerical range such as “6 to 10” refers to each integer in the given range; e.g., "6 to 10 ring atoms” means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms.
  • the term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of ring atoms) groups.
  • an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
  • Heteroaryl or, alternatively, “heteroaromatic” refers to a 5- to 18-membered aromatic radical (e.g., C5-C13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system.
  • a numerical range such as “5 to 18” refers to each integer in the given range; e.g., "5 to 18 ring atoms” means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms.
  • Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene.
  • heteroaryl refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.
  • the polycyclic heteroaryl group may be fused or non-fused.
  • the heteroatom(s) in the heteroaryl radical is optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • the heteroaryl is attached to the rest of the molecule through any atom of the ring(s).
  • heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1 ,3-benzodioxolyl, benzofuranyl, benzoxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[6][l,4]dioxepinyl, benzo[b][l,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl), benzotlueno[3,2-d]pyrimi
  • a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -OR a , -SR a ,
  • each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • aryl-alkyl arylalkyl
  • arylalkyl arylalkyl
  • aralkyl a group wherein the alkyl chain can be branched or straight chain forming a linking portion with the terminal aryl, as defined above, of the aryl-alkyl moiety.
  • aryl-alkyl groups include, but are not limited to, optionally substituted benzyl, phenethyl, phenpropyl and phenbutyl such as 4- chlorobenzyl, 2,4-dibromobenzyl, 2-methylbenzyl, 2-(3-fluorophenyl)ethyl, 2-(4- methylphenyl)ethyl, 2-(4-(trifluoromethyl)phenyl)ethyl, 2-(2-methoxyphenyl)ethyl, 2-(3- nitropheny ethyl, 2-(2,4-dichlorophenyl)ethyl, 2-(3,5-dimethoxyphenyl)ethyl, 3-phenylpropyl, 3-(3-chlorophenyl)propyli 3-(2-methylphenyl)propyl, 3-(4-methoxyphenyl)propyl, 3-(4- (trifluoromethyl)phenyl)propyl
  • heteroarylalkyl used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heteroaralkyl moiety with the terminal heteroaryl portion, as defined above, for example 3-furylmethyl, thenyl, furfuryl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclyl refers to a four-, five-, six-, or seven-membered ring containing one, two, three or four heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • the four-membered ring has zero double bonds
  • the five-membered ring has zero to two double bonds
  • the six- and seven-membered rings have zero to three double bonds.
  • heterocyclyl also includes bicyclic groups in which the heterocyclyl ring is fused to another monocyclic heterocyclyl group, or a four- to seven-membered aromatic or nonaromatic carbocyclic ring.
  • the heterocyclyl group can be attached to the parent molecular moiety through any carbon atom or nitrogen atom in the group.
  • Heterocycloalkyl refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Whenever it appears herein, a numerical range such as “3 to 18" refers to each integer in the given range; e.g., "3 to 18 ring atoms” means that the heterocycloalkyl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, it is a C5-C10 heterocycloalkyl. In some embodiments, it is a C4-C10
  • heterocycloalkyl In some embodiments, it is a C3-C 10 heterocycloalkyl.
  • the heterocycloalkyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • the heteroatoms in the heterocycloalkyl radical may be optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • the heterocycloalkyl radical is partially or fully saturated.
  • the heterocycloalkyl may be attached to the rest of the molecule through any atom of the ring(s).
  • heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl,
  • 2-oxopiperidinyl 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl,
  • each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl or heteroarylalkyl.
  • Heterocycloalkyl also includes bicyclic ring systems wherein one non-aromatic ring, usually with 3 to 7 ring atoms, contains at least 2 carbon atoms in addition to 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms; and the other ring, usually with 3 to 7 ring atoms, optionally contains 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen and is not aromatic.
  • heterocyclylalkyl refers to the divalent derivative of heterocycloalkyl.
  • alkoxy refers to the group -O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like.
  • Lower alkoxy refers to alkoxy groups containing one to six carbons. In some embodiments, C1-C4 alkyl, is an alkyl group which encompasses both straight and branched chain alkyls of from 1 to 4 carbon atoms.
  • alkylthio includes both branched and straight chain alkyl groups attached to a linking sulfur atom, for example methylthio and the like.
  • oxo refers to an oxygen that is double bonded to a carbon atom.
  • an "oxo” requires a second bond from the atom to which the oxo is attached. Accordingly, it is understood that oxo cannot be substituted onto an aryl or heteroaryl ring, unless it forms part of the aromatic system as a tautomer.
  • a sulfonamido group is optionally substituted by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
  • Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Compounds may be shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of the disclosed compounds and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • the present invention includes all manner of rotamers and conformationally restricted states of an inhibitor of the invention.
  • R ⁇ R", R'" and R" each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1 -3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R 1 , R", R"' and R"" groups when more than one of these groups is present.
  • R* and R" or R" and R m are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring.
  • -NR'R is meant to include, but not be limited to, 1-pyrrolidinyl, 4 piperazinyl, and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF 3 and -CH 2 CF 3 ) and acyl (e.g., -C(0)CH 3 , -C(0)CF 3 , - C(0)CH 2 OCH 3 , and the like).
  • haloalkyl e.g., -CF 3 and -CH 2 CF 3
  • acyl e.g., -C(0)CH 3 , -C(0)CF 3 , - C(0)CH 2 OCH 3 , and the like.
  • exemplary substituents for aryl and heteroaryl groups are varied and are selected from, for example: halogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, -OR', -NR'R", -SR', -halogen, -SiR'R"R" ⁇ -OC(0)R',
  • R', R", R"' and R" are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl.
  • each of the R groups is independently selected as are each R 1 , R", R'" and R"" groups when more than
  • 0-2 in the context of -S(0)(o-2)- are integers of 0, 1, and 2.
  • Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(0)-(CRR') q -U-, wherein T and U are independently -NR-, -0-, - CRR'- or a single bond, and q is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) B-, wherein A and B are independently -CRR'-, -0-, -NR-, -S-, -S(O)-, -S(0)2-, -S(0)2NR'- or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -(CRR') s -X'-(C"R" l )d-, where s and d are independently integers of from 0 to 3, and X* is -0-, -NR'-, -S-, -S(O)-, -S(0) 2 -, or -S(0) 2 NR'-.
  • R, R', R" and R'" are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures wherein hydrogen is replaced by deuterium or tritium, or wherein carbon atom is replaced by 13 C- or l C-enriched carbon are within the scope of this invention.
  • the present invention may include compounds that contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • the present invention may include pharmaceutically acceptable salts and radioactive isomers of compound A.
  • intermittent regimen refers to administration of a pharmaceutically active ingredient, (including but not limited to a PI3Ka inhibitor and/or a taxane) to a subject for at least one day, followed by an intermission, or rest period, of at least one day.
  • a pharmaceutically active ingredient including but not limited to a PI3Ka inhibitor and/or a taxane
  • an intermittent regimen involves administering a pharmaceutically active ingredient for three consecutive days followed by a rest of 4 days in a 7- day treating period.
  • an intermittent regimen involves administering a pharmaceutically active ingredient on three alternating days and not on days in between within a given treating period.
  • an intermittent regimen involves administering a pharmaceutically active ingredient consecutively for at least 2, 3, 4, 5, 6, or more days, followed by an intermission of at least 1, 2, 3, 4, 5, 6 or more days over a given treating period.
  • the pharmaceutically active ingredient can be administered on three alternating days and not administered on the days in between within the 7-day period.
  • PBKct inhibitor a pharmaceutically active ingredient
  • a taxane disclosed herein refers to a ⁇ iministering the more than one ingredient at the same time, or at two different time points that are separated by no more than 2 hours.
  • a PI3Ka inhibitor and a taxane disclosed herein refers to acirninistering the more than one ingredient at two different time points that are separated by more than 2 hours, e.g., about 5 hours, 8 hours, lday, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days or even longer.
  • the term "intermission” refers to a period that is that subsequent to the administration of a particular pharmaceutically active ingredient, (including but not limited to a PI3K.cc inhibitor and/or a taxane) of an intermittent regimen. Intermission refers to a rest period wherein a particular pharmaceutically active ingredient, (including but not limited to a PI3Ka inhibitor and/or a taxane) is not adrninistered for at least one day.
  • a week or “7-day cycle”, as used herein, are used interchangeably. These terms refer to a continuous period of time covering the duration of seven consecutive days. For example, a week can start on Monday and end on the following Sunday, or a 7-day cycle can start on Wednesday and end on the next Tuesday.
  • the PI3Kct inhibitors e.g., compound A and its pharmaceutically acceptable salts and prodrugs
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • the present invention includes all manner of rotamers and conformationally restricted states of the PBKa inhibitor.
  • the PBKa inhibitor of the present invention also include crystalline and amorphous forms of those compounds, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof.
  • Crystal form may be used interchangeably herein, and are meant to include all crystalline and amorphous forms of the compound, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms, as well as mixtures thereof, unless a particular crystalline or amorphous form is referred to.
  • the PI3Ka inhibitor described herein can be optionally contacted with a pharmaceutically acceptable acid to form the corresponding acid addition salts.
  • Pharmaceutically acceptable forms of the compounds recited herein include pharmaceutically acceptable salts, chelates, non-covalent complexes, prodrugs, and mixtures thereof.
  • the PI3Ka inhibitor described herein are in the form of pharmaceutically acceptable salts. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition salts.
  • taxane as used herein to describe the compounds that are derived from the plants of genus Taxus (yews). Synonyms for taxane include but are not limited to, taxoid or taxol derivative. Examples of taxanes include, but are not limited to, docetaxel, paclitaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof.
  • the present invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the taxane is not paclitaxel.
  • the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
  • the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
  • the neoplastic condition is non-small cell lung cancer.
  • the non-small cell lung cancer is squamous non-small cell lung cancer.
  • the non-small cell lung cancer is non-squamous non-small cell lung cancer.
  • the neoplastic condition is small cell lung cancer.
  • the subject has a PIK3CA mutation and/or amplification.
  • the present invention provides a method of enhancing apoptosis in cancer cells comprising administering to the cells simultaneously or sequentially a
  • the administration takes place in vitro. In other embodiments, the administration takes place in vivo.
  • the present invention provides a method of suppressing tumor regrowth in a subject in need thereof administering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3Ka) inhibitor and a taxane.
  • the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the PI3 a inhibitor or the taxane.
  • the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the taxane.
  • the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the PDKa inhibitor or the taxane.
  • the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the PI3 a inhibitor or the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the taxane.
  • a PO a inhibitor and a taxane can be administered sequentially, where the two agents are introduced into a subject at two different time points.
  • the two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, or according to any intermittent regimen schedule disclosed herein.
  • a taxane is administered prior to the administration of the POKa inhibitor.
  • Pre-dosing of taxane may occur at about 1, 5, 8, 10, 12, 24, 36, or 72 hours before administering the PI3Ka inhibitor. Pre-dosing may also take place at about 1, 2, 3, 4, 5, 6, 7, or more days before administering the PI3Ka inhibitor.
  • Pre-dosing of taxane may provide a synergistic therapeutic effect as compared to simultaneous administration of the two agents or administration of either agent alone in down regulating PI3Ka signaling, increasing apoptosis of cancer cells size and/or reducing tumor inhibition of tumor regrowth.
  • the PI3 a inhibitor and the taxane are administered simultaneously.
  • Simultaneous administration may take the format of co-administration of the two agents in same formulation, or in different formulations but at the same time.
  • the subject methods utilize a therapeutically effective amount of a combination of a PI3Ka inhibitor and a taxane.
  • the combination is sufficient to effect the intended application including but not limited to disease treatment, as defined herein.
  • a therapeutically effective amount of a PI3Ka inhibitor and/or a taxane in combination to effect such treatment.
  • Also contemplated in the subject methods is the use of a sub-therapeutic amount of a PI3Ka inhibitor and/or a taxane in the combination for treating an intended disease condition.
  • the PI3 a inhibitor and taxane individually, though present in sub-therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
  • the therapeutically effective amount of the subject combination of compounds may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the effect of inhibiting PO a pathway may be measured, for example, as a percentage decrease in phosphorylation of a protein chosen from p4EBPl, pS6, and pPRAS40.
  • pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of p4EBPl.
  • phosphorylation of p4EBPl is reduced by at least 60%.
  • pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of pS6.
  • pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of pPRAS40.
  • phosphorylation of pPRAS40 is reduced by at least 60%.
  • pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of p4EBPl, pS6, and pRAS40.
  • pathway inhibition is measured in peripheral blood cells.
  • pathway inhibition is measured in a biopsy, for example a skin biopsy.
  • the present invention provides for the treatment of a disorder mediated by PI3-kinase a (PI3 a), wherein the regimen comprises simultaneous or sequential aciministration of at least one taxane and at least one PI3Kct inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel.
  • the regimen comprises at least one cycle wherein the taxane is administered before the PI3Kot inhibitor in a given cycle and wherein the taxane and the PDKoc inhibitor are not administered on the same day.
  • the cycle comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 25, 30, 40 or more days.
  • the cycle comprises least 5 to 15 days. In some embodiments, the cycle consists of 7 days. In some embodiments the cycle consists of 21 days. In some embodiments, the PI3K.cc inhibitor is administered nine times in a 21 -day cycle. In certain such embodiments, the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17, and 18. In certain such embodiments, the PI3Ka inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18, and 20.
  • the taxane is administered for at least one day followed by an intermission in which the taxane is not administered for at least one day. In some embodiments, the taxane is administered at least once per 7-day cycle. In some embodiments, the taxane is administered at least once per 21 -day cycle. In some embodiments, the taxane is administered twice per 21 -day cycle, such as on Day 1 and Day 8 of a 21 -day cycle. In some embodiments, the taxane is administered once per 21 -day cycle, such as on Day 1. [00130] In some embodiments, the taxane and the PI3-kinase a inhibitor are administered simultaneously.
  • the taxane is administered to the subject prior to adniinistration of the PI3Ka inhibitor. In some embodiments, the taxane is administered to the subject about 1, 5, 8, 10, 12, 24, 36 or 72 hours prior to administration of the PI3 a inhibitor. In some embodiments, taxane is aciministered to the subject about 12 to about 96 hours prior to administration of the PI3Ka inhibitor. In some embodiments, the taxane is administered to the subject about 12 to about 72 hours prior to administration of the PI3 a inhibitor. In some embodiments, the taxane is administered to the subject about 12 to about 36 hours prior to administration of the PDKct inhibitor.
  • the taxane is administered to the subject about 12 hours prior to administration of the PDKct inhibitor. In some embodiments, the taxane is administered to the subject about 18 hours prior to administration of the ⁇ 3 ⁇ inhibitor. In some embodiments, the taxane is administered to the subject about 24 hours prior to administration of the PDKct inhibitor. In some embodiments, the taxane is administered to the subject about 30 hours prior to aclrninistration of the PI3 a inhibitor. In some
  • the taxane is aciministered to the subject about 36 hours prior to administration of the PDKct inhibitor.
  • a given dosing schedule comprises one or more administrations of a PI3Ka inhibitor, wherein at least one administration of a PI3 a inhibitor, such as described herein, may be repeated or cycled on a daily, weekly, biweekly, monthly, bimonthly, annually, semi-annually, or any other period.
  • a repeated dosing schedule or cycle may be repeated for a fixed period of time determined at the start of the schedule; may be terminated, extended, or otherwise adjusted based on a measure of therapeutic effect, such as a level of reduction in the presence of detectable disease tissue (e.g. a reduction of at least 50%, 60%, 70%, 80%, 90%, 95%, 99%, or 100%); or may be terminated, extended, or otherwise adjusted for any other reason as determined by a medical professional.
  • the PI3K inhibitor is administered according to an intermittent regimen. In some embodiments, the PI3Ka inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission.
  • the PDKct inhibitor is administered for at least 1 day, followed by an intermission in which the PDKct inhibitor is not administered for at least 1 day for at least one cycle.
  • the PI3Kct inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PDKct inhibitor is not administered for at least 1 day.
  • the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
  • the PDKa inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
  • the PI3Ka inhibitor is administered for 7, 8, 9, 10, 11, 12, 13, or 14 consecutive days, followed by an intermission where the ⁇ 3 ⁇ inhibitor is not administered for at least 1, 2, 3, 4, 5, 6, or 7 days. In other embodiments, the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3Ka inhibitor is not administered for at least 3, 4, or 5 consecutive days.
  • the ⁇ 3 ⁇ inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission.
  • the PI3Koc inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days for at least one 7-day cycle.
  • the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of at least one day per 7-day cycle.
  • the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle.
  • the PI3Ka inhibitor is administered for 4 consecutive days followed by an intermission of 3 consecutive days for at least one 7-day cycle.
  • the PI3Ka inhibitor is administered for 5 consecutive days followed by an intermission of 2 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 6 consecutive days followed by an intermission of 1 day for at least one 7-day cycle.
  • the PI3Ka inhibitor is administered for at least 1 day for at least one 7- day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 2 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 3 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 4 days for at least one 7-day cycle. In some embodiments, the PI3Kcc inhibitor is administered for at least 5 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 6 days for at least one 7-day cycle.
  • the PI3Ka inhibitor is administered every other day (i.e., 7 dosing days in 2 weeks). In some embodiments, the PI3Ka inhibitor is administered for three non-consecutive days within a 7-day cycle. In some embodiments, the PI3Koc inhibitor is administered on alternate days in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission. For example, the ⁇ 3 ⁇ inhibitor is administered at least 2 times on alternate days within a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered at least 3 times on alternate days within a 7- day cycle. In some embodiments, the PI3 a inhibitor is administered at least 4 times on alternate days within a 7-day cycle.
  • the taxane is administered on Day 1 and the PI3 a inhibitor is administered on the Days 2, 3 and 4 of a 7-day cycle as shown in Figure 7.
  • the taxane is administered on Day 1 and the PI3 a inhibitor is aclministered on Days 3, 4, and 5 of a 7-day cycle. In some embodiments, the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 4, 5 and 6 of a 7-day cycle. In some embodiments, the taxane is administered on Day land the PI3K.cc inhibitor is administered on Days 5, 6, and 7 of a 7-day cycle. In some embodiments, the taxane is a ⁇ lministered on the Day 1 and the PI3Ka inhibitor is administered on Days 2, 4, and 6 of a 7-day cycle as shown in Figure 7. In some embodiments, the taxane is administered on Day 1 and the PI3Koc inhibitor is administered on Days 3, 5, and 7 of a 7- day cycle.
  • the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17 and 18 of a 21 -day cycle as shown in Figure 8A. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 4, 5, 10, 11, 12, 17, 18 and 19 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is aoministered on Days 4, 5, 6, 11, 12, 13, 18, 19 and 20 of a 21 -day cycle. In some
  • the taxane is administered on Days 1 and 8 and the PI3Koc inhibitor is administered on Days 5, 6, 7, 12, 13, 14, 19, 20 and 21 of a 21-day cycle.
  • the taxane is administered on Days 1 and 8 and the PI3Koc inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18 and 20 of a 21-day cycle as shown in Figure 8B. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 5, 7, 10, 12, 14, 17, 19 and 21 of a 21-day cycle.
  • the PI3Ka inhibitor is administered at least once a day (QD) in each of the days that the PI3Ka inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered once a day (QD) in each of the days that the PI3Ka inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is aclministered twice a day (BID) in each of the days that the PI3Ka inhibitor is administered to the subject.
  • QD a day
  • BID twice a day
  • a PI3K inhibitor and/or any additional therapeutic compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day or per week. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In some embodiments, cycles of administering a PI3Ka inhibitor followed by periods of intermission (or rest) are repeated for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, repetition of a dosing cycle comprising administration of a PI3 a inhibitor followed by intermission (or rest) is continued as long as necessary.
  • a PI3Kcc inhibitor of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, 21, or 28 days. In some embodiments, a PI3 a inhibitor of the invention is administered for less than 28, 21, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a PI3Ka inhibitor of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • the amount of the PI3Kcc inhibitor administered herein may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the PI3Ka inhibitor may be administered in any suitable amount. In some embodiments,
  • the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 900 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3 a inhibitor is administered to a subject within a range of about an amount of 900 mg to about 6300 mg of the PI3Koc inhibitor in a 7-day cycle.
  • this inhibitor is administered to a subject at a dosage of about 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, 3000, 3100, 3200, 3300, 3400, 3600, 4500, 5400, or 6300 mg in 7-day cycle.
  • the amount of PI3Ka inhibitor administered in a 7-day cycle is about 300 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 900 mg.
  • the amount of PI3Ka inhibitor aclministered in a 7-day cycle is about 1800 mg. In some embodiments, the amount of PI3Kcc inhibitor administered in a 7-day cycle is about 2700 mg. In some embodiments, the amount of PI3Kct inhibitor administered in a 7-day cycle is about 3600 mg. In some embodiments, the amount of PI3Koc inhibitor administered in a 7-day cycle is about 4500 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 5400 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 6300 mg.
  • a dose (i.e., as applied to a single administration) of the PI3Ka inhibitor is about 100 to about 1200 mg. In some embodiments, a dose (i.e., as applied to a single administration) of the PI3 a inhibitor is about 100 to about 2100 mg. In some embodiments, a dose of the PDKot inhibitor is about 300 to about 1200 mg. In some embodiments, a dose of the PI3 a inhibitor ts about 300 to about 2100 mg. In some embodiments a dose of the PI3 a inhibitor is about 100 mg, about 300 mg, about 600 mg, or about 900 mg.
  • a dose of the PDKot inhibitor is about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 600 mg.
  • a dose of the PI3Ka inhibitor is about 900 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1200 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1 00 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1800 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 2100 mg.
  • a dose of PI3Ka inhibitor is within a range of about 1 mg/kg- 100 mg kg per day, such as about, less than about, or more than about, 1 mg/kg, 2 mg/kg, 3 mg kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg, 40 mg/kg, 45 mg/kg, 50 mg/kg, 60 mg/kg, 70 mg/kg, 80 mg/kg, 90 mg kg, or 100 mg/kg per day.
  • the administration schedule may be repeated according to any regimen according to the invention, including any adrninistration schedule described herein.
  • a dose of PI3Kct inhibitor may be about, at least about, or at most about 0.1 , 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1400, 1425, 1450 mg or mg/kg, or any range derivable therein.
  • such a dose can range from about 30-120 mg/kg (e.g., 40-80 mg/kg such as about 50 or about 60 mg/kg). It is contemplated that a dosage of mg/kg refers to the mg amount of inhibitor per kg of total body weight of the subject. It is contemplated that when multiple doses are given to a patient, the doses may vary in amount or they may be the same.
  • the taxane may be administered in any suitable amount. In some embodiments, the taxane is administered at a dose of any suitable amount in a single administration. In some embodiments, the taxane is administered to a subject at a dose of about 25 mg m 2 to about 100 mg/m 2 in a single administration.
  • the taxane is aclministered at a dose of about 25 mg/m 2 , 26 mg/m 2 , 27 mg/m 2 , 28 mg/m 2 , 29 mg/m 2 , 30 mg/m 2 , 31 mg/m 2 , 32 mg/m 2 , 33 mg/m 2 , 34 mg/m 2 , 35 mg/m 2 , 36 mg/m 2 , 37 mg/m 2 , 38 mg/m 2 , 39 mg/m 2 , 40 mg/m 2 , 41 mg/m 2 , 42 mg/m 2 , 43 mg/m 2 , 44 mg/m 2 , 45 mg/m 2 , 46 mg/m 2 , 47 mg m 2 , 48 mg/m 2 , 49 mg/m 2 , 50 mg m 2 , 51 mg/m 2 , 52 mg/m 2 , 53 mg m 2 , 54 mg/m 2 , 55 mg/m 2 , 56 mg/m 2 , 57 mg/m 2 ,
  • the amount of PI3 a inhibitor or taxane administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • the PI3 a inhibitor and the taxane disclosed herein can be prepared in a dosage form for administration to a subject.
  • the dosage form can be a single capsule, tablet, or pill, or alternatively can be comprised of multiple capsules, tablets or pills (e.g., a single dose of 900 mg may be administered as three dosage forms, such as three tablets, each comprising 300 mg of the PI3Ka inhibitor or the taxane).
  • a dosage form may be administered to a subject once or multiple times per day. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell or tissue being treated, and the subject being treated.
  • Single or multiple administrations e.g. about or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, or more doses
  • the PI3Ka inhibitor is a compound of the following formula:
  • W 1 is CR 3 ;
  • R 1 is hydrogen
  • R 2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamide, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
  • amido of formula -C(0)N(R) 2 or -NHC(0)R wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R) 2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring. In a further embodiment, R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 6-membered ring. In a further embodiment, R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring.
  • R 2 is amino. In a further embodiment, R 2 is NH 2 .
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring and R 2 is amino.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a 6-membered ring and R 2 is amino.
  • R 3 is amido of formula -C(0)N(R) 2 wherein the (R) 2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring and R 2 is NH 2 .
  • the PI3 a inhibitor is a compound with the following structure: or a pharmaceutically acceptable salt thereof.
  • the PI3Ka inhibitor is (6-(2-aminobenzo[d]oxazol-5- yl)imidazo[l ,2-a]pyridin-3-yl)(morpholino)methanone) or compound A.
  • Taxanes are diterpenes produced from the plants of the genus Taxus (yews). Taxanes interfere with cell growth by disrupting with microtubule function. Examples of taxanes include, but are not limited to, docetaxel. Docetaxel, (2R, 3S) ⁇ N-carboxy-3-phenylisoserine, N- tert-butyl ester, 13-ester with 5 ⁇ -20-epoxy- 1 ,2 ⁇ ,4, 7 ⁇ , 10 ⁇ , 13a-hexahydroxytax- 1 - 1 -en-9-one 4- acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as
  • TAXOTERE® Docetaxel is indicated for the treatment of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
  • taxanes contemplated for use include semisynthetic taxanes, including but not limited to, cabazitaxel (taxoid XRP6258 and Jevtana®), ortataxel (SB-T- 101131, IDN5109, and BAY59-8862), larotaxel (XRP9881 and RPR 109881), tesetaxel (DJ- 927), 10-deacetyltaxol.
  • Other examples of taxanes include cephalomannine (Taxol® B).
  • the taxane is docetaxel. In some embodiments, the taxane is docetaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof.
  • the subject methods and regimen are useful for treating any disease conditions, for example neoplastic diseases.
  • the disease condition is a proliferative disorder, such as described herein, including but not limited to cancer.
  • the disease condition is associated with PI3-kinase.
  • PI3-kinase a one of the four isoforms of type I PI3-kinases has been implicated, for example, in a variety of human proliferative disorders, such as cancers.
  • Angiogenesis has been shown to selectively require PI3Ka in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6).
  • Mutations in the gene coding for PI3 a or mutations which lead to upregulation of PI3 a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers.
  • mutations in the gene coding for PI3 a are point mutations clustered within several hotspots in helical and kinase domains, such as E542 , E545 , and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K a mutations, targeting of this pathway provides valuable therapeutic opportunities. While other PI3K isoforms such as PI3K ⁇ or PI3 ⁇ are expressed primarily in hematopoietic cells, PI3K a, along with PI3K ⁇ , is expressed constitutively.
  • Disease conditions associated with PI3-kinase can also be characterized by abnormally high level of activity and/or expression of downstream messengers of mTOR and PI3-kinase.
  • proteins or messengers such as PIP2, PIP3, PDK, Akt, PTEN, PRAS40, GSK-3 , p21 , p27 may be present in abnormal amounts which can be identified by any assays known in the art.
  • PI3K/Akt mTOR pathway Deregulation of the PI3K/Akt mTOR pathway is emerging as a common theme in diverse human diseases and as a consequence drugs that target PI3 a have therapeutic value.
  • the diseases associated with deregulation of PI3 a include, but are not limited to, tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), both of which are caused by mutations in TSC1 or TSC2 tumor suppressors.
  • TSC tuberous sclerosis complex
  • LAM lymphangioleiomyomatosis
  • Inhibition of PI3Kct may help patients with Peutz-Jeghers cancer-prone syndrome caused by the LKB 1 mutation.
  • PI3Ka may also have role in the genesis of sporadic cancers.
  • Akt/mTOR pathway is activated in many cancers.
  • Activated Akt regulates cell survival, cell proliferation and metabolism by phosphorylating proteins such as BAD, FOXO, NF- KB, p21Cipl, p27Kipl, GSK3P and others.
  • Akt might also promote cell growth by phosphorylating TSC2.
  • Akt activation may promote cellular transformation and resistance to apoptosis by collectively promoting growth, proliferation and survival, while inhibiting apoptotic pathways.
  • the subject to be treated is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to the PI3Ka inhibitor and/or taxane.
  • a diagnostic assay to determine the sensitivity of tumor cells to the PI3Ka inhibitor and/or taxane. Any method known in the art that can determine the sensitivity of the tumor cells of a subject to a PI3Ka inhibitor and/or a taxane can be employed.
  • one or more additional anticancer agents or treatments can be co-administered according to a treatment regimen of the invention using the combination of a PI3Ka inhibitor and a taxane, as judged to be appropriate by the adrninistering physician given the prediction of the likely responsiveness of the subject to the combination of the PI3Ka inhibitor and taxane, in combination with any additional circumstances pertaining to the individual subject.
  • Non-limiting examples of such conditions include but are not limited to Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblasts leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar soft part sarcoma, Ameloblastic fibroma, Anal cancer, Anaplastic large cell lymph
  • Carcinoma in situ Carcinoma of the penis, Carcinoma of Unknown Primary Site,
  • Carcinosarcoma Castleman's Disease, Central Nervous System Embryonal Tumor, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Cholangiocarcinoma, Chondroma, Chondrosarcoma, Chordoma, Choriocarcinoma, Choroid plexus papilloma, Chronic
  • Lymphocytic Leukemia Chronic monocytic leukemia, Chronic myelogenous leukemia, Chronic Myeloproliferative Disorder, Chronic neutrophilic leukemia, Clear-cell tumor, Colon Cancer, Colorectal cancer, Craniopharyngioma, Cutaneous T-cell lymphoma, Degos disease,
  • Dermatofibrosarcoma protuberans Dermoid cyst, Desmoplastic small round cell tumor, Diffuse large B cell lymphoma, Dysembryoplastic neuroepithelial tumor, Embryonal carcinoma, Endodermal sinus tumor, Endometrial cancer, Endometrial Uterine Cancer, Endometrioid tumor, Enteropathy-associated T-cell lymphoma, Ependymoblastoma, Ependymoma, Epithelioid sarcoma, Erythroleukemia,Esophageal cancer, Esthesioneuroblastoma, Ewing Family of Tumor, Ewing Family Sarcoma, Ewing's sarcoma, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Extramammary Paget's disease, Fallopian tube cancer, Fetus in fetu, Fibroma, Fibrosarcoma, Follicular lymphoma, Folli
  • Hematological malignancy Hepatocellular carcinoma, Hepatosplenic T-cell lymphoma, Hereditary breast-ovarian cancer syndrome, Hodgkin Lymphoma, Hodgkin's lymphoma,
  • Lymphoepithelioma Lymphoid leukemia, Lymphoma, Macroglobulinemia, Malignant Fibrous
  • Histiocytoma Malignant fibrous histiocytoma, Malignant Fibrous Histiocytoma of Bone,
  • Malignant rhabdoid tumor Malignant triton tumor, MALT lymphoma, Mantle cell lymphoma,
  • Mast cell leukemia Mediastinal germ cell tumor, Mediastinal tumor, Medullary thyroid cancer,
  • Medulloblastoma Medulloblastoma, Medulloblastoma, Medulloepithelioma, Melanoma, Melanoma, Meningioma,
  • Mouth Cancer Mucinous tumor, Multiple Endocrine Neoplasia Syndrome, Multiple myeloma,
  • Mycosis Fungoides Mycosis fungoides, Myelodysplastic Disease, Myelodysplasia Syndromes,
  • Neuroblastoma Neuroblastoma, Neuroblastoma, Neurofibroma, Neuroma, Nodular melanoma, Non-Hodgkin
  • Tumor Ovarian Low Malignant Potential Tumor, Paget's disease of the breast, Pancoast tumor,
  • Pancreatic Cancer Pancreatic cancer, Papillary thyroid cancer, Papillomatosis, Paraganglioma,
  • Neoplasm Pleuropulmonary blastoma, Polyembryoma, Precursor T-lymphoblastic lymphoma,
  • Prostate cancer Pseudomyxoma peritonei, Rectal Cancer, Renal cell carcinoma, Respiratory
  • Tract Carcinoma Involving the NUT Gene on Chromosome 15, Retinoblastoma, Rhabdomyoma,
  • Sarcoma Schwannomatosis, Sebaceous gland carcinoma, Secondary neoplasm, Seminoma, Serous tumor, Sertoli-Leydig cell tumor, Sex cord-stromal tumor, Sezary Syndrome, Signet ring cell carcinoma, Skin Cancer, Small blue round cell tumor, Small cell carcinoma, Small Cell Lung Cancer, Small cell lymphoma, Small intestine cancer, Soft tissue sarcoma,
  • Somatostatinoma Soot wart, Spinal Cord Tumor, Spinal tumor, Splenic marginal zone lymphoma, Squamous cell carcinoma, Stomach cancer, Superficial spreading melanoma, Supratentorial Primitive Neuroectodermal Tumor, Surface epithelial-stromal tumor, Synovial sarcoma, T-cell acute lymphoblastic leukemia, T-cell large granular lymphocyte leukemia, T- cell leukemia, T-cell lymphoma, T-cell prolymphocyte leukemia, Teratoma, Terminal lymphatic cancer, Testicular cancer, Thecoma, Throat Cancer, Thymic Carcinoma, Thymoma, Thyroid cancer, Transitional Cell Cancer of Renal Pelvis and Ureter, Transitional cell carcinoma, Urachal cancer, Urethral cancer, Urogenital neoplasm, Uterine sarcoma, Uveal melanoma, Vaginal Cancer, Verner Morrison syndrome, Verrucous carcinoma,
  • the methods or treatment regimen involve administering to a combination of a PI3Ka inhibitor and a taxane for the treatment of a cancer selected from the group consisting of lung cancer, breast cancer, endometrial cancer, ovarian cancer, bladder cancer, prostate cancer, neuroendocrine cancer, renal cancer, lymphoma, myeloma and leukemia.
  • the methods or treatment regimen involve administering to a combination of a PD a inhibitor and a taxane for the treatment of a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocaricoma, and squamous cell carcinoma of head and neck cancer.
  • NSCLC non-small cell lung cancer
  • the disease or condition mediated by PI3-kinase is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
  • the disease or condition mediated by PI3-kinase is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
  • the cancer is gastrointestinal cancer.
  • the cancer is selected from gastric cancer and gastroesophageal adenocarcinoma.
  • the cancer is gastric cancer.
  • the cancer is gastroesophageal adenocarcinoma.
  • the cancer is gastric cancer. In some embodiments, the cancer is small cell lung cancer. In some embodiments, the cancer is non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is squamous non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer. In some embodimdents, the disorder is non-small cell lung cancer and the subject has been identified with a PIK3CA mutation or amplification.
  • the methods or treatment regimen involves administering a combination of a PI3 a inhibitor and a taxane for the treatment of solid tumors.
  • Solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary.
  • exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
  • Additional exemplary solid tumors include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma,
  • lymphangioendotheliosarcoma synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastrointestinal system carcinomas, colon carcinoma, pancreatic cancer, breast cancer, genitourinary system carcinomas, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms 1 tumor, cervical cancer, endocrine system carcinomas, testicular tumor, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma,
  • medulloblastoma craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma.
  • the methods or treatment regimen of the invention involves administering a combination of a PI3Ka inhibitor and a taxane for the treatment of multiple myeloma and/or Waldenstrom's macroglobulinemia.
  • the methods or treatment regimen involves administering a combination of a PDKct inhibitor and a taxane for the treatment of renal cell carcinoma (also known as RCC or hypernephroma).
  • Renal cell carcinoma is a kidney cancer that originates in the lining of the proximal convoluted tubule.
  • Any known type of renal cell carcinoma may be treated using the treatment regimens of the invention, including clear renal cell carcinoma, papillary renal cell carcinoma, chromophobe renal cell carcinoma and collecting duct carcinoma.
  • Any stage of the disease may be treated using the methods of the invention, including early stage as well as later stages (e.g. metastatic renal cell carcinoma).
  • Certain embodiments contemplate a human subject such as a subject that has been diagnosed as having or being at risk for developing or acquiring a proliferative disorder condition.
  • a non-human subject for example a non-human primate such as a macaque, chimpanzee, gorilla, vervet, orangutan, baboon or other non- human primate, including such non-human subjects that can be known to the art as preclinical models, including preclinical models for inflammatory disorders.
  • non-human subject that is a mammal, for example, a mouse, rat, rabbit, pig, sheep, horse, bovine, goat, gerbil, hamster, guinea pig or other mammal.
  • subject or biological source can be a non- mammalian vertebrate, for example, another higher vertebrate, or an avian, amphibian or reptilian species, or another subject or biological source.
  • a transgenic animal is utilized.
  • a transgenic animal is a non-human animal in which one or more of the cells of the animal includes a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • therapeutic efficacy is measured based on an effect of treating a proliferative disorder, such as cancer.
  • a proliferative disorder such as cancer.
  • therapeutic efficacy of the methods and compositions of the invention, with regard to the treatment of a proliferative disorder e.g.
  • cancer whether benign or malignant
  • vascularization vascularization, the eradication of tumor cells, and/or a reduction in the size of at least one tumor such that a human is treated for the proliferative disorder.
  • Several parameters to be considered in the determination of therapeutic efficacy are discussed herein. The proper combination of parameters for a particular situation can be established by the clinician.
  • the progress of the inventive method in treating cancer e.g., reducing tumor size or eradicating cancerous cells
  • the primary efficacy parameter used to evaluate the treatment of cancer by the inventive method and compositions preferably is a reduction in the size of a tumor.
  • Tumor size can be figured using any suitable technique, such as measurement of dimensions, or estimation of tumor volume using available computer software, such as FreeFlight software developed at Wake Forest University that enables accurate estimation of tumor volume.
  • Tumor size can be determined by tumor visualization using, for example, CT, ultrasound, SPECT, spiral CT, MRI, photographs, and the like.
  • the presence of tumor tissue and tumor size can be determined by gross analysis of the tissue to be resected, and/or by pathological analysis of the resected tissue.
  • tumor size is reduced as a result of the inventive method preferably without significant adverse events in the subject.
  • Adverse events are categorized or "graded" by the Cancer Therapy Evaluation Program (CTEP) of the National Cancer Institute (NCI), with Grade 0 representing minimal adverse side effects and Grade 4 representing the most severe adverse events.
  • CEP Cancer Therapy Evaluation Program
  • NCI National Cancer Institute
  • the NCI toxicity scale (published April 1999) and Common Toxicity Criteria Manual (updated August 1999) is available through the NCI, e.g., through the NCI internet website at www.ctep.info.nih.gov or in the Investigator's Handbook for participants in clinical trials of investigational agents sponsored by the Division of Cancer Treatment and Diagnosis, NCI.
  • the inventive method is associated with minimal adverse events, e.g. Grade 0, Grade 1, or Grade 2 adverse events, as graded by the CTEP/NCI.
  • the growth of a tumor is stabilized (i.e., one or more tumors do not increase more than 1%, 5%, 10%, 15%, or 20% in size, and/or do not metastasize ) as a result of the inventive method and compositions.
  • Such stabilization may be evidenced by a longer period of stable disease as characterized by the RECIST guidelines.
  • a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more weeks.
  • a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more months.
  • a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more years.
  • the inventive method reduces the size of a tumor at least about 5%
  • tumor size is reduced at least about 30% (e.g., at least about 35%, 40%, 45%, 50%, 55%, 60%, or 65%). Even more preferably, tumor size is reduced at least about 70% (e.g., at least about 75%, 80%, 85%, 90%, or 95%). In some embodiments, tumor regrowth is not detected to at least 20, 25, 30, 40, 45, 50,
  • a subject remains tumor free (e.g. in remission) for at least about 1, 2, 3, 4, 5, 6, 7,
  • a subject remains tumor free for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more months following treatment. In some embodiments, no detectable amount of tumor is found in the subject for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more years after the treatment.
  • the efficacy of the inventive method in reducing tumor size can be determined by measuring the percentage of resected tissue that is necrotic (i.e., dead).
  • a treatment is therapeutically effective if the necrosis percentage of the resected tissue is greater than about 20% (e.g., at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%), more preferably about 90% or greater (e.g., about 90%, 95%, or 100%).
  • the necrosis percentage of the resected tissue is 100%, that is, no tumor tissue is present or detectable.
  • a number of secondary parameters can be employed to determine the efficacy of the inventive method.
  • secondary parameters include, but are not limited to, detection of new tumors, detection of tumor antigens or markers (e.g., CEA, PSA, or CA-125), biopsy, surgical downstaging (i.e., conversion of the surgical stage of a tumor from unresectable to resectable), PET scans, survival, disease progression-free survival, time to disease progression, quality of life assessments such as the Clinical Benefit Response Assessment, and the like, all of which can point to the overall progression (or regression) of cancer in a human.
  • Biopsy is particularly useful in detecting the eradication of cancerous cells within a tissue.
  • Radioimmunodetection is used to locate and stage tumors using serum levels of markers (antigens) produced by and/or associated with tumors ("tumor markers” or “tumor-associated antigens”), and can be useful as a pre-treatment diagnostic predicate, a post-treatment diagnostic indicator of recurrence, and a post-treatment indicator of therapeutic efficacy.
  • tumor markers or tumor-associated antigens that can be evaluated as indicators of therapeutic efficacy include, but are not limited to, carcinembryonic antigen (CEA) prostate-specific antigen (PSA), CA-125, CA19-9, ganglioside molecules (e.g., GM2, GD2, and GD3), MART-1, heat shock proteins (e.g., gp96), sialyl Tn (STn), tyrosinase, MUC-1, HER-2/neu, c-erb-B2, KSA, PSMA, p53, RAS, EGF-R, VEGF, MAGE, and gplOO.
  • CCA carcinembryonic antigen
  • PSA prostate-specific antigen
  • CA-125 CA19-9
  • CA19-9 ganglioside molecules
  • ganglioside molecules e.g., GM2, GD2, and GD3
  • MART-1 heat shock proteins
  • STn sialyl Tn
  • STn sialy
  • the treatment of cancer in a human patient is evidenced by one or more of the following results: (a) the complete disappearance of a tumor (i.e., a complete response), (b) about a 25% to about a 50% reduction in the size of a tumor for at least four weeks after completion of the therapeutic period as compared to the size of the tumor before treatment, (c) at least about a 50% reduction in the size of a tumor for at least four weeks after completion of the therapeutic period as compared to the size of the tumor before the therapeutic period, (d) at least a 2% decrease (e.g., about a 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% decrease) in a specific tumor-associated antigen level at about 4-12 weeks after completion of the therapeutic period as compared to the tumor-associated antigen level before the therapeutic period or (e) a longer period of stable disease, for example longer by 1, 2, 3, 4, or 5 months.
  • any decrease in the tumor-associated antigen level is evidence of treatment of a cancer in a patient by the inventive method.
  • treatment can be evidenced by at least a 10% decrease in the CA19- 9 tumor-associated antigen level at 4-12 weeks after completion of the therapeutic period as compared to the CA19-9 level before the therapeutic period.
  • treatment can be evidenced by at least a 10% decrease in the CEA tumor-associated antigen level at 4-12 weeks after completion of the therapeutic period as compared to the CEA level before the therapeutic period.
  • the therapeutic benefit of the treatment in accordance with the invention can be evidenced in terms of pain intensity, analgesic consumption, and/or the Karnofsky Performance Scale score.
  • the Karnofsky Performance Scale allows patients to be classified according to their functional impairment.
  • the Karnofsky Performance Scale is scored from 0-100. In general, a lower Karnofsky score is predictive of a poor prognosis for survival.
  • the treatment of cancer in a human patient is evidenced by (a) at least a 50% decrease (e.g., at least a 60%, 70%, 80%, 90%, or 100% decrease) in pain intensity reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of treatment, as compared to the pain intensity reported by the patient before treatment, (b) at least a 50% decrease (e.g., at least a 60%, 70%, 80%, 90%, or 100% decrease) in analgesic consumption reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of treatment as compared to the analgesic consumption reported by the patient before treatment, and/or (c) at least a 20 point increase (e.g., at least a 30 point, 50 point, 70 point, or 90 point increase) in the Karnofsky Performance Scale score reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of the therapeutic period as compared to the Karnofsky Performance Scale score
  • a 50% decrease e
  • a proliferative disorder e.g. cancer, whether benign or malignant
  • a human patient desirably is evidenced by one or more (in any combination) of the foregoing results, although alternative or additional results of the referenced tests and/or other tests can evidence treatment efficacy.
  • Detection, monitoring, and rating of various cancers in a human are further described in Cancer Facts and Figures 2001, American Cancer Society, New York, N.Y., and International Patent Application WO 01/24684. Accordingly, a clinician can use standard tests to determine the efficacy of the various embodiments of the inventive method in treating cancer. However, in addition to tumor size and spread, the clinician also may consider quality of life and survival of the subject in evaluating efficacy of treatment.
  • administration of a taxane at least 24 hours prior to the administration of a PI3 a inhibitor provides improved therapeutic efficacy over a treatment wherein the taxane and PI3 a inhibitor are administered simultaneously.
  • Improved efficacy may be measured using any method known in the art, including but not limited to those described herein.
  • the improved therapeutic efficacy is an improvement of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, 110%, 120%, 150%, 200%, 300%, 400%, 500%, 600%, 700%, 1000%, 10000% or more, using an appropriate measure (e.g., inhibiting PI3 a signaling tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival).
  • an appropriate measure e.g., inhibiting PI3 a signaling tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival.
  • Improved efficacy may also be expressed as fold improvement, such as at least about 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60- fold, 70-fold, 80-fold, 90-fold, 100-fold, 1000-fold, 10000-fold, or more, using an appropriate measure (e.g. tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival).
  • fold improvement such as at least about 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60- fold, 70-fold, 80-fold, 90-fold, 100-fold, 1000-fold, 10000-fold, or more, using an appropriate measure (e.g. tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival).
  • the invention provides, in one aspect, a combination treatment utilizing a PI3 a inhibitor and a taxane.
  • the therapeutic agents (including compounds) that are provided for use in the combination therapies of the invention can be administered simultaneously or separately.
  • This administration in combination includes, for example, simultaneous administration of two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration.
  • multiple therapeutic agents can be formulated together in the same dosage form and administered simultaneously.
  • multiple therapeutic agents can be simultaneously administered, wherein both the agents are present in separate formulations.
  • a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa.
  • a compound of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart.
  • the term "combination treatments" also embraces the administration of the therapeutic agents as described herein in further combination with other biologically active compounds or ingredients and non-drug therapies (e.g., surgery or radiation treatment).
  • Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action.
  • An effective amount of an inhibitor of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • Sequential or substantially simultaneous administration of each inhibitor or therapeutic agent can be effected by any appropriate route as noted above and including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues.
  • the therapeutic agents can be administered by the same route or by different routes.
  • a first therapeutic agent of the combination e.g. in some embodiments a taxane
  • the other therapeutic agent a PI3 a inhibitor
  • all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
  • adrninistration of the compounds of the invention can be effected in one dose, continuously or intermittently throughout the course of treatment.
  • Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell or tissue being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • each compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg kg/day, in single or divided doses.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g., by dividing such larger doses into several small doses for administration throughout the day.
  • a combination treatment of the invention is administered in a single dose comprising at least one PD ct inhibitor and at least one taxane.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • other routes may be used as appropriate.
  • a single dose of a combination treatment of the invention may also be used for treatment of an acute condition.
  • the subject pharmaceutical compositions can be administered as a combination of a PI3Ka inhibitor and a taxane, or in further combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions.
  • the subject combinations and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
  • a pharmaceutical composition of the invention typically contains an active ingredient (e.g., a compound) of the present invention or a pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including but not limited to inert solid diluents and fillers, diluents, sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • compositions for oral administration are provided.
  • the invention provides a pharmaceutical composition for oral administration containing a compound of the invention, and a pharmaceutical excipient suitable for oral administration.
  • the invention provides a solid pharmaceutical composition for oral administration containing: (i) a compound which is a PI3Ka inhibitor; (ii) a second compound which is a taxane; and (iii) a pharmaceutical excipient suitable for oral
  • composition further contains: (iv) a third agent or even a fourth agent.
  • each compound or agent is present in a
  • one or more compounds or agents is present in a sub-therapeutic amount, and the compounds or agents act synergistically to provide a therapeutically effective pharmaceutical composition.
  • the invention provides for a pharmaceutical composition comprising a combination of a PI3Ka inhibitor and a taxane.
  • the PI3-kinase a inhibitor and the taxane can be packaged as a single oral dosage form.
  • the PI3Ka inhibitor and the taxane inhibitor can be packaged as separate dosage forms, such as a tablet.
  • the present invention provides an oral dosage form comprising 100 mg to 1.5g of an inhibitor of the invention.
  • the oral dosage form can be a tablet, formulated in form of liquid, in immediate or sustained release format.
  • the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption.
  • Pharmaceutical compositions of the invention suitable for oral adniinistration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or nonaqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion.
  • Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients.
  • compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds.
  • water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and or storage is expected.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits.
  • suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
  • An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier can take a wide variety of forms depending on the form of preparation desired for administration.
  • any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro- crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose.
  • suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, com starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
  • natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyr
  • suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art. About 0.5 to about 15 weight percent of disintegrant, or about 1 to about
  • Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, hydroxypropyl cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
  • Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, colloidal silicon dioxide, or mixtures thereof.
  • a lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
  • the active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • the tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • a suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
  • HLB hydrophilic-lipophilic balance
  • HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.
  • lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10.
  • HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids,
  • oligopeptides, and polypeptides lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di- glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • ionic surfactants include, by way of example:
  • lecithins lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di- glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Ionic surfactants may be the ionized forms of lecithin, lysolecithin,
  • phosphatidylcholine phosphatidylethanolamine
  • phosphatidylglycerol phosphatidic acid
  • phosphatidylserine lysophosphatidylcholine
  • lysophosphatidylethanolamine phosphatidylethanolamine
  • lysophosphatidylglycerol lysophosphatidic acid, lysophosphatidylserine, PEG- phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate, caprate, laurate, myristate, palmitate, oleate, ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl sulfate, docusate, lauroyl carnitines, palmitoyl carnitines, myristoyl carn
  • Hydrophilic non-ionic surfactants may include, but are not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene stearoyl
  • hydrophilic-non-ionic surfactants include, without limitation, PEG- 10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG- 15 stearate, PEG-32 distearate, PEG-40 stearate, PEG- 100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl o
  • Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil- soluble vitarnms/vitamin derivatives; and mixtures thereof.
  • preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection.
  • a solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
  • solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol,
  • alcohols and polyols such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol,
  • N-alkylpyrrolidone N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide and polyvinylpyrrolidone
  • esters such as ethyl propionate, tributylcitrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, ⁇ -caprolactone and isomers thereof, ⁇ -valerolactone and isomers thereof, ⁇ -butyrolactone and isomers thereof; and other solubilizers known in the art, such as dimethyl acetamide, dimethyl isosorbide, N-methyl pyrrolidones, monooctanoin
  • Mixtures of solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone,
  • N-hydroxyethylpyrrolidone polyvinylpyrrolidone, hydroxypropyl methylcellulose
  • hydroxypropyl cyclodextrins ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide.
  • Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
  • the amount of solubilizer that can be included is not particularly limited.
  • the amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art.
  • the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients.
  • very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less.
  • the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
  • the composition can further include one or more pharmaceutically acceptable additives and excipients.
  • additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons.
  • pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like.
  • bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like.
  • a pharmaceutically acceptable acid such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids
  • Salts of polyprotic acids such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used.
  • the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like.
  • Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
  • Suitable acids are pharmaceutically acceptable organic or inorganic acids.
  • suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like.
  • suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid and the like.
  • compositions for injection are provided.
  • the invention provides a pharmaceutical composition for injection containing at least one compound of the present invention and a pharmaceutical excipient suitable for injection.
  • a pharmaceutical composition for injection comprising at least one PI3 ct inhibitor and at least one taxane.
  • pharmaceutical compositions comprising a PI3Ka inhibitor, and pharmaceutical compositions comprising a taxane, where the PI3Ka inhibitor is administered separately or together with the taxane.
  • the PI3 a inhibitor and the taxane may be formulated separately, and may further include a third therapeutic agent. Components and amounts of agents in the compositions are as described herein.
  • Aqueous solutions in saline are also conventionally used for injection.
  • Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions for topical (e.g.. transdermal) delivery containing at least one compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery.
  • a pharmaceutical composition for topical delivery comprising at least one PI3Ka inhibitor and at least one taxane.
  • pharmaceutical compositions for topical delivery comprising a PI3 a inhibitor, and pharmaceutical compositions for topical delivery comprising a taxane, where the PB a inhibitor is administered separately or together with the taxane.
  • the PI3 a inhibitor and the taxane may be formulated separately, and may further include a third therapeutic agent.
  • compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical acUninistration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions.
  • DMSO dimethylsulfoxide
  • carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients.
  • a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
  • compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • suitable solid or gel phase carriers or excipients which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin.
  • penetration-enhancing molecules known to those trained in the art of topical formulation.
  • humectants e.g., urea
  • glycols e.g., propylene glycol
  • alcohols e.g., ethanol
  • fatty acids e.g., oleic acid
  • surfactants e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.g., isopropyl myristate and sodium lauryl sulfate
  • pyrrolidones e.glycerol monolaurate, sulfoxides, terpenes (e.g., menthol)
  • amines amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • transdermal delivery devices patches
  • Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts, either with or without another agent.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Pharmaceutical compositions for inhalation are well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139.
  • Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine.
  • Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • a pharmaceutical composition for respiratory delivery is provided comprising at least one PI3Ka inhibitor and a taxane. Also provided are
  • compositions for respiratory delivery comprising a PI3 a inhibitor
  • pharmaceutical compositions for respiratory delivery comprising a taxane
  • the PI3Ka inhibitor is administered separately or together with the taxane.
  • Compositions comprising a PI3 a inhibitor and a taxane may be formulated separately, and may further include a third therapeutic agent.
  • compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration.
  • Preparations for such pharmaceutical compositions are well-known in the art. See, e.g., See, e.g., Anderson, Philip O.; noben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, New York, 1990; atzung, ed., Basic and Clinical
  • Administration of the compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also abe administered intraadiposally or intrathecally.
  • a compound of the invention is administered in a single dose.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • other routes may be used as appropriate.
  • a single dose of a compound of the invention may also be used for treatment of an acute condition.
  • a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • Administration of the compounds of the invention may continue as long as necessary.
  • a compound of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days.
  • a compound of the invention is aa'ministered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day.
  • a compound of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • the compounds of the invention may be aaministered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
  • the subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • kits include the compounds of the present invention as described herein, in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like.
  • kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider.
  • Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • the kit may further contain another agent.
  • the compounds of the present invention and the agent are provided as separate compositions in separate containers within the kit. In some embodiments, the compound of the present invention and the agent are provided as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like) are known in the art and may be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • the present invention also provides methods for further combination therapies in which, in addition to an PDKct inhibitor and a taxane, one or more agents known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes is used or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • such therapy includes but is not limited to the combination of the composition comprising at least one PBKa inhibitor and at least one taxane, as described herein, with one or more additional therapeutic agents such as anticancer agents,
  • chemotherapeutic agents therapeutic antibodies, and radiation treatment, to provide, where desired, a synergistic or additive therapeutic effect.
  • Pathways that may be targeted by administering another agent include, but are not limited to, MAP kinase, Akt, NFkB, WNT, RAS/ RAF7MEK/ERK, JNK/SAP , p38 MAPK, Src Family Kinases, JAK/STAT and/or PKC signaling pathways.
  • Other agents may target one or more members of one or more signaling pathways.
  • NFkB nuclear factor-kappaB
  • Representative members of the nuclear factor-kappaB (NFkB) pathway include but are not limited to RelA (p65), RelB, c-Rel, p50/pl05 (NF- ⁇ 1), p52/p 100 (NF-KB2), IkB, and IkB kinase.
  • Non-limiting examples of receptor tyrosine kinases that are members of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway that may be targeted by one or more agents include FLT3 LIGAND, EGFR, IGF-1R, HER2/neu, VEGFR, and PDGFR.
  • Downstream members of the PI3K/AKT pathway that may be targeted by agents according to the methods of the invention include, but are not limited to, forkhead box O transcription factors, Bad, GSK-3P, I-KB, mTOR, MDM-2, and S6 ribosomal subunit.
  • This invention further relates to a method for using the compounds or pharmaceutical composition in combination with other tumor treatment approaches, including surgery, ionizing radiation, photodynamic therapy, or implants, e.g., with corticosteroids, hormones, or used as radiosensitizers.
  • One such approach may be, for example, radiation therapy in inhibiting abnormal cell growth or treating the proliferative disorder in the mammal.
  • Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein.
  • the administration of the compounds of the invention in this combination therapy can be determined as described herein.
  • Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy.
  • brachytherapy refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g., At-211, 1-131, 1-125, Y-90,
  • Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids.
  • the radiation source can be a radionuclide, such as 1-125, 1-131,
  • the radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90.
  • the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
  • the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal the combination of a taxane and a PI3 a inhibitor of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which combined amounts are effective in sensitizing abnormal cells to treatment with radiation.
  • the amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
  • Photodynamic therapy includes therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers.
  • photodynamic therapy include treatment with compounds, such as e.g., VISUDYNE and porfuner sodium.
  • Angiostatic steroids include compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortexolone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids include compounds, such as e.g., fluocinolone and dexamethasone.
  • Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • NCI-H1048 cells were plated in 6-well plates with 750,000 cells/well in 1ml of HITES growth media supplemented with 5% of fetal bovine serum (FBS) and allowed to adhere overnight. Cells were treated with 3 ⁇ compound A, lOnM docetaxel or ⁇ , DMSO for controls and allowed to incubate for 2 hr, 24 hr, or 48 hr. After the timecourse, media was removed by aspiration and cells were washed with 1 mL ice cold DPBS.
  • FBS fetal bovine serum
  • NCI-H1048 tumors approximately 400 -500 mm 3 in size were collected at 2, 24 and 48 h post last dose and frozen in liquid nitrogen. Tumor chunks were homogenized in 700 Lof M- PER lysis buffer supplemented with 25 ⁇ NaF, 1 ⁇ NaOrtho Vanadate, 25 ⁇ ⁇ -GP, and protease inhibitor cocktail tablets. The supernatants were assayed for protein concentration using a BCA Protein Assay kit (Thermo Scientific).
  • Blots were washed with TBST and incubated with fluorescently-labeled secondary antibody AlexaFluor 680 Goat Anti-Rabbit IgG (H+L) (Life Technologies) for 1 hr at RT.
  • Membranes were imaged using Odyssey LI-COR Infrared Imager /scanner) (LI-COR Inc). Li-cor Odyssey Software 2.1 was used to quantitate the proteins/PD markers on the Western blots.
  • the objective of the study was to determine the pharmacodynamic effect of docetaxel in Hs746T gastric xenograft tumor samples.
  • Hs746T tumors tumors approximately 600 mm 3 in size were collected at 2, 7, 24 and 48 h post last dose and frozen in liquid nitrogen. Tumor chunks were homogenized in ⁇ 800 ⁇ M-PER lysis buffer supplemented with 25 ⁇ NaF, 1 ⁇ NaOrtho Vanadate, 25 ⁇ ⁇ - GP, and protease inhibitor cocktail tablets. The supernatants were assayed for protein concentration using a BCA Protein Assay kit (Thermo Scientific).
  • Blots were washed with TBST and incubated with fluorescently-labeled secondary antibody AlexaFluor 680 Goat Anti-Rabbit IgG (H+L) (Life Technologies) for 1 hr at RT.
  • Membranes were imaged using Odyssey LI-COR Infrared Imager /scanner) (LI-COR Inc). Li-cor Odyssey Software 2.1 was used to quantitate the proteins/PD markers on the Western blots.
  • Compound A was prepared according to methods disclosed in WO 2011/022439 and WO 2013/071272. Compound A was formulated in 100% PEG400 and stored at approximately 25 °C, shielded from light. The control/vehicle used in this study was 100% PEG400 + 0.9% saline.
  • NCI-H1048 cells were grown in HITES Medium supplemented with 5% fetal bovine serum (FBS) until approximately 80% confluence was reached. Prior to injection, the cells were detached with Trypsin (Invitrogen), washed twice with phosphate buffered saline (PBS), and re- suspended in KITES media (Invitrogen,) without supplements. NCI-H1048 cells were resuspended to a final concentration of 4.0 x 10 7 cells/mL in HITES media without supplements, with 50% Matrigel (BD Biosciences).
  • FBS fetal bovine serum
  • vehicle 100% PEG400 + 0.9% saline
  • compound A at 140 mg kg on QDX3 schedule via oral gavage, PO
  • docetaxel at 5mg/kg, on QW schedule, intravenously IV
  • TGI tumor growth inhibition
  • BW percent body weight
  • AAUC change in areas under the tumor volume-versus-time curves
  • BIW twice weekly
  • IP IP
  • aDose volumes for PO and IP administration were 5 mL kg body weight.
  • Dose volume for IV administation was 10 mL/kg body
  • bTGI values were calculated on Day 21 post treatment initiation.
  • dAAUC Statistical analysis was performed with a linear mixed effects regresssion model. A p value of ⁇ 0.05 was considered.
  • Figures 5A and 5B show the increased apoptosis observed in vivo in NCI-H1048 when pre-dosing docetaxel in combination with compound A.
  • Figure 5A shows quantified Cleaved lamin A and
  • Figure 5B shows quantified cleaved PARP.
  • Figures 6A and 6B show the effect that pre-dosing docetaxel in combination with compound A has on in vivo antitumor activity compared to simultaneous combination dosing in NCI-H1048 cells.
  • Figure 6A shows the effect of pre-dosing docetaxel at low clinically relevant doses
  • Figure 6B shows the effect of pre-dosing docetaxel at 10 mg/kg.
  • Pre-dosing docetaxel in combination with compound A results in improved or equal antitumor activity compared to simultaneous combination dosing.
  • Figure 6A shows that the pre- dosing effect of docetaxel is more pronounced at low clinically relevant doses for docetaxel.
  • Figure 7 shows the effect that pre-dosing docetaxel in combination with compound A has on enhanced tumor growth delay (compound A: 140 mg/kg QD3; docetaxel: 10 mg/kg QW).
  • Figure 7 shows the effect on NCI-H1048 cells where tumor regrowth is suppressed for at least 20, 30 or even 40 days.
  • MTV mean tumor volume
  • mice Body weight and the tumor growth were monitored twice a week. On day 1 mice began treatment with assigned test materials, either vehicle (100% PEG400 + 0.9% saline), compound A (at 140 mg/kg on QD x3 schedule via oral gavage, PO), docetaxel (at 10 mg/kg, on QW schedule, intravenously IV), or a combination of compound A with docetaxel over a 21 day treatment period. The percentage of tumor growth inhibition (TGI) and percent body weight (BW) change were determined on Day 21. Statistical comparisons of tumor growth between the treatment and vehicle groups were conducted using a linear mixed effects regression analysis on the change in area under the tumor volume (TV)-versus- time curves (AAUC) values (see Table 3 for details). All p values less than 0.05 were considered significant. Table 3: Study Design and Noteworthy Findings for Compound A and Docetaxel in GA0098 model
  • Docetaxel 10 rV/(QWx3) Female/8 Mus TGI 48.20% days 1, 8, 15 musculu AAUC 32.2;
  • AAUC change in areas under the tumor volume-versus-time curves; BWL body wieght loss; a TGI values were calculated on Day 21 post treatment initiation.
  • Example 4 Compound A with or without Docetaxel in Participants with Locally Advanced or Metastatic Non-small Cell Lung Cancer
  • the purpose of this study is to determine the recommended phase 2 dose (RP2D) of Compound A when administered in combination with docetaxel in patients with non-small cell lung cancer (NSCLC) and to evaluate efficacy, safety, and tolerability of Compound A administered alone and in combination with docetaxel at the RP2D in patients with locally advanced or metastatic non-small cell lung cancer.
  • This study consists of 2 phases: Phase lb - dose escalation phase; Phase 2 - expansion phase.
  • Phase lb Approximately 15 patients with NSCLC who have been treated with multiple prior lines of therapies are enrolled for Phase lb. The participants receive docetaxel (36 mg/m 2 ) intravenous (IV) and Compound A tablets, orally administered, once daily in 21 -day dosing cycles. Compound A dose is escalated until recommended Phase 2 dose (RP2D) is determined. Up to 140 adults with locally advanced or metastatic NSCLC refractory or resistant to 1 prior line platinum-based, non-docetaxel containing systemic chemotherapy are enrolled for Phase 2 (dose expansion). Phase 2 expansion study uses a sequential, multistage Bayesian adaptive design and consists of up to 3 parts evaluating the following patient populations:
  • Part 1 NSCLC (inclusive of both squamous and nonsquamous histology and all PIK3CA genotypes).
  • Part 2 Histology-specific NSCLC (either squamous or nonsquamous).
  • Part 3 Histology- and genotype-specific NSCLC (PD 3CA MUT/AMP positive squamous NSCLC if squamous histology is selected in Part 2).
  • Each part of the adaptive Phase 2 study is designed as a stand-alone, randomized study evaluating Progression-Free Survival (PFS) as the primary efficacy measure in a total of 60 patients between the 2 treatment arms: Compound A plus docetaxel versus docetaxel alone.
  • PFS Progression-Free Survival
  • An event-driven analysis of PFS is performed after each part of the Phase 2 study. On the basis of the PFS analysis of the preceding part of the study, the study may be stopped for efficacy or futility, or proceed to the next part.
  • PFS in Phase 2 is is evaluated for approximately 6 months after last dose of study drug.
  • PFS is defined as the time from the date of randomization to the date of first documentation of PD or death due to any cause, whichever occurs first.
  • Study drug is administered in 21 -day dosing cycles. During each phase of the study, patients are treated with a maximum of 9 cycles of either docetaxel alone or docetaxel plus Compound A. Patients treated with docetaxel plus Compound A may continue to receive Compound A monotherapy until progression of disease, occurrence of unacceptable toxicities or death. The maximum duration of treatment for patients is 12 months unless it is determined that a patient would derive benefit from continued treatment beyond 12 months. Patients are followed after discontinuation of study drug to collect PFS and OS data. Patients may withdraw from therapy at any time. The overall time to participate in this study is up to 24 months.
  • DLT Dose-Limiting Toxicities
  • MTD Maximum Tolerated Dose (MTD) in Phase lb is evaluated for approximately 9 months.
  • MTD is defined as the highest dose level of Compound A when administered with weekly docetaxel at which no more than 1 of the 6 patients experiences a DLT during the first cycle (21 days) of therapy in Phase lb.
  • RP2D Phase 2 Dose in Phase lb
  • Compound A plus docetaxel is evaluated for approximately 9 months.
  • RP2D is determined in Phase lb based on safety, tolerability, PK and other observations in both Cycle 1 and Cycle 2 and beyond. The RP2D does not exceed the MTD.
  • AE adverse events
  • An Adverse Event is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign
  • a serious adverse event is any untoward medical occurrence or effect that at any dose results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability / incapacity, is a congenital anomaly / birth defect or is medically important due to other reasons than the above mentioned criteria.
  • Response Rate is assessed approximately 12 months. Response rate is defined as complete response + partial response.
  • Disease Control Rate is assessed at the end of the Cycle 2 , 4, and 6, and then every 3 cycles thereafter until the patient discontinues study drug due to disease progression, unacceptable toxicity, or death (approximately 12 months).
  • Disease control rate is defined as complete response [CR] + partial response [PR] +stable disease [SD].
  • Duration of Response is assessed from the date of first documented response up to the first documentation of progression of disease (approximately 12 months).
  • the duration of response is defined as the time from the date of first documentation of a response to the date of first documentation of progression of disease.
  • Time to Progression is assessed from the date of randomization to the date of first documentation of progressive disease (approximately 24 months). Time to progression is defined as the time from the date of randomization to the date of first documentation of progression of disease.
  • C max Single dose Maximum (peak) plasma concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. Maximum observed plasma concentration
  • (Cmax) is the peak plasma concentration of a drug after administration, obtained directly from the plasma concentration-time curve.
  • T max single-dose first time of occurrence of maximum (peak) concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose.
  • T ma x is defined as time to reach the maximum plasma concentration (C max ).
  • AUC,a Area under the concentration time curve from time 0 to the next dose in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose.
  • AUC,a U is defined as area under the plasma concentration versus time curve from zero to next dose.
  • AUC(iast) Area Under the Plasma Concentration-Time Curve From Time 0 to the Time of the Last Quantifiable Concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose.
  • AUC ( i aS i ) is defined as area under the plasma concentration versus time curve from zero to the time of the last quantifiable concentration.
  • CL F Apparent total body clearance in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. CL/F is apparent clearance of the drug after extravascular administration.
  • ⁇ ' Terminal disposition half-life in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. Ti /2 is the time required for half of the drug to be eliminated from the plasma.
  • Compound A plasma concentrations when administered 1 day after docetaxel is 1 day post docetaxel dose. The plasma concentration of Compound A was evaluated one day following docetaxel dosing.
  • the age Eligible for the study is 18 Years and older. Both male and female genders are eligible for the study.
  • Phase 2 has archived or fresh tumor biopsy samples obtained during screening sufficient for genotyping. Has adequate organ function, before the first dose of study drug. Has Eastern Cooperative Oncology Group (ECOG) Performance Status of 0 or 1.
  • ECG Eastern Cooperative Oncology Group
  • Male participants agree to practice effective barrier contraception during the entire study treatment period and through 30 days after the last dose of study drug, or agree to practice true abstinence.
  • Previous treatment with a PI3 or AKT inhibitor Prior cancer therapy or other investigational therapy within 2 weeks before the first administration of study drug or failed to recover from the reversible effects of prior anticancer therapies. For prior therapies with a half-life longer than 3 days, the interval must be at least 28 days before the first administration of study drug, and the patient must have documented progressive disease.
  • Has taken strong inhibitors or strong inducers of CYP3A4 within 14 days before the first dose of study drug.
  • Has taken histamine-H2 receptor antagonists and/or neutralizing antacids within 24 hours before the first administration of study drug.
  • Fridericia's corrected QT interval > 475 milliseconds (msec) (males) or > 450 msec (females) on a 12-lead
  • ECG during the Screening period or abnormalities on 12-lead ECG including, but not limited to, changes in rhythm and intervals that in the opinion of the investigator are considered to be clinically significant.
  • Has active secondary malignancy that requires treatment. Has any serious medical or psychiatric illness, including drug or alcohol abuse.
  • Example 5 Compound A with or without Docetaxel in Participants with Locallv Advanced and Metastatic Gastric or Gastroesophageal Adenocarcinoma
  • the primary objective in Part 1 is to determine dose-limiting toxicity (DLT) and the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) for Compound A when administered with each of the combination partners.
  • DLT dose-limiting toxicity
  • MTD maximum tolerated dose
  • R2D recommended phase 2 dose
  • the primary objective in Part 2 is to evaluate the overall response rate (ORR) as the primary efficacy measure of Compound A in combination with each of the combination partners in patients with gastric or gastroesophageal adenocarcinoma.
  • the secondary objectives are to evaluate the safety and tolerability of Compound A in combination with each of the combination partners, to evaluate additional efficacy measures, such as progression-free survival (PFS), disease control rate, response duration, time to progression (TTP), and overall survival (OS) of Compound A in combination with each of the combination partners in patients with gastric or gastroesophageal adenocarcinoma, and to evaluate the pharmacokinetics (PK) of Compound A when dosed in combination with alisertib, Compound B, docetaxel, or paclitaxel, and to evaluate the PK of alisertib and Compound B when these agents are dosed in combination with Compound A.
  • PFS progression-free survival
  • TTP time to progression
  • OS overall survival
  • Inclusion criteria Male and female patients aged 18 years or older at the time of consent. In Part 1 (dose escalation), patients must have a histologically confirmed diagnosis of advanced solid tumor, including but not limited to gastric or gastroesophageal adenocarcinoma, and are refractory to or relapsed after prior line(s) of therapy with no effective therapeutic options available.
  • Part 1 dose escalation
  • patients must have a histologically confirmed diagnosis of advanced solid tumor, including but not limited to gastric or gastroesophageal adenocarcinoma, and are refractory to or relapsed after prior line(s) of therapy with no effective therapeutic options available.
  • Exclusion criteria Patients who have received prior systemic anticancer therapies, or other investigational agents or radiotherapy within 2 weeks before first dose of study drug; are receiving treatment with P-glycoprotein (P gp) inhibitors/inducers (Compound A + Compound B arm only); have received strong cytochrome P 450 (CYP) 3A4 inducers/inhibitors or proton pump inhibitors (PPIs) within 7 days before the first administration of study drug or have conditions that require the concomitant use of CYP3A4 inducers/inhibitors or PPIs during the course of the study; have poorly controlled diabetes mellitus; have signs of peripheral neuropathy >NCI CTCAE Grade 2; have symptomatic brain metastases or brain metastases with a stable neurologic status for ⁇ 2 weeks after completion of the definitive therapy and steroids.
  • P gp P-glycoprotein
  • PPIs proton pump inhibitors
  • the dose of Compound A will be escalated (planned doses of 300 mg, 600 mg, and 900 mg) according to a 3+3 dose escalation scheme, while Compound B, alisertib, paclitaxel, and docetaxel will be administered at a fixed dose and regimen until the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) is determined.
  • MTD maximum tolerated dose
  • R2D recommended phase 2 dose
  • Compound A will be administered PO QD for 3 days on (i.e., Days 2, 3, 4; 9, 10, 11;
  • the RP2D of Compound A when administrated concomitantly with docetaxel will be determined in the Part 1 dose escalation phase and used in the Part 2 dose expansion phase.
  • Docetaxel will be administered IV at 75 mg/m2 on Day 1 once every 3 weeks in a 21 -day cycle.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

COMBINATION TREATMENT WITH PI3Ka INHIBITORS AND TAXANES
BACKGROUND OF THE INVENTION
[0001] This application claims the benefit under 35 U.S.C. § 119, of United States Application No. 62/054,905 filed on September 24, 2014; United States Application No. 62/132,931 filed on March 13, 2015; and United States Application No. 62/195,479 filed on July 22, 2015. The entire contents of each of the aforesaid applications are incorporated by reference herein in their entireties.
BACKGROUND OF THE INVENTION
[0002] The phosphoinositide 3-kinases (PI3Ks) are members of a unique and conserved family of intracellular lipid kinases that phosphorylate the 3'-OH group on phosphatidylinositols or phosphoinositides. The PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation ( atso et al., 2001). The class I PI3Ks (pi 10a, pi 10β, pi 106, and pi ΙΟγ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), which engages downstream effectors such as those in the Akt/PDKl pathway, mTOR, the Tec family kinases, and the Rho family GTPases. The class II and ΙΠ PI3-Ks play a key role in intracellular trafficking through the synthesis of PI(3)P and PI(3,4)P2. The PIKKs are protein kinases that control cell growth (mTORCl) or monitor genomic integrity (ATM, ATR, DNA-P , and hSmg-1). The production of PIP3 initiates potent growth and survival signals. In some epithelial cancers the PI3K pathway is activated by direct genetic mutation. As PI3K signaling pathway plays a pivotal role in cell proliferation and differentiation, inhibition of this pathway has been shown to be beneficial in hyperproliferative diseases.
[0003] The alpha (a) isoform of PI3 (PI3Ks) has been implicated, for example, in a variety of human cancers. Angiogenesis has been shown to selectively require the a isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453;662-6). Mutations in the gene coding for PI3Koc or mutations which lead to upregulation of PI3Ka are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers. Often, mutations in the gene coding for PI3Kot are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K mutations, targeting of this pathway may provide valuable therapeutic opportunities. While other PI3K isoforms such as ΡΙ3Κγ or PI3K5 are expressed primarily in hematopoietic cells, PI3 a, along with ΡΙ3Κβ, is expressed constitutively.
[0004] Taxanes are diterpenes produced from the plants of the genus Taxus (yews). Taxanes are anticancer agents that interfere with cell growth by disrupting microtubule function. Examples of taxanes include, but are not limited to, paclitaxel, docetaxel, cabazitaxel, ortataxel, larotaxel, tesetaxel, 10-deacetyltaxol and cephalomannine. Docetaxel, (2R, 3S)— N-carboxy-3- phenylisoserine, N-tert-butyl ester, 13-ester with 5β-20-βροχν-1,2α,4,7β,10β,13α- hexahydroxytax-l-l-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®. Docetaxel is indicated for the treatment of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric
adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
SUMMARY OF THE INVENTION
[0005] The present invention provides compositions and methods for treating a variety of neoplastic diseases. Furthermore, the present invention also provides compositions and methods for treating a variety of diseases mediated by PI3K-kinase a.
[0006] In one aspect, the invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a
therapeutically effective amount of a combination of a PI3K-kinase a (PI3 a) inhibitor and a taxane, wherein the taxane is not paclitaxel. In another aspect, the invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a therapeutically effective amount of a combination of a PI3K- kinase a (PI3Ka) inhibitor and a taxane, wherein the taxane is not paclitaxel, and wherein the PI3 a inhibitor is a compound of the following formula:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and R3 is amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
[0007] In some embodiments, the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
[0008] In some embodiments, the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
[0009] In other embodiments, the neoplastic condition is a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer. In other embodiments, the neoplastic condition is gastric cancer. In certain embodiments, the cancer is gastrointestinal cancer. In certain such embodiments, the cancer is selected from gastric cancer and gastroesophageal adenocarcinoma. In certain such embodiments, the cancer is gastric cancer. In certain such embodiments, the cancer is gastroesophageal adenocarcinoma. In other embodiments, the neoplastic condition is non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is squamous non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer. In some embodiments, the subject has a PIK3CA mutation and/or amplification.
[0010] In another aspect, the invention provides a method of enhancing apoptosis in cancer cells comprising administering to the cells simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane. In another aspect, the invention provides for a method of enhancing apoptosis in cancer cells comprising aciministering to the cells simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3Ka) inhibitor and a taxane, wherein the PI3 a inhibitor is a compound of the following formula:
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
R3 is amido of formula -C(0)N(R)2 or-NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
[0011] In some embodiments, the administration takes place in vivo. In some embodiments, the administration takes place in vivo.
[0012] In yet another aspect, the invention provides a method of suppressing tumor regrowth in a subject in need thereof comprising administering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PD a) inhibitor and a taxane. In yet another aspect, the invention provides a method of method of suppressing tumor regrowth in a subject in need thereof comprising adrninistering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the PDKa inhibitor is a compound of the following formula:
Figure imgf000005_0002
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
R3 is amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
[0013] In some embodiments, the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the PI3 a inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the PD a inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the PI3Ka inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the taxane.
[0014] In some embodiments, the taxane and the PI3Ka inhibitor are administered
simultaneously. In some embodiments, the taxane and the PI3Ka inhibitor are administered sequentially, wherein the taxane and the PDKa inhibitor are introduced at two different time points. In some embodiments, the taxane is administered to the subject prior to administration of the PI3 a inhibitor. In some embodiments, the taxane is administered to the subject about 1, 5, 8, 10, 12, 24, 36 or 72 hours prior to administration of the PD a inhibitor. In some embodiments, taxane is administered to the subject about 12 to about 96 hours prior to administration of the PDKcc inhibitor. In some embodiments, the taxane is administered to the subject about 12 to about 72 hours prior to administration of the PI3Ka inhibitor. In some embodiments, taxane is administered to the subject about 12 to about 36 hours prior to administration of the PDKa inhibitor. In some embodiments, the taxane is administered to the subject about 24 hours prior to administration of the PI3Ka inhibitor.
[0015] In another but related aspect, the invention provides for a pharmaceutical regimen for the treatment of a disorder mediated by PI3 -kinase a (PDKa), wherein the regimen comprises simultaneous or sequential administration of at least one taxane and at least one PDKa inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel. In another aspect, the invention provides for a pharmaceutical regimen for the treatment of a disorder mediated by PD-kinase a (PDKa), wherein the regimen comprises simultaneous or sequential administration of at least one taxane and at least one PI3K.cc inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel; and wherein the PI3Ka inhibitor is a compound of the following formula:
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamide, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
R3 is amido of formula -C(0)N(R)2 or-NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
[0016] In some embodiments, the disorder is a cancer selected from the group consisting of non- small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
[0017] In some embodiments, the disorder is a cancer selected from the group consisting of non- small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
[0018] In other embodiments, the disorder is non-small cell lung cancer. In some embodiments, the disorder is a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer. In other embodiments the disorder is gastric cancer. In some embodiments, the disorder is squamous non-small cell lung cancer. In some
embodiments, the disorder is non-squamous non-small cell lung cancer. In some embodiments, the subject has a PIK3CA mutation and/or amplification [0019] In some embodiments, the regimen comprises at least one cycle wherein the taxane is administered before the PI3Ka inhibitor and wherein the taxane and the PI3Ka inhibitor are not administered on the same day. In some embodiments, the cycle comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 25, 30, 40 or more days. In some embodiments, the cycle comprises least 5 to 15 days. In some embodiments, the cycle consists of 7 days. In some embodiments the cycle consists of 21 days. In some embodiments, the PI3Ka inhibitor is administered nine times in a 21 -day cycle. In certain such embodiments, the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17, and 18. In certain such embodiments, the PI3Kct inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18, and 20.
[0020] In some embodiments, the PI3Kct inhibitor is administered according to an intermittent regimen. In some embodiments, the PI3K.cc inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission.
[0021] In some embodiments, the PI3Koc inhibitor is administered for at least 1 day, followed by an intermission in which the PI3Ka inhibitor is not administered for at least 1 day for at least one cycle. In some embodiments, the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3Ka inhibitor is not administered for at least 1 day. In some embodiments, the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1 , 2, 3, 4, 5, or 6 consecutive days. In some embodiments, the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days. In some
embodiments, the PI3Koc inhibitor is administered for 7, 8, 9, 10, 11, 12, 13, or 14 consecutive days, followed by an intermission where the PI3Ka inhibitor is not administered for at least 1, 2, 3, 4, 5, 6, or 7 days. In other embodiments, the PBKoc inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3K<x inhibitor is not administered for at least 3, 4, or 5 consecutive days.
[0022] In some embodiments, the PI3Ka inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In other embodiments, the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for 3 consecutive days of a 7-day cycle followed by an intermission of at least one day. In some embodiments, the PI3Kct inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 4 consecutive days followed by an intermission of 3 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Kot inhibitor is administered for 5 consecutive days followed by an intermission of 2 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Kct inhibitor is administered for 6 consecutive days followed by an intermission of 1 day for at least one 7-day cycle.
[0023] In some embodiments, the PI3Ka inhibitor is administered for at least 1 day for at least one 7- day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 2 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 3 days for at least one 7-day cycle. In some embodiments, the PI3Kct inhibitor is adrriinistered for at least 4 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 5 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 6 days for at least one 7-day cycle.
[0024] In some embodiments, the PI3Koc inhibitor is administered every other day. In some embodiments, the PI3Ka inhibitor is administered for three non-consecutive days in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle. In some embodiments, the PI3 a inhibitor is administered on alternate days in a 7-day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered at least 3 times on alternate days within a 7-day cycle. In some embodiments, the PI3Koc inhibitor is administered at least 4 times on alternate days within a 7-day cycle.
[0025] In some embodiments, the taxane is administered on Day 1 and the PI3Ka inhibitor is adrriinistered on the Days 2, 3 and 4 of a 7-day cycle. In some embodiments, the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 3, 4, and 5 of a 7-day cycle. In some embodiments, the taxane is admimstered on Day 1 and the ΡΙ3 α inhibitor is administered on Days 4, 5 and 6 of a 7-day cycle. In some embodiments, the taxane is administered on Day land the PI3Ka inhibitor is administered on Days 5, 6, and 7 of a 7-day cycle. In some embodiments, the taxane is administered on the Day 1 and the PI3Ka inhibitor is administered on Days 2, 4, and 6 of a 7-day cycle. In some embodiments, the taxane is admimstered on Day 1 and the PI3Ka inhibitor is administered on Days 3, 5, and 7 of a 7- day cycle.
[0026] In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17 and 18 of a 21 -day cycle as shown in Figure 8A. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is adrriinistered on Days 3, 4, 5, 10, 11, 12, 17, 18 and 19 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is adniinistered on Days 4, 5, 6, 11, 12, 13, 18, 19 and 20 of a 21 -day cycle. In some
embodiments, the taxane is administered on Days 1 and 8 and the PDKct inhibitor is administered on Days 5, 6, 7, 12, 13, 14, 19, 20 and 21 of a 21 -day cycle. In some
embodiments, the taxane is administered on Days 1 and 8 and the PDKot inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18 and 20 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 5, 7, 10, 12, 14, 17, 19 and 21 of a 21 -day cycle.
[0027] In some embodiments, the PI3Ka inhibitor is administered at least once a day in each of the days that the PI3 a inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered once daily in each of the days that the ΡΙ3Κα inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered twice daily in each of the days that the PI3Koc inhibitor is administered to the subject.
[0028] In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 6300 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 900 mg to about 3600 mg of the PI3Koc inhibitor in a 7-day cycle. In some embodiments, the PI3 ct inhibitor is administered to a subject within a range of about an amount of 900 mg to about 6300 mg of the PDKct inhibitor in a 7-day cycle.In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 300 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 900 mg. In some embodiments, the amount of PDKcc inhibitor administered in a 7-day cycle is about 1800 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 2700 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 3600 mg. In some embodiments, the amount of PDKct inhibitor administered in a 7-day cycle is about 4500 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 5400 mg. In some embodiments, the amount of PDKct inhibitor administered in a 7-day cycle is about 6300 mg.
[0029] In some embodiments, a dose of the PI3Ka inhibitor is about 100 to about 1200 mg. In some embodiments, a dose of the PDKct inhibitor is about 100 to about 2100 mg. In some embodiments, a dose of the PDKct inhibitor is about 300 to about 1200 mg. In some embodiments, a dose of the PI3 a inhibitor is about 300 to about 2100 mg. In some
embodiments a dose of the P13 a inhibitor is about 100 mg, about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, or about 1200 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, or about 1200 mg. In some embodiments a dose of the PI3 a inhibitor is about 100 mg, about 300 mg, about 600 mg, about
900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some
embodiments, a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3 a inhibitor is about 100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 600 mg.
In some embodiments, a dose of the PI3 a inhibitor is about 900 mg. In some embodiments, a dose of the PI3 a inhibitor is about 1200 mg. In some embodiments, a dose of the PDKa inhibitor is about 1500 mg. In some embodiments, a dose of the PI3 a inhibitor is about 1800 mg. In some embodiments, a dose of the PI3 a inhibitor is about 2100 mg.
[0030] In some embodiments, the taxane is administered to a subject at a dose of about 25 mg/m2 to about 100 mg m2 in a single administration. In some embodiments, the taxane is administered at a dose of about 25 mg/m2, 26 mg/m2, 27 mg/m2, 28 mg m2, 29 mg m2, 30 mg/m2, 31 mg m2, 32 mg m2, 33 mg/m2, 34 mg/m2, 35 mg/m2, 36 mg/m2, 37 mg/m2, 38 mg/m2,
39 mg/m2, 40 mg/m2, 41 mg m2, 42 mg/m2, 43 mg/m2, 44 mg/m2, 45 mg/m2, 46 mg/m2, 47 mg/m2, 48 mg/m2, 49 mg/m2, 50 mg/m2, 51 mg/m2, 52 mg/m2, 53 mg/m2, 54 mg/m2, 55 mg/m2,
56 mg/m , 57 mg/m", 58 mg/m', 59 mg/m", 60 mg/m , 61 mg/m", 62 mg/m , 63 mg/m , 64 mg/m , 65 mg/m , 66 mg/m", 67 mg m , 68 mg/m", 69 mg m", 70 mg/m , 71 mg/m", 72 mg/m ,
73 mg/m", 74 mg/m , 75 mg/m", 76 mg/m", 77 mg/m , 78 mg/m", 79 mg/m , 80 mg/m", 81 mg/m2, 82 mg/m2, 83 mg/m2, 84 mg/m2, 85 mg m2, 86 mg m2, 87 mg/m2, 88 mg/m2, 89 mg/m2',
90 mg/m2, 91 mg/m2, 2 mg/m2, 93 mg/m2, 94 mg/m2, 95 mg/m2, 96 mg m2, 97 mg/m2, 98 mg/m2, 99 mg/m2, and 100 mg/m2 in a single administration. In some embodiments, the taxane is administered at a dose of about 36 mg/m2 in a single administration. In some embodiments, the taxane is administered at a dose of about 75 mg/m2 in a single administration.
[0031] In some embodiments, the taxane is docetaxel. In some embodiments, the taxane is docetaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof.
[0032] In some embodiments, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 6-membered ring. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring. In some embodiments, R2 is amino. In a further embodiment, R2 is Nt .
[0033] In some embodiments, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring and R2 is amino. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 6-membered ring and R2 is amino. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring and R2 is NH2.
[0034] In some embodiments, the PO a inhibitor is a compound with the following structure:
Figure imgf000012_0001
or a pharmaceutically acceptable salt thereof.
INCORPORATION BY REFERENCE
[0035] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS
[0036] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
[0037] Figure 1 shows the effect of docetaxel on the phosphorylation of AKT, S6 and 4EBP1 in NCI-H1048 (PIK3CA-mutated) cells.
[0038] Figure 2A shows the effect of docetaxel and compound A on the in vitro
phosphorylation of AKT(308) in NCI-H1048. Figure 2B shows the effect of docetaxel and compound A on the in vitro phosphorylation of S6 240/244 in NCI-H1048 cells.
[0039] Figure 3A shows a Western blot depicting the effects of compound A and docetaxel on the in vitro apoptosis of NCI-H1048 cells. Figure 3B shows quantified cleaved PARP. [0040] Figure 4 shows the effect of docetaxel on the phosphorylation of AKT, S6 and 4EBP1 in Hs746T gastric xenograft tumor samples.
[0041] Figures 5A and 5B show the increased apoptosis observed in vivo in NCI-H1048 when pre-dosing docetaxel in combination with compound A. Figure 4A shows quantified Cleaved lamin A and Figure 5B shows quantified cleaved PARP.
[0042] Figures 6A and 6B show the effect that pre-dosing docetaxel in combination with compound A has on in vivo anti-tumor activity compared to simultaneous combination dosing in NCI-H1048 cells. Figure 6A shows the effect of pre-dosing docetaxel at a low clinically relevant dose. Figure 6B shows the effect of pre-dosing docetaxel at 10 mg kg.
[0043] Figure 7 shows the effect that pre-dosing docetaxel in combination with compound A has on enhanced tumor delay on NCI-H1048 cells (compound A: 140 mg kg QD3; docetaxel: 10 mg kg QW).
[0044] Figure 8 shows the effect that pre-dosing docetaxel for 24h in combination with compound A has on in vivo antitumor activity compared to simultaneous combination dosing in GA0098 xenografts
[0045] Figure 9 shows the administration of a combination of a taxane and a PI3Ka inhibitor wherein the PI3Kot inhibitor is administered according to an intermittent regimen for a 7-day cycle.
[0046] Figures 10A and 10B show the aclministration of a combination of a taxane and a PI3Kcc inhibitor wherein the PI3Ka inhibitor is administered according to an intermittent regimen for a 21 -day cycle.
DETAILED DESCRIPTION OF THE INVENTION
[0047] Several aspects of the invention are described below with reference to example applications for illustration. It should be understood that numerous specific details,
relationships, and methods are set forth to provide a full understanding of the invention. One having ordinary skill in the relevant art, however, will readily recognize that the invention can be practiced without one or more of the specific details or with other methods. Unless stated otherwise, the present invention is not limited by the illustrated ordering of acts or events, as some acts may occur in different orders and/or concurrently with other acts or events.
Furthermore, not all illustrated acts or events are required to implement a methodology in accordance with the present invention.
[0048] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms "including", "includes", "having", "has", "with", or variants thereof are used in either the detailed description and/or the claims, such terms are intended to be inclusive in a manner similar to the term "comprising".
[00491 The term "about" or "approximately" means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, "about" can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, "about" can mean a range of up to 20%, up to 10%, up to 5%, or up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term "about" meaning within an acceptable error range for the particular value should be assumed.
[0050] "Treatment", "treating", "palliating" and "ameliorating", as used herein, are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be aclministered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
[0051] As used herein, the term "neoplastic condition" or "neoplastic disorder" refers to the presence of cells possessing abnormal growth characteristics, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, perturbed oncogenic signaling, and certain characteristic morphological features. This includes but is not limited to the growth of: (1) benign or malignant cells (e.g., tumor cells) that correlates with
overexpression of a tyrosine or serine/threonine kinase; (2) benign or malignant cells (e.g., tumor cells) that correlates with abnormally high level of tyrosine or serine/threonine kinase activity or lipid kinase activity. Exemplary tyrosine kinases implicated in a neoplastic condition include but are not limited to receptor tyrosine kinases such as epidermal growth factor receptors (EGF receptor), platelet derived growth factor (PDGF) receptors, and cytosolic tyrosine kinases such as src and abl kinase. Non-limiting serine/threonine kinases implicated in neoplastic condition include but are not limited to raf, mek, mTor, and akt. Exemplary lipid kinases include but are not limited to PI3 kinases such as ΡΒΚα, ΡΒΚβ, PI3K5, and ΡΙ3 γ.
[0052] The term "effective amount" or "therapeutically effective amount" refers to that amount of an inhibitor described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or down regulation of activity of a target protein. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
[0053] A "sub-therapeutic amount" of an agent or therapy is an amount less than the effective amount for that agent or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a result desired by the physician, due to, for example, synergy in the resulting efficacious effects, or reduced side effects.
[0054] A "synergistically effective" therapeutic amount, "synergistically effective" amount of an agent or therapy is an amount which, when combined with an effective or sub-therapeutic amount of another agent or therapy, produces a greater effect than when either of the two agents are used alone. In some embodiments, a synergistically effective therapeutic amount of an agent or therapy produces a greater effect when used in combination than the additive effects of each of the two agents or therapies when used alone. The term "greater effect" encompasses not only a reduction in symptoms of the disorder to be treated, but also an improved side effect profile, improved tolerability, improved patient compliance, improved efficacy, or any other improved clinical outcome.
[0055] The terms "synergistic" and "synergistically" as applied to the effect of two or more pharmaceutically active ingredients used in combination (whether simultaneously or sequentially) refer to a greater effect than when either of the two agents are used alone.
[0056] As used herein, "agent" or "biologically active agent" refers to a biological,
pharmaceutical, or chemical compound or other moiety. Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound. Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures. In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
[0057] The term "agonist" or "activator" as used herein refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term "agonist" is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g., bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
[0058] The term "dose" as used herein refers to the amount of a particular pharmaceutically active ingredient used in a single/one-time administration. For example, the "dose" of a PI3 a inhibitor disclosed herein may be administered at a "dose" of 300 mg each time, and an amount of 900 mg over a course of a 7-day cycle during which the PI3 a inhibitor is administered on Day 2, Day 4 and Day 6 only.
[0059] The terms "antagonist" and "inhibitor" are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms "antagonist" and "inhibitors" are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition. A preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
[0060] The phrase "PI3Kcc inhibitor" refers to a PI3Ka inhibitor that interacts with and reduces activity of PI3Ka kinase. For example, the PI3 a inhibitor can be (6-(2-aminobenzo[d]oxazol- 5-yl)-midazo[l,2-a]pyridin-3-yl)(mo holino)methanone and its pharmaceutically acceptable salts, prodrugs, or radioactive isomers. As used herein, compound A is (6-(2- ammobenzo[d]oxazol-5-yl)imidazo[l,2-a]pyridin-3-yl)(mo holino)methanone and has the following structure:
Figure imgf000017_0001
[0061] An "anti-neoplastic", "anti-cancer agent", "anti-tumor agent" or "chemotherapeutic agent" refers to any agent useful in the treatment of a neoplastic condition. One class of anticancer agents comprises chemotherapeutic agents. "Chemotherapy" means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
[0062] The term "cell proliferation" refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
[0063] The terms "co-administration," "administered in combination with," and their grammatical equivalents, encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Coadministration includes simultaneous a<lministration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present. Co-administered agents may be in the same formulation. Co-administered agents may also be in different formulations.
[0064] A "therapeutic effect," as used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[0065] The term "pharmaceutically acceptable salt" refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as
isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, and sodium, calcium and magnesium salts.
[0066] "Pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
[0067] "Signal transduction" is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response. A modulator of a signal transduction pathway refers to a compound that modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway. A modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
[0068] "Subject" refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human.
[0069] The term "/« vivo" refers to an event that takes place in a subject's body.
[0070] The term "w vitro" refers to an event that takes places outside of a subject's body. For example, an in vitro assay encompasses any assay run outside of a subject assay. In vitro assays encompass cell-based assays in which cells alive or dead are employed. In vitro assays also encompass a cell-free assay in which no intact cells are employed.
[0071] The following abbreviations and terms have the indicated meanings throughout: PI3K = Phosphoinositide-3-kinase; PI = phosphatidylinositol.
[0072] Unless otherwise stated, the connections of compound name moieties are at the rightmost recited moiety. That is, the substituent name starts with a terminal moiety, continues with any linking moieties, and ends with the linking moiety. For example, heteroarylthio C alkyl has a heteroaryl group connected through a thio sulfur to a Ci^ alkyl radical that connects to the chemical species bearing the substituent. This condition does not apply where a formula such as, for example "-L-CMO alkyl -
Figure imgf000018_0001
is represented. In such case, the terminal group is a C3.8cycloalkyl group attached to a linking CMO alkyl moiety which is attached to an element L, which is itself connected to the chemical species bearing the substituent.
[0073] "Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., Ci-Cio alkyl). Whenever it appears herein, a numerical range such as "1 to 10" refers to each integer in the given range; e.g., "1 to 10 carbon atoms" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term "alkyl" where no numerical range is designated. In some embodiments, it is a C|-C4 alkyl group. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like. The alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), n-propyl, 1-methylethyl (iso-propyl), «-butyl, M-pentyl,
1,1-dimethylethyl (/-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more of substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, -SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, -N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tR8 (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0),N(Ra)2 (where t is 1 or 2), or P03(Ra)2 where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0074] The term "halo" or "halogen" refers to fluoro, chloro, bromo, or iodo.
[0075] The term "haloalkyl" refers to an alkyl group substituted with one or more halo groups, for example chloromethyl, 2-bromoethyl, 3-iodopropyl, trifluoromethyl, perfluoropropyl, 8- chlorononyl, and the like.
[0076] "Acyl" refers to the groups (alkyl)-C(O)-, (aryl)-C(O)-, (heteroaryl)-C(O)-,
(heteroalkyl)-C(O)-, and (heterocycloalkyl)-C(O)-, wherein the group is attached to the parent structure through the carbonyl functionality. In some embodiments, it is a Ci-Cio acyl radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocycloalkyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e. three other ring or chain atoms plus carbonyl. If the R radical is heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms. Unless stated otherwise specifically in the specification, the "R" of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, SRa, -OC(0)-R\ -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0),N(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0077] "Cycloalkyl" refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (i.e., C3-C10 cycloalkyl). Whenever it appears herein, a numerical range such as "3 to 10" refers to each integer in the given range; e.g., "3 to 10 carbon atoms" means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C3-C8 cycloalkyl radical. In some embodiments, it is a C3-C5 cycloalkyl radical. Illustrative examples of cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbomyl, and the like. Unless stated otherwise specifically in the specification, a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa,SRa, -OC(0)-Ra, -N(Ra)2, -C(0)R\ -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0078] As used herein, the term "heteroatom" or "ring heteroatom" is meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
[0079] "Heteroalkyl" includes optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof. A numerical range may be given, e.g., C1-C4 heteroalkyl which refers to the chain length in total, which in this example is 1 to 4 atoms long. For example, a -CH2OCH2CH3 radical is referred to as a "C4" heteroalkyl, which includes the heteroatom center in the atom chain length description. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl chain. A heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, SRa,
-OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2) -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0),N(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0080] An "alkene" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an "alkyne" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
[0081] "Alkenyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (i.e., C2-Cio alkenyl). Whenever it appears herein, a numerical range such as "2 to 10" refers to each integer in the given range; e.g., "2 to 10 carbon atoms" means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to five carbon atoms (e.g., C2-C5 alkenyl). The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-l-enyl (i.e., allyl), but-l-enyl, pent-l-enyl, penta-l,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, SRa, -OC(0)-R\ -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2> -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0082] Unless otherwise specified, the term "cycloalkenyl" refers to a cyclic aliphatic 3 to 8 membered ring structure, optionally substituted with alkyl, hydroxy and halo, having 1 or 2 ethylenic bonds such as methylcyclopropenyl, trifluoromethylcyclopropenyl, cyclopentenyl, cyclohexenyl, 1,4-cyclohexadienyl, and the like. [0083] "Alkynyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (i.e., C2-Cio alkynyl). Whenever it appears herein, a numerical range such as "2 to 10" refers to each integer in the given range; e.g., "2 to 10 carbon atoms" means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms. In other
embodiments, an alkynyl has two to five carbon atoms (e.g., C2-C5 alkynyl). The alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2,
-N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0084] "Amino" or "amine" refers to a -N(Ra)2 radical group, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification. When a -N(Ra)2 group has two Ra other than hydrogen they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring. For example, -N(Ra)2 is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. Unless stated otherwise specifically in the specification, an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, -SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)R\ - N(Ra)C(0)N(Ra)2, -N(Ra)C(NRa)N(Ra)2, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl and each of these moieties may be optionally substituted as defined herein.
[0085] "Amide" or "amido" refers to a chemical moiety with formula -C(0)N(R)2 or -
NHC(0)R, where R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), each of which moiety may itself be optionally substituted. In some embodiments it is a C1-C4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical. The R2' of - N(R)2 of the amide may optionally be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring. Unless stated otherwise specifically in the specification, an amido group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl. An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified. The procedures and specific groups to make such amides are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
[0086] "Aromatic" or "aryl" refers to an aromatic radical with six to ten ring atoms (e.g., C6-Cio aromatic or Ce-Cio aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl). Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals. Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl" by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
Whenever it appears herein, a numerical range such as "6 to 10" refers to each integer in the given range; e.g., "6 to 10 ring atoms" means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms. The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of ring atoms) groups. Unless stated otherwise specifically in the specification, an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano,
trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, -ORa, -SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -OC(0)N(Ra)2, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, - N(Ra)C(0)N(Ra)2, N(Ra)C(NRa)N(Ra)2) -N(Ra)S(0)tRa (where t is 1 or 2), -S(0),ORa (where t is 1 or 2), -S(0),N(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl or heteroarylalkyl. [0087] "Heteroaryl" or, alternatively, "heteroaromatic" refers to a 5- to 18-membered aromatic radical (e.g., C5-C13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system. Whenever it appears herein, a numerical range such as "5 to 18" refers to each integer in the given range; e.g., "5 to 18 ring atoms" means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms. Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl" by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene. An N-containing "heteroaromatic" or "heteroaryl" moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom. The polycyclic heteroaryl group may be fused or non-fused. The heteroatom(s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring(s). Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1 ,3-benzodioxolyl, benzofuranyl, benzoxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[6][l,4]dioxepinyl, benzo[b][l,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl), benzotlueno[3,2-d]pyrimidinyl, benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, cyclopenta[d]pyrimidinyl, 6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrirnidinyl, 5,6-dihydrobenzo[h]quinazolinyl, 5,6-dihydrobenzo[h]cinnolinyl, 6,7-dihydro-5H- benzo[6,7]cyclohepta[l,2-c]pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furazanyl, furanonyl, furo[3,2-c]pyridinyl, 5,6,7,8,9, 10-hexahydrocycloocta[d]pyrimidinyl,
5,6,7,8,9, 10-hexahydrocycloocta[d]pyridazinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridinyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-5,6,7,8-tetrahydroquinazolinyl, naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl,
5,6,6a, 7,8,9, 10, 1 Oa-octahydrobenzo[h]quinazolinyl, 1 -phenyl- lH-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrrolyl, pyrazolyl, pyrazolo[3,4-d]pyrirnidinyl, pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl,
5.6.7.8- tetrahydrobenzo[4,5]tMeno[2,3-d]pyrimidinyl,
6.7.8.9- tetrahydro-5H-cyclohepta[4,5]thieno[2,3-d]pyrimidinyl, 5,6,7,8-tetrahydropyrido[4,5-c]pyridazinyl, thiazolyl, thiadiazolyl, thiapyranyl, triazolyl, tetrazolyl, tnazinyl, thieno[2,3-d]pyrimidinyl, thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pyridinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, -SRa,
-OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -N(Ra)C(0)Ra, -N(Ra)S(0),Ra (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0)tN(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
[0088] The terms "aryl-alkyl", "arylalkyl" and "aralkyl" are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion with the terminal aryl, as defined above, of the aryl-alkyl moiety. Examples of aryl-alkyl groups include, but are not limited to, optionally substituted benzyl, phenethyl, phenpropyl and phenbutyl such as 4- chlorobenzyl, 2,4-dibromobenzyl, 2-methylbenzyl, 2-(3-fluorophenyl)ethyl, 2-(4- methylphenyl)ethyl, 2-(4-(trifluoromethyl)phenyl)ethyl, 2-(2-methoxyphenyl)ethyl, 2-(3- nitropheny ethyl, 2-(2,4-dichlorophenyl)ethyl, 2-(3,5-dimethoxyphenyl)ethyl, 3-phenylpropyl, 3-(3-chlorophenyl)propyli 3-(2-methylphenyl)propyl, 3-(4-methoxyphenyl)propyl, 3-(4- (trifluoromethyl)phenyl)propyl, 3-(2,4-dichlorophenyl)propyl, 4-phenylbutyl, 4-(4- chlorophenyl)butyl, 4-(2-methylphenyl)butyl, 4-(2,4-dichlorophenyl)butyl, 4-(2- methoxphenyl)butyl, and 10-phenyldecyl. Either portion of the moiety is unsubstituted or substituted.
[0089] The terms "heteroarylalkyl", "heteroarylalkyl", "heteroaryl-alkyl", "heteroaryl-alkyl", "hetaralkyl" and "heteroaralkyl" are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heteroaralkyl moiety with the terminal heteroaryl portion, as defined above, for example 3-furylmethyl, thenyl, furfuryl, and the like. Either portion of the moiety is unsubstituted or substituted.
[0090] The term "heterocyclyl" refers to a four-, five-, six-, or seven-membered ring containing one, two, three or four heteroatoms independently selected from nitrogen, oxygen and sulfur. The four-membered ring has zero double bonds, the five-membered ring has zero to two double bonds, and the six- and seven-membered rings have zero to three double bonds. The term "heterocyclyl" also includes bicyclic groups in which the heterocyclyl ring is fused to another monocyclic heterocyclyl group, or a four- to seven-membered aromatic or nonaromatic carbocyclic ring. The heterocyclyl group can be attached to the parent molecular moiety through any carbon atom or nitrogen atom in the group.
[0091] "Heterocycloalkyl" refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Whenever it appears herein, a numerical range such as "3 to 18" refers to each integer in the given range; e.g., "3 to 18 ring atoms" means that the heterocycloalkyl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, it is a C5-C10 heterocycloalkyl. In some embodiments, it is a C4-C10
heterocycloalkyl. In some embodiments, it is a C3-C 10 heterocycloalkyl. Unless stated otherwise specifically in the specification, the heterocycloalkyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocycloalkyl radical may be optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocycloalkyl radical is partially or fully saturated. The heterocycloalkyl may be attached to the rest of the molecule through any atom of the ring(s). Examples of such heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl,
2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl,
tetrahydropyranyl, thiomorpholinyl, tWamorpholinyl, l-oxo-thiomo holinyl, and
1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocycloalkyl moiety is optionally substituted by one or more substituents which
independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, -ORa, SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa,
-N(Ra)C(0)Ra, -N(Ra)S(0),Ra (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0),N(Ra)2 (where t is 1 or 2), or P03(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl or heteroarylalkyl.
[0092] "Heterocycloalkyl" also includes bicyclic ring systems wherein one non-aromatic ring, usually with 3 to 7 ring atoms, contains at least 2 carbon atoms in addition to 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms; and the other ring, usually with 3 to 7 ring atoms, optionally contains 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen and is not aromatic. [0093] The terms "heterocyclylalkyl", "heterocyclyl-alkyl", "hetcyclylalkyl", and "hetcyclyl- alkyl" are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heterocyclylalkyl moiety with the terminal heterocyclyl portion, as defined above, for example 3-piperidinylmethyl and the like. The term "heterocycloalkylene" refers to the divalent derivative of heterocycloalkyl.
[0094] The term "alkoxy" refers to the group -O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. "Lower alkoxy" refers to alkoxy groups containing one to six carbons. In some embodiments, C1-C4 alkyl, is an alkyl group which encompasses both straight and branched chain alkyls of from 1 to 4 carbon atoms.
[0095] The term "alkylthio" includes both branched and straight chain alkyl groups attached to a linking sulfur atom, for example methylthio and the like.
[0096] The term "oxo" refers to an oxygen that is double bonded to a carbon atom. One in the art understands that an "oxo" requires a second bond from the atom to which the oxo is attached. Accordingly, it is understood that oxo cannot be substituted onto an aryl or heteroaryl ring, unless it forms part of the aromatic system as a tautomer.
[0097] "Sulfonamidyl" or "sulfonamido" refers to a -S(=0)2-NR'R' radical, where each R' is selected independently from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). The R' groups in -NR'R' of the -S(=0)2-NRrR' radical may be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring. A sulfonamido group is optionally substituted by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
[0098] Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Compounds may be shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of the disclosed compounds and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers. [0099] The present invention includes all manner of rotamers and conformationally restricted states of an inhibitor of the invention.
[00100] Substituents for alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl monovalent and divalent derivative radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, alkynyl, cycloalkyl, and heterocycloalkyl) can be one or more of a variety of groups selected from, but not limited to: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, -OR', =0, =NR', =N-OR', -NR'R", - SR', -halogen, -SiR'R"R"\ -OC(0)R\ -C(0)R', -C02R',-C(0)NR'R", -OC(0)NR'R", - NR"C(0)R', -NR*-C(0)NR"R'", -NR"C(0)OR', -NR-C(NR'R")=NR"', -S(0)R', -S(0)2R\ - S(0)2NR'R", -NRS02R', -CN and -N02 in a number ranging from zero to (2m'+l), where m' is the total number of carbon atoms in such radical. R\ R", R'" and R"" each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1 -3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups. When an inhibitor of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R1, R", R"' and R"" groups when more than one of these groups is present.
[00101] When R* and R" or R" and Rm are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring. For example, -NR'R" is meant to include, but not be limited to, 1-pyrrolidinyl, 4 piperazinyl, and 4-morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term "alkyl" is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF3 and -CH2CF3) and acyl (e.g., -C(0)CH3, -C(0)CF3, - C(0)CH2OCH3, and the like).
[00102] Similar to the substituents described for alkyl radicals above, exemplary substituents for aryl and heteroaryl groups ( as well as their divalent derivatives) are varied and are selected from, for example: halogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, -OR', -NR'R", -SR', -halogen, -SiR'R"R"\ -OC(0)R',
-C(0)R\ -C02R', -C(0)NR'R", - OC(0)NR'R", -NR"C(0)R', -NR'-C(0)NR"R'",
-NR"C(0)OR', -NR-C(NR*R"R"')=NR"", -NR-C(NR'R")=NR"\ -S(0)R', -S(0)2R\
-S(0)2NR'R", -NRS02R\ -CN and -N02, -R\ -N3, -CH(Ph)2, fluoro(Ci-C4)alkoxo, and fluoro(Ci-C4)alkyl, in a number ranging from zero to the total number of open valences on aromatic ring system; and where R', R", R"' and R"" are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl. When an inhibitor of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R1, R", R'" and R"" groups when more than one of these groups is present.
[00103] As used herein, 0-2 in the context of -S(0)(o-2)- are integers of 0, 1, and 2.
[00104] Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(0)-(CRR')q-U-, wherein T and U are independently -NR-, -0-, - CRR'- or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH2) B-, wherein A and B are independently -CRR'-, -0-, -NR-, -S-, -S(O)-, -S(0)2-, -S(0)2NR'- or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -(CRR')s-X'-(C"R"l)d-, where s and d are independently integers of from 0 to 3, and X* is -0-, -NR'-, -S-, -S(O)-, -S(0)2-, or -S(0)2NR'-. The substituents R, R', R" and R'" are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
[00105] Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures wherein hydrogen is replaced by deuterium or tritium, or wherein carbon atom is replaced by 13C- or l C-enriched carbon, are within the scope of this invention.
[00106] The present invention may include compounds that contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention. For example, the present invention may include pharmaceutically acceptable salts and radioactive isomers of compound A.
[00107] The term "intermittent regimen" or "intermittent administration" refers to administration of a pharmaceutically active ingredient, (including but not limited to a PI3Ka inhibitor and/or a taxane) to a subject for at least one day, followed by an intermission, or rest period, of at least one day. For example, an intermittent regimen involves administering a pharmaceutically active ingredient for three consecutive days followed by a rest of 4 days in a 7- day treating period. In another example, an intermittent regimen involves administering a pharmaceutically active ingredient on three alternating days and not on days in between within a given treating period. In yet another example, an intermittent regimen involves administering a pharmaceutically active ingredient consecutively for at least 2, 3, 4, 5, 6, or more days, followed by an intermission of at least 1, 2, 3, 4, 5, 6 or more days over a given treating period. In still another example, the pharmaceutically active ingredient can be administered on three alternating days and not administered on the days in between within the 7-day period.
[00108] The term "simultaneous" or "simultaneously" as applied to administering more than one pharmaceutically active ingredient (e.g., a PBKct inhibitor and a taxane disclosed herein) refers to a<iministering the more than one ingredient at the same time, or at two different time points that are separated by no more than 2 hours. The term "sequentially" as applied to administering more than one pharmaceutically active ingredient (e.g., a PI3Ka inhibitor and a taxane disclosed herein) refers to acirninistering the more than one ingredient at two different time points that are separated by more than 2 hours, e.g., about 5 hours, 8 hours, lday, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days or even longer.
[00109] As used herein, the term "intermission" refers to a period that is that subsequent to the administration of a particular pharmaceutically active ingredient, (including but not limited to a PI3K.cc inhibitor and/or a taxane) of an intermittent regimen. Intermission refers to a rest period wherein a particular pharmaceutically active ingredient, (including but not limited to a PI3Ka inhibitor and/or a taxane) is not adrninistered for at least one day.
[00110] The terms "a week" or "7-day cycle", as used herein, are used interchangeably. These terms refer to a continuous period of time covering the duration of seven consecutive days. For example, a week can start on Monday and end on the following Sunday, or a 7-day cycle can start on Wednesday and end on the next Tuesday.
[00111] When ranges are used herein for physical properties, such as such as molecular weight, or chemical properties, such as chemical formulae, or dose ranges, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary from, for example, between 1% and 15% of the stated number or numerical range. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including") includes those embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, that "consist of or "consist essentially of the described features.
[00112] The PI3Kct inhibitors (e.g., compound A and its pharmaceutically acceptable salts and prodrugs) described herein can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
[00113] The present invention includes all manner of rotamers and conformationally restricted states of the PBKa inhibitor.
[00114] The PBKa inhibitor of the present invention also include crystalline and amorphous forms of those compounds, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof. "Crystalline form," "polymorph," and "novel form" may be used interchangeably herein, and are meant to include all crystalline and amorphous forms of the compound, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms, as well as mixtures thereof, unless a particular crystalline or amorphous form is referred to.
[00115] The PI3Ka inhibitor described herein can be optionally contacted with a pharmaceutically acceptable acid to form the corresponding acid addition salts. Pharmaceutically acceptable forms of the compounds recited herein include pharmaceutically acceptable salts, chelates, non-covalent complexes, prodrugs, and mixtures thereof. In certain embodiments, the PI3Ka inhibitor described herein are in the form of pharmaceutically acceptable salts. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition salts.
[00116] The term "taxane" as used herein to describe the compounds that are derived from the plants of genus Taxus (yews). Synonyms for taxane include but are not limited to, taxoid or taxol derivative. Examples of taxanes include, but are not limited to, docetaxel, paclitaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof. Methods and Treatment Regimen
[00117] In one aspect, the present invention provides a method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the taxane is not paclitaxel. In some embodiments, the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
[00118] In some embodiments, the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
[00119] In some embodiments, the neoplastic condition is non-small cell lung cancer. In. some embodiments, the non-small cell lung cancer is squamous non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer. In some embodiments, the neoplastic condition is small cell lung cancer. In other embodiments, the subject has a PIK3CA mutation and/or amplification.
[00120] In another aspect, the present invention provides a method of enhancing apoptosis in cancer cells comprising administering to the cells simultaneously or sequentially a
therapeutically effective amount of a combination of a PI3-kinase a (PI3Ka) inhibitor and a taxane. In some embodiments, the administration takes place in vitro. In other embodiments, the administration takes place in vivo.
[00121] In another aspect, the present invention provides a method of suppressing tumor regrowth in a subject in need thereof administering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3Ka) inhibitor and a taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the PI3 a inhibitor or the taxane. In some embodiments, the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the PDKa inhibitor or the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the PI3 a inhibitor or the taxane. In other embodiments, the subject does not exhibit tumor regrowth for at least 40 days from the first date of administration of the taxane.
[00122] In practicing any of the discussed methods, a PO a inhibitor and a taxane can be administered sequentially, where the two agents are introduced into a subject at two different time points. The two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, or according to any intermittent regimen schedule disclosed herein.
[00123] In some embodiments, a taxane is administered prior to the administration of the POKa inhibitor. Pre-dosing of taxane may occur at about 1, 5, 8, 10, 12, 24, 36, or 72 hours before administering the PI3Ka inhibitor. Pre-dosing may also take place at about 1, 2, 3, 4, 5, 6, 7, or more days before administering the PI3Ka inhibitor. Pre-dosing of taxane may provide a synergistic therapeutic effect as compared to simultaneous administration of the two agents or administration of either agent alone in down regulating PI3Ka signaling, increasing apoptosis of cancer cells size and/or reducing tumor inhibition of tumor regrowth.
[00124] In certain embodiments, the PI3 a inhibitor and the taxane are administered simultaneously. Simultaneous administration may take the format of co-administration of the two agents in same formulation, or in different formulations but at the same time.
[00125] As used herein, the subject methods utilize a therapeutically effective amount of a combination of a PI3Ka inhibitor and a taxane. In general, the combination is sufficient to effect the intended application including but not limited to disease treatment, as defined herein. Encompassed in the subject methods is the use of a therapeutically effective amount of a PI3Ka inhibitor and/or a taxane in combination to effect such treatment. Also contemplated in the subject methods is the use of a sub-therapeutic amount of a PI3Ka inhibitor and/or a taxane in the combination for treating an intended disease condition. The PI3 a inhibitor and taxane individually, though present in sub-therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
[00126] The therapeutically effective amount of the subject combination of compounds may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried. [00127] The effect of inhibiting PO a pathway may be measured, for example, as a percentage decrease in phosphorylation of a protein chosen from p4EBPl, pS6, and pPRAS40. In some embodiments, pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of p4EBPl. For example, phosphorylation of p4EBPl is reduced by at least 60%. In other embodiments, pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of pS6. For example, phosphorylation of pS6 is reduced by at least 60%. In yet other embodiments, pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of pPRAS40. For example, phosphorylation of pPRAS40 is reduced by at least 60%. In yet other embodiments, pathway inhibition is measured as a 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater decrease in phosphorylation of p4EBPl, pS6, and pRAS40. For example, phosphorylation of p4EBPl, pS6, and pPRAS40 is reduced by at least 60%. In some embodiments, pathway inhibition is measured in peripheral blood cells. In other embodiments, pathway inhibition is measured in a biopsy, for example a skin biopsy.
[00128] In a separate but related aspect, the present invention provides for the treatment of a disorder mediated by PI3-kinase a (PI3 a), wherein the regimen comprises simultaneous or sequential aciministration of at least one taxane and at least one PI3Kct inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel. In some embodiments, the regimen comprises at least one cycle wherein the taxane is administered before the PI3Kot inhibitor in a given cycle and wherein the taxane and the PDKoc inhibitor are not administered on the same day. In some embodiments, the cycle comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 25, 30, 40 or more days. In some embodiments, the cycle comprises least 5 to 15 days. In some embodiments, the cycle consists of 7 days. In some embodiments the cycle consists of 21 days. In some embodiments, the PI3K.cc inhibitor is administered nine times in a 21 -day cycle. In certain such embodiments, the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17, and 18. In certain such embodiments, the PI3Ka inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18, and 20.
[00129] In some embodiments, the taxane is administered for at least one day followed by an intermission in which the taxane is not administered for at least one day. In some embodiments, the taxane is administered at least once per 7-day cycle. In some embodiments, the taxane is administered at least once per 21 -day cycle. In some embodiments, the taxane is administered twice per 21 -day cycle, such as on Day 1 and Day 8 of a 21 -day cycle. In some embodiments, the taxane is administered once per 21 -day cycle, such as on Day 1. [00130] In some embodiments, the taxane and the PI3-kinase a inhibitor are administered simultaneously. In some embodiments, the taxane is administered to the subject prior to adniinistration of the PI3Ka inhibitor. In some embodiments, the taxane is administered to the subject about 1, 5, 8, 10, 12, 24, 36 or 72 hours prior to administration of the PI3 a inhibitor. In some embodiments, taxane is aciministered to the subject about 12 to about 96 hours prior to administration of the PI3Ka inhibitor. In some embodiments, the taxane is administered to the subject about 12 to about 72 hours prior to administration of the PI3 a inhibitor. In some embodiments, the taxane is administered to the subject about 12 to about 36 hours prior to administration of the PDKct inhibitor. In some embodiments, the taxane is administered to the subject about 12 hours prior to administration of the PDKct inhibitor. In some embodiments, the taxane is administered to the subject about 18 hours prior to administration of the ΡΙ3Κα inhibitor. In some embodiments, the taxane is administered to the subject about 24 hours prior to administration of the PDKct inhibitor. In some embodiments, the taxane is administered to the subject about 30 hours prior to aclrninistration of the PI3 a inhibitor. In some
embodiments, the taxane is aciministered to the subject about 36 hours prior to administration of the PDKct inhibitor.
[00131] In some embodiments, a given dosing schedule comprises one or more administrations of a PI3Ka inhibitor, wherein at least one administration of a PI3 a inhibitor, such as described herein, may be repeated or cycled on a daily, weekly, biweekly, monthly, bimonthly, annually, semi-annually, or any other period. A repeated dosing schedule or cycle may be repeated for a fixed period of time determined at the start of the schedule; may be terminated, extended, or otherwise adjusted based on a measure of therapeutic effect, such as a level of reduction in the presence of detectable disease tissue (e.g. a reduction of at least 50%, 60%, 70%, 80%, 90%, 95%, 99%, or 100%); or may be terminated, extended, or otherwise adjusted for any other reason as determined by a medical professional.
[00132] In some embodiments, the PI3K inhibitor is administered according to an intermittent regimen. In some embodiments, the PI3Ka inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission.
[00133) In some embodiments, the PDKct inhibitor is administered for at least 1 day, followed by an intermission in which the PDKct inhibitor is not administered for at least 1 day for at least one cycle. In some embodiments, the PI3Kct inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PDKct inhibitor is not administered for at least 1 day. In some embodiments, the PI3Ka inhibitor is administered for 1, 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days. In some embodiments, the PDKa inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days. In some
embodiments, the PI3Ka inhibitor is administered for 7, 8, 9, 10, 11, 12, 13, or 14 consecutive days, followed by an intermission where the ΡΙ3Κα inhibitor is not administered for at least 1, 2, 3, 4, 5, 6, or 7 days. In other embodiments, the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6 or 7 consecutive days followed by an intermission in which the PI3Ka inhibitor is not administered for at least 3, 4, or 5 consecutive days.
[00134] In some embodiments, the ΡΙ3Κα inhibitor is administered on consecutive days in a 7- day cycle followed by an intermission. In yet other embodiments, the PI3Koc inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of at least one day per 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 4 consecutive days followed by an intermission of 3 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 5 consecutive days followed by an intermission of 2 consecutive days for at least one 7-day cycle. In yet other embodiments, the PI3Ka inhibitor is administered for 6 consecutive days followed by an intermission of 1 day for at least one 7-day cycle.
[00135] In some embodiments, the PI3Ka inhibitor is administered for at least 1 day for at least one 7- day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 2 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 3 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 4 days for at least one 7-day cycle. In some embodiments, the PI3Kcc inhibitor is administered for at least 5 days for at least one 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered for at least 6 days for at least one 7-day cycle.
[00136] In some embodiments, the PI3Ka inhibitor is administered every other day (i.e., 7 dosing days in 2 weeks). In some embodiments, the PI3Ka inhibitor is administered for three non-consecutive days within a 7-day cycle. In some embodiments, the PI3Koc inhibitor is administered on alternate days in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered on alternate days in a 7-day cycle followed by an intermission. For example, the ΡΙ3Κα inhibitor is administered at least 2 times on alternate days within a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered at least 3 times on alternate days within a 7- day cycle. In some embodiments, the PI3 a inhibitor is administered at least 4 times on alternate days within a 7-day cycle.
[00137] In some embodiments, the taxane is administered on Day 1 and the PI3 a inhibitor is administered on the Days 2, 3 and 4 of a 7-day cycle as shown in Figure 7. In some
embodiments, the taxane is administered on Day 1 and the PI3 a inhibitor is aclministered on Days 3, 4, and 5 of a 7-day cycle. In some embodiments, the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 4, 5 and 6 of a 7-day cycle. In some embodiments, the taxane is administered on Day land the PI3K.cc inhibitor is administered on Days 5, 6, and 7 of a 7-day cycle. In some embodiments, the taxane is a<lministered on the Day 1 and the PI3Ka inhibitor is administered on Days 2, 4, and 6 of a 7-day cycle as shown in Figure 7. In some embodiments, the taxane is administered on Day 1 and the PI3Koc inhibitor is administered on Days 3, 5, and 7 of a 7- day cycle.
[00138] In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 2, 3, 4, 9, 10, 11, 16, 17 and 18 of a 21 -day cycle as shown in Figure 8A. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 4, 5, 10, 11, 12, 17, 18 and 19 of a 21 -day cycle. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is aoministered on Days 4, 5, 6, 11, 12, 13, 18, 19 and 20 of a 21 -day cycle. In some
embodiments, the taxane is administered on Days 1 and 8 and the PI3Koc inhibitor is administered on Days 5, 6, 7, 12, 13, 14, 19, 20 and 21 of a 21-day cycle. In some
embodiments, the taxane is administered on Days 1 and 8 and the PI3Koc inhibitor is administered on Days 2, 4, 6, 9, 11, 13, 16, 18 and 20 of a 21-day cycle as shown in Figure 8B. In some embodiments, the taxane is administered on Days 1 and 8 and the PI3Ka inhibitor is administered on Days 3, 5, 7, 10, 12, 14, 17, 19 and 21 of a 21-day cycle.
[00139] In some embodiments, the PI3Ka inhibitor is administered at least once a day (QD) in each of the days that the PI3Ka inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is administered once a day (QD) in each of the days that the PI3Ka inhibitor is administered to the subject. In some embodiments, the PI3Ka inhibitor is aclministered twice a day (BID) in each of the days that the PI3Ka inhibitor is administered to the subject.
[00140] In some embodiments, a PI3K inhibitor and/or any additional therapeutic compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day or per week. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In some embodiments, cycles of administering a PI3Ka inhibitor followed by periods of intermission (or rest) are repeated for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, repetition of a dosing cycle comprising administration of a PI3 a inhibitor followed by intermission (or rest) is continued as long as necessary. Administration of the treatment regimens of the invention may continue as long as necessary and/or as long as the subject does not suffer from severe and intolerable toxicity. In some embodiments, a PI3Kcc inhibitor of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, 21, or 28 days. In some embodiments, a PI3 a inhibitor of the invention is administered for less than 28, 21, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a PI3Ka inhibitor of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[00141] The amount of the PI3Kcc inhibitor administered herein may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
[00142] The PI3Ka inhibitor may be administered in any suitable amount. In some
embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 300 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3Ka inhibitor is administered to a subject within a range of about an amount of 900 mg to about 3600 mg of the PI3Ka inhibitor in a 7-day cycle. In some embodiments, the PI3 a inhibitor is administered to a subject within a range of about an amount of 900 mg to about 6300 mg of the PI3Koc inhibitor in a 7-day cycle. For example, this inhibitor is administered to a subject at a dosage of about 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, 3000, 3100, 3200, 3300, 3400, 3600, 4500, 5400, or 6300 mg in 7-day cycle. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 300 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 900 mg. In some embodiments, the amount of PI3Ka inhibitor aclministered in a 7-day cycle is about 1800 mg. In some embodiments, the amount of PI3Kcc inhibitor administered in a 7-day cycle is about 2700 mg. In some embodiments, the amount of PI3Kct inhibitor administered in a 7-day cycle is about 3600 mg. In some embodiments, the amount of PI3Koc inhibitor administered in a 7-day cycle is about 4500 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 5400 mg. In some embodiments, the amount of PI3Ka inhibitor administered in a 7-day cycle is about 6300 mg.
[00143] In some embodiments, a dose (i.e., as applied to a single administration) of the PI3Ka inhibitor is about 100 to about 1200 mg. In some embodiments, a dose (i.e., as applied to a single administration) of the PI3 a inhibitor is about 100 to about 2100 mg. In some embodiments, a dose of the PDKot inhibitor is about 300 to about 1200 mg. In some embodiments, a dose of the PI3 a inhibitor ts about 300 to about 2100 mg. In some embodiments a dose of the PI3 a inhibitor is about 100 mg, about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PDKot inhibitor is about 300 mg, about 600 mg, or about 900 mg. In some embodiments a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 100 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 300 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 600 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 900 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1200 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1 00 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 1800 mg. In some embodiments, a dose of the PI3Ka inhibitor is about 2100 mg.
[00144] In some embodiments, a dose of PI3Ka inhibitor is within a range of about 1 mg/kg- 100 mg kg per day, such as about, less than about, or more than about, 1 mg/kg, 2 mg/kg, 3 mg kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, 15 mg/kg, 16 mg/kg, 17 mg/kg, 18 mg/kg, 19 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg, 40 mg/kg, 45 mg/kg, 50 mg/kg, 60 mg/kg, 70 mg/kg, 80 mg/kg, 90 mg kg, or 100 mg/kg per day. The administration schedule may be repeated according to any regimen according to the invention, including any adrninistration schedule described herein.
[00145] A dose of PI3Kct inhibitor may be about, at least about, or at most about 0.1 , 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1400, 1425, 1450 mg or mg/kg, or any range derivable therein. For example, such a dose can range from about 30-120 mg/kg (e.g., 40-80 mg/kg such as about 50 or about 60 mg/kg). It is contemplated that a dosage of mg/kg refers to the mg amount of inhibitor per kg of total body weight of the subject. It is contemplated that when multiple doses are given to a patient, the doses may vary in amount or they may be the same.
[00146] The taxane may be administered in any suitable amount. In some embodiments, the taxane is administered at a dose of any suitable amount in a single administration. In some embodiments, the taxane is administered to a subject at a dose of about 25 mg m2 to about 100 mg/m2 in a single administration. In some embodiments, the taxane is aclministered at a dose of about 25 mg/m2, 26 mg/m2, 27 mg/m2, 28 mg/m2, 29 mg/m2, 30 mg/m2, 31 mg/m2, 32 mg/m2, 33 mg/m2, 34 mg/m2, 35 mg/m2, 36 mg/m2, 37 mg/m2, 38 mg/m2, 39 mg/m2, 40 mg/m2, 41 mg/m2, 42 mg/m2, 43 mg/m2, 44 mg/m2, 45 mg/m2, 46 mg/m2, 47 mg m2, 48 mg/m2, 49 mg/m2, 50 mg m2, 51 mg/m2, 52 mg/m2, 53 mg m2, 54 mg/m2, 55 mg/m2, 56 mg/m2, 57 mg/m2, 58 mg m2, 59 mg/m2, 60 mg/m2, 61 mg/m2, 62 mg/m2, 63 mg/m2, 64 mg/m2, 65 mg/m2, 66 mg/m2, 67 mg/m2, 68 mg/m2, 69 mg/m2, 70 mg/m2, 71 mg/m2, 72 mg/m2, 73 mg/m2, 74 mg m2, 75 mg/m2, 76 mg/m2, 77 mg/m2, 78 mg/m2, 79 mg/m2, 80 mg/m2, 81 mg/m2, 82 mg m2, 83 mg/m2, 84 mg/m2, 85 mg/m2, 86 mg m2, 87 mg/m2, 88 mg/m2, 89 mg/m2', 90 mg/m2, 91 mg/m2, 92 mg/m2, 93 mg/m2, 94 mg/m2, 95 mg/m2, 96 mg/m2, 97 mg/m2, 98 mg m2, 99 mg/m2, and 100 mg/m2 in a single administration. In some embodiments, the taxane is administered at a dose of about 36 mg m2 in a single administration. In some embodiments, the taxane is administered at a dose of about 75 mg/m2 in a single administration.
[00147] The amount of PI3 a inhibitor or taxane administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
[00148] The PI3 a inhibitor and the taxane disclosed herein can be prepared in a dosage form for administration to a subject. When the dosage form is a solid, the dosage form can be a single capsule, tablet, or pill, or alternatively can be comprised of multiple capsules, tablets or pills (e.g., a single dose of 900 mg may be administered as three dosage forms, such as three tablets, each comprising 300 mg of the PI3Ka inhibitor or the taxane). A dosage form may be administered to a subject once or multiple times per day. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell or tissue being treated, and the subject being treated. Single or multiple administrations (e.g. about or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, or more doses) can be carried out with the dose level and pattern being selected by the treating physician.
PDKot Inhibitors [00149] In one aspect, the PI3Ka inhibitor is a compound of the following formula:
Figure imgf000041_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamide, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
[00150] In some embodiments, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 6-membered ring. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring.
[00151] In some embodiments, R2 is amino. In a further embodiment, R2 is NH2.
[00152] In some embodiments, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 4-, 5-, 6-, or 7-membered ring and R2 is amino. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a 6-membered ring and R2 is amino. In a further embodiment, R3 is amido of formula -C(0)N(R)2 wherein the (R)2 groups taken together with the nitrogen to which they are attached form a morpholinyl ring and R2 is NH2.
[00153] In some embodiments, the PI3 a inhibitor is a compound with the following structure:
Figure imgf000041_0002
or a pharmaceutically acceptable salt thereof.
[00154] In some embodiments, the PI3Ka inhibitor is (6-(2-aminobenzo[d]oxazol-5- yl)imidazo[l ,2-a]pyridin-3-yl)(morpholino)methanone) or compound A.
Taxanes
[00155] Taxanes are diterpenes produced from the plants of the genus Taxus (yews). Taxanes interfere with cell growth by disrupting with microtubule function. Examples of taxanes include, but are not limited to, docetaxel. Docetaxel, (2R, 3S)~N-carboxy-3-phenylisoserine, N- tert-butyl ester, 13-ester with 5 β-20-epoxy- 1 ,2α,4, 7β, 10β, 13a-hexahydroxytax- 1 - 1 -en-9-one 4- acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as
TAXOTERE®. Docetaxel is indicated for the treatment of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocarcinoma, and squamous cell carcinoma of head and neck cancer.
[00156] Other examples of taxanes contemplated for use include semisynthetic taxanes, including but not limited to, cabazitaxel (taxoid XRP6258 and Jevtana®), ortataxel (SB-T- 101131, IDN5109, and BAY59-8862), larotaxel (XRP9881 and RPR 109881), tesetaxel (DJ- 927), 10-deacetyltaxol. Other examples of taxanes include cephalomannine (Taxol® B).
[00157] In some embodiments, the taxane is docetaxel. In some embodiments, the taxane is docetaxel, cabazitaxel, ortataxel, larotaxel, 10-deacetyltaxol, tesetaxel, or derivatives thereof.
[00158] The subject methods and regimen are useful for treating any disease conditions, for example neoplastic diseases. In some embodiments, the disease condition is a proliferative disorder, such as described herein, including but not limited to cancer.
[00159] In some embodiments, the disease condition is associated with PI3-kinase. A vast diversity of disease conditions associated with PI3-kinase have been reported. PI3-kinase a, one of the four isoforms of type I PI3-kinases has been implicated, for example, in a variety of human proliferative disorders, such as cancers. Angiogenesis has been shown to selectively require PI3Ka in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6). Mutations in the gene coding for PI3 a or mutations which lead to upregulation of PI3 a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers. Often, mutations in the gene coding for PI3 a are point mutations clustered within several hotspots in helical and kinase domains, such as E542 , E545 , and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K a mutations, targeting of this pathway provides valuable therapeutic opportunities. While other PI3K isoforms such as PI3K δ or PI3 γ are expressed primarily in hematopoietic cells, PI3K a, along with PI3K β, is expressed constitutively.
[00160] Disease conditions associated with PI3-kinase can also be characterized by abnormally high level of activity and/or expression of downstream messengers of mTOR and PI3-kinase. For example, proteins or messengers such as PIP2, PIP3, PDK, Akt, PTEN, PRAS40, GSK-3 , p21 , p27 may be present in abnormal amounts which can be identified by any assays known in the art.
[00161] Deregulation of the PI3K/Akt mTOR pathway is emerging as a common theme in diverse human diseases and as a consequence drugs that target PI3 a have therapeutic value. The diseases associated with deregulation of PI3 a include, but are not limited to, tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), both of which are caused by mutations in TSC1 or TSC2 tumor suppressors. Inhibition of PI3Kct may help patients with Peutz-Jeghers cancer-prone syndrome caused by the LKB 1 mutation. PI3Ka may also have role in the genesis of sporadic cancers. Inactivation of several tumor suppressors, in particular PTEN, p53, VHL and NF1, has been linked to mTORCl activation. The PI3K Akt/mTOR pathway is activated in many cancers. Activated Akt regulates cell survival, cell proliferation and metabolism by phosphorylating proteins such as BAD, FOXO, NF- KB, p21Cipl, p27Kipl, GSK3P and others. Akt might also promote cell growth by phosphorylating TSC2. Akt activation may promote cellular transformation and resistance to apoptosis by collectively promoting growth, proliferation and survival, while inhibiting apoptotic pathways.
[00162] Where desired, the subject to be treated is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to the PI3Ka inhibitor and/or taxane. Any method known in the art that can determine the sensitivity of the tumor cells of a subject to a PI3Ka inhibitor and/or a taxane can be employed. In these methods one or more additional anticancer agents or treatments can be co-administered according to a treatment regimen of the invention using the combination of a PI3Ka inhibitor and a taxane, as judged to be appropriate by the adrninistering physician given the prediction of the likely responsiveness of the subject to the combination of the PI3Ka inhibitor and taxane, in combination with any additional circumstances pertaining to the individual subject.
[00163] The data presented in the Examples herein below demonstrate that the anti-tumor effects of administering sequentially or simultaneously a combination of a taxane and a PI3 a inhibitor. As such, the subject methods or treatment regimen is particularly useful for treating a proliferative disorder, such as a neoplastic condition. Non-limiting examples of such conditions include but are not limited to Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblasts leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar soft part sarcoma, Ameloblastic fibroma, Anal cancer, Anaplastic large cell lymphoma, Anaplastic thyroid cancer, Angioimmunoblastic T-cell lymphoma, Angiomyolipoma, Angiosarcoma, Appendix cancer, Astrocytoma, Atypical teratoid rhabdoid tumor, Basal cell carcinoma, Basal- like carcinoma, B-cell leukemia, B-cell lymphoma, Bellini duct carcinoma, Biliary tract cancer, Bladder cancer, Blastoma, Bone Cancer, Bone tumor, Brain Stem Glioma, Brain Tumor, Breast Cancer, Brenner tumor, Bronchial Tumor, Bronchioloalveolar carcinoma, Brown tumor, Burkitt's lymphoma, Cancer of Unknown Primary Site, Carcinoid Tumor, Carcinoma,
Carcinoma in situ, Carcinoma of the penis, Carcinoma of Unknown Primary Site,
Carcinosarcoma, Castleman's Disease, Central Nervous System Embryonal Tumor, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Cholangiocarcinoma, Chondroma, Chondrosarcoma, Chordoma, Choriocarcinoma, Choroid plexus papilloma, Chronic
Lymphocytic Leukemia, Chronic monocytic leukemia, Chronic myelogenous leukemia, Chronic Myeloproliferative Disorder, Chronic neutrophilic leukemia, Clear-cell tumor, Colon Cancer, Colorectal cancer, Craniopharyngioma, Cutaneous T-cell lymphoma, Degos disease,
Dermatofibrosarcoma protuberans, Dermoid cyst, Desmoplastic small round cell tumor, Diffuse large B cell lymphoma, Dysembryoplastic neuroepithelial tumor, Embryonal carcinoma, Endodermal sinus tumor, Endometrial cancer, Endometrial Uterine Cancer, Endometrioid tumor, Enteropathy-associated T-cell lymphoma, Ependymoblastoma, Ependymoma, Epithelioid sarcoma, Erythroleukemia,Esophageal cancer, Esthesioneuroblastoma, Ewing Family of Tumor, Ewing Family Sarcoma, Ewing's sarcoma, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Extramammary Paget's disease, Fallopian tube cancer, Fetus in fetu, Fibroma, Fibrosarcoma, Follicular lymphoma, Follicular thyroid cancer, Gallbladder Cancer, Gallbladder cancer, Ganglioglioma, Ganglioneuroma, Gastric Cancer, Gastric lymphoma, Gastrointestinal cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Stromal Tumor, Gastrointestinal stromal tumor, Germ cell tumor, Germinoma, Gestational choriocarcinoma, Gestational Trophoblastic Tumor, Giant cell tumor of bone, Glioblastoma multiforme, Glioma, Gliomatosis cerebri, Glomus tumor, Glucagonoma, Gonadoblastoma, Granulosa cell tumor, Hairy Cell Leukemia, Hairy cell leukemia, Head and Neck Cancer, Head and neck cancer, Heart cancer, Hemangioblastoma, Hemangiopericytoma, Hemangiosarcoma,
Hematological malignancy, Hepatocellular carcinoma, Hepatosplenic T-cell lymphoma, Hereditary breast-ovarian cancer syndrome, Hodgkin Lymphoma, Hodgkin's lymphoma,
Hypopharyngeal Cancer, Hypothalamic Glioma, Inflammatory breast cancer, Intraocular
Melanoma, Islet cell carcinoma, Islet Cell Tumor, Juvenile myelomonocytic leukemia, Sarcoma,
Kaposi's sarcoma, Kidney Cancer, Klatskin tumor, Krukenberg tumor, Laryngeal Cancer,
Laryngeal cancer, Lentigo maligna melanoma, Leukemia, Leukemia, Lip and Oral Cavity
Cancer, Liposarcoma, Lung cancer, Luteoma, Lymphangioma, Lymphangiosarcoma,
Lymphoepithelioma, Lymphoid leukemia, Lymphoma, Macroglobulinemia, Malignant Fibrous
Histiocytoma, Malignant fibrous histiocytoma, Malignant Fibrous Histiocytoma of Bone,
Malignant Glioma, Malignant Mesothelioma, Malignant peripheral nerve sheath tumor,
Malignant rhabdoid tumor, Malignant triton tumor, MALT lymphoma, Mantle cell lymphoma,
Mast cell leukemia, Mediastinal germ cell tumor, Mediastinal tumor, Medullary thyroid cancer,
Medulloblastoma, Medulloblastoma, Medulloepithelioma, Melanoma, Melanoma, Meningioma,
Merkel Cell Carcinoma, Mesothelioma, Mesothelioma, Metastatic Squamous Neck Cancer with
Occult Primary, Metastatic urothelial carcinoma, Mixed Mullerian tumor, Monocytic leukemia,
Mouth Cancer, Mucinous tumor, Multiple Endocrine Neoplasia Syndrome, Multiple myeloma,
Mycosis Fungoides, Mycosis fungoides, Myelodysplastic Disease, Myelodysplasia Syndromes,
Myeloid leukemia, Myeloid sarcoma, Myeloproliferative Disease, Myxoma, Nasal Cavity
Cancer, Nasopharyngeal Cancer, Nasopharyngeal carcinoma, Neoplasm, Neurinoma,
Neuroblastoma, Neuroblastoma, Neurofibroma, Neuroma, Nodular melanoma, Non-Hodgkin
Lymphoma, Non-Hodgkin lymphoma, Nonmelanoma Skin Cancer, Non-Small Cell Lung
Cancer, Ocular oncology, Oligoastrocytoma, Oligodendroglioma, Oncocytoma, Optic nerve sheath meningioma, Oral Cancer, Oral cancer, Oropharyngeal Cancer, Osteosarcoma,
Osteosarcoma, Ovarian Cancer, Ovarian cancer, Ovarian Epithelial Cancer, Ovarian Germ Cell
Tumor, Ovarian Low Malignant Potential Tumor, Paget's disease of the breast, Pancoast tumor,
Pancreatic Cancer, Pancreatic cancer, Papillary thyroid cancer, Papillomatosis, Paraganglioma,
Paranasal Sinus Cancer, Parathyroid Cancer, Penile Cancer, Perivascular epithelioid cell tumor,
Pharyngeal Cancer, Pheochromocytoma, Pineal Parenchymal Tumor of Intermediate
Differentiation, Pineoblastoma, Pituicytoma, Pituitary adenoma, Pituitary tumor, Plasma Cell
Neoplasm, Pleuropulmonary blastoma, Polyembryoma, Precursor T-lymphoblastic lymphoma,
Primary central nervous system lymphoma, Primary effusion lymphoma, Primary Hepatocellular
Cancer, Primary Liver Cancer, Primary peritoneal cancer, Primitive neuroectodermal tumor,
Prostate cancer, Pseudomyxoma peritonei, Rectal Cancer, Renal cell carcinoma, Respiratory
Tract Carcinoma Involving the NUT Gene on Chromosome 15, Retinoblastoma, Rhabdomyoma,
Rhabdomyosarcoma, Richter's transformation, Sacrococcygeal teratoma, Salivary Gland Cancer,
Sarcoma, Schwannomatosis, Sebaceous gland carcinoma, Secondary neoplasm, Seminoma, Serous tumor, Sertoli-Leydig cell tumor, Sex cord-stromal tumor, Sezary Syndrome, Signet ring cell carcinoma, Skin Cancer, Small blue round cell tumor, Small cell carcinoma, Small Cell Lung Cancer, Small cell lymphoma, Small intestine cancer, Soft tissue sarcoma,
Somatostatinoma, Soot wart, Spinal Cord Tumor, Spinal tumor, Splenic marginal zone lymphoma, Squamous cell carcinoma, Stomach cancer, Superficial spreading melanoma, Supratentorial Primitive Neuroectodermal Tumor, Surface epithelial-stromal tumor, Synovial sarcoma, T-cell acute lymphoblastic leukemia, T-cell large granular lymphocyte leukemia, T- cell leukemia, T-cell lymphoma, T-cell prolymphocyte leukemia, Teratoma, Terminal lymphatic cancer, Testicular cancer, Thecoma, Throat Cancer, Thymic Carcinoma, Thymoma, Thyroid cancer, Transitional Cell Cancer of Renal Pelvis and Ureter, Transitional cell carcinoma, Urachal cancer, Urethral cancer, Urogenital neoplasm, Uterine sarcoma, Uveal melanoma, Vaginal Cancer, Verner Morrison syndrome, Verrucous carcinoma, Visual Pathway Glioma, Vulvar Cancer, Waldenstrom's macroglobulinemia, Warthin's tumor, Wilms' tumor, or any combination thereof.
[00164J In some embodiments, the methods or treatment regimen involve administering to a combination of a PI3Ka inhibitor and a taxane for the treatment of a cancer selected from the group consisting of lung cancer, breast cancer, endometrial cancer, ovarian cancer, bladder cancer, prostate cancer, neuroendocrine cancer, renal cancer, lymphoma, myeloma and leukemia. In some embodiments, the methods or treatment regimen involve administering to a combination of a PD a inhibitor and a taxane for the treatment of a cancer selected from the group consisting of breast cancer, non-small cell lung cancer (NSCLC), hormone refractory prostate cancer, gastric adenocaricoma, and squamous cell carcinoma of head and neck cancer.
[00165] In some embodiments, the disease or condition mediated by PI3-kinase is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer and endometrial cancer.
[00166] In some embodiments, the disease or condition mediated by PI3-kinase is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
[00167] In certain embodiments, the cancer is gastrointestinal cancer. In certain such embodiments, the cancer is selected from gastric cancer and gastroesophageal adenocarcinoma. In certain such embodiments, the cancer is gastric cancer. In certain such embodiments, the cancer is gastroesophageal adenocarcinoma.
[00168] In some embodiments, the cancer is gastric cancer. In some embodiments, the cancer is small cell lung cancer. In some embodiments, the cancer is non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is squamous non-small cell lung cancer. In some embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer. In some embodimdents, the disorder is non-small cell lung cancer and the subject has been identified with a PIK3CA mutation or amplification.
[00169] In some embodiments, the methods or treatment regimen involves administering a combination of a PI3 a inhibitor and a taxane for the treatment of solid tumors. Solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary. Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine. Additional exemplary solid tumors include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma,
lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastrointestinal system carcinomas, colon carcinoma, pancreatic cancer, breast cancer, genitourinary system carcinomas, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms1 tumor, cervical cancer, endocrine system carcinomas, testicular tumor, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma,
medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma.
[00170] In some embodiments, the methods or treatment regimen of the invention involves administering a combination of a PI3Ka inhibitor and a taxane for the treatment of multiple myeloma and/or Waldenstrom's macroglobulinemia.
[00171] In some embodiments, the methods or treatment regimen involves administering a combination of a PDKct inhibitor and a taxane for the treatment of renal cell carcinoma (also known as RCC or hypernephroma). Renal cell carcinoma is a kidney cancer that originates in the lining of the proximal convoluted tubule. Any known type of renal cell carcinoma may be treated using the treatment regimens of the invention, including clear renal cell carcinoma, papillary renal cell carcinoma, chromophobe renal cell carcinoma and collecting duct carcinoma. Any stage of the disease may be treated using the methods of the invention, including early stage as well as later stages (e.g. metastatic renal cell carcinoma).
[00172] Certain embodiments contemplate a human subject such as a subject that has been diagnosed as having or being at risk for developing or acquiring a proliferative disorder condition. Certain other embodiments contemplate a non-human subject, for example a non- human primate such as a macaque, chimpanzee, gorilla, vervet, orangutan, baboon or other non- human primate, including such non-human subjects that can be known to the art as preclinical models, including preclinical models for inflammatory disorders. Certain other embodiments contemplate a non-human subject that is a mammal, for example, a mouse, rat, rabbit, pig, sheep, horse, bovine, goat, gerbil, hamster, guinea pig or other mammal. There are also contemplated other embodiments in which the subject or biological source can be a non- mammalian vertebrate, for example, another higher vertebrate, or an avian, amphibian or reptilian species, or another subject or biological source. In certain embodiments of the present invention, a transgenic animal is utilized. A transgenic animal is a non-human animal in which one or more of the cells of the animal includes a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
[00173] In some embodiments, therapeutic efficacy is measured based on an effect of treating a proliferative disorder, such as cancer. In general, therapeutic efficacy of the methods and compositions of the invention, with regard to the treatment of a proliferative disorder (e.g.
cancer, whether benign or malignant), may be measured by the degree to which the methods and compositions promote inhibition of tumor cell proliferation, the inhibition of tumor
vascularization, the eradication of tumor cells, and/or a reduction in the size of at least one tumor such that a human is treated for the proliferative disorder. Several parameters to be considered in the determination of therapeutic efficacy are discussed herein. The proper combination of parameters for a particular situation can be established by the clinician. The progress of the inventive method in treating cancer (e.g., reducing tumor size or eradicating cancerous cells) can be ascertained using any suitable method, such as those methods currently used in the clinic to track tumor size and cancer progress. The primary efficacy parameter used to evaluate the treatment of cancer by the inventive method and compositions preferably is a reduction in the size of a tumor. Tumor size can be figured using any suitable technique, such as measurement of dimensions, or estimation of tumor volume using available computer software, such as FreeFlight software developed at Wake Forest University that enables accurate estimation of tumor volume. Tumor size can be determined by tumor visualization using, for example, CT, ultrasound, SPECT, spiral CT, MRI, photographs, and the like. In embodiments where a tumor is surgically resected after completion of the therapeutic period, the presence of tumor tissue and tumor size can be determined by gross analysis of the tissue to be resected, and/or by pathological analysis of the resected tissue.
[00174] In some embodiments, tumor size is reduced as a result of the inventive method preferably without significant adverse events in the subject. Adverse events are categorized or "graded" by the Cancer Therapy Evaluation Program (CTEP) of the National Cancer Institute (NCI), with Grade 0 representing minimal adverse side effects and Grade 4 representing the most severe adverse events. The NCI toxicity scale (published April 1999) and Common Toxicity Criteria Manual (updated August 1999) is available through the NCI, e.g., through the NCI internet website at www.ctep.info.nih.gov or in the Investigator's Handbook for participants in clinical trials of investigational agents sponsored by the Division of Cancer Treatment and Diagnosis, NCI. Desirably, the inventive method is associated with minimal adverse events, e.g. Grade 0, Grade 1, or Grade 2 adverse events, as graded by the CTEP/NCI.
[00175] As discussed herein, reduction of tumor size, although preferred, is not required in that the actual size of tumor may not shrink despite the eradication of tumor cells. Eradication of cancerous cells is sufficient to realize a therapeutic effect. Likewise, any reduction in tumor size is sufficient to realize a therapeutic effect.
[00176] Desirably, the growth of a tumor is stabilized (i.e., one or more tumors do not increase more than 1%, 5%, 10%, 15%, or 20% in size, and/or do not metastasize ) as a result of the inventive method and compositions. Such stabilization may be evidenced by a longer period of stable disease as characterized by the RECIST guidelines. In some embodiments, a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more weeks. In some embodiments, a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more months. In some embodiments, a tumor is stabilized for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more years. Preferably, the inventive method reduces the size of a tumor at least about 5%
(e.g., at least about 10%, 15%, 20%, or 25%). More preferably, tumor size is reduced at least about 30% (e.g., at least about 35%, 40%, 45%, 50%, 55%, 60%, or 65%). Even more preferably, tumor size is reduced at least about 70% (e.g., at least about 75%, 80%, 85%, 90%, or 95%). In some embodiments, tumor regrowth is not detected to at least 20, 25, 30, 40, 45, 50,
60, 70 days or even longer after the first administration of a P13 a inhibitor and a taxane. Most preferably, the tumor is completely eliminated, or reduced below a level of detection. In some embodiments, a subject remains tumor free (e.g. in remission) for at least about 1, 2, 3, 4, 5, 6, 7,
8, 9, 10, 11, 12, or more weeks following treatment. In some embodiments, a subject remains tumor free for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more months following treatment. In some embodiments, no detectable amount of tumor is found in the subject for at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more years after the treatment.
[00177] When a tumor is subject to surgical resection following completion of the therapeutic period, the efficacy of the inventive method in reducing tumor size can be determined by measuring the percentage of resected tissue that is necrotic (i.e., dead). In some embodiments, a treatment is therapeutically effective if the necrosis percentage of the resected tissue is greater than about 20% (e.g., at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%), more preferably about 90% or greater (e.g., about 90%, 95%, or 100%). Most preferably, the necrosis percentage of the resected tissue is 100%, that is, no tumor tissue is present or detectable.
[00178] A number of secondary parameters can be employed to determine the efficacy of the inventive method. Examples of secondary parameters include, but are not limited to, detection of new tumors, detection of tumor antigens or markers (e.g., CEA, PSA, or CA-125), biopsy, surgical downstaging (i.e., conversion of the surgical stage of a tumor from unresectable to resectable), PET scans, survival, disease progression-free survival, time to disease progression, quality of life assessments such as the Clinical Benefit Response Assessment, and the like, all of which can point to the overall progression (or regression) of cancer in a human. Biopsy is particularly useful in detecting the eradication of cancerous cells within a tissue.
Radioimmunodetection (RAID) is used to locate and stage tumors using serum levels of markers (antigens) produced by and/or associated with tumors ("tumor markers" or "tumor-associated antigens"), and can be useful as a pre-treatment diagnostic predicate, a post-treatment diagnostic indicator of recurrence, and a post-treatment indicator of therapeutic efficacy. Examples of tumor markers or tumor-associated antigens that can be evaluated as indicators of therapeutic efficacy include, but are not limited to, carcinembryonic antigen (CEA) prostate-specific antigen (PSA), CA-125, CA19-9, ganglioside molecules (e.g., GM2, GD2, and GD3), MART-1, heat shock proteins (e.g., gp96), sialyl Tn (STn), tyrosinase, MUC-1, HER-2/neu, c-erb-B2, KSA, PSMA, p53, RAS, EGF-R, VEGF, MAGE, and gplOO. Other tumor-associated antigens are known in the art. RAID technology in combination with endoscopic detection systems also efficiently distinguishes small tumors from surrounding tissue (see, for example, U.S. Pat. No. 4,932,412).
[00179] Desirably, in accordance with the inventive method, the treatment of cancer in a human patient is evidenced by one or more of the following results: (a) the complete disappearance of a tumor (i.e., a complete response), (b) about a 25% to about a 50% reduction in the size of a tumor for at least four weeks after completion of the therapeutic period as compared to the size of the tumor before treatment, (c) at least about a 50% reduction in the size of a tumor for at least four weeks after completion of the therapeutic period as compared to the size of the tumor before the therapeutic period, (d) at least a 2% decrease (e.g., about a 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% decrease) in a specific tumor-associated antigen level at about 4-12 weeks after completion of the therapeutic period as compared to the tumor-associated antigen level before the therapeutic period or (e) a longer period of stable disease, for example longer by 1, 2, 3, 4, or 5 months. While at least a 2% decrease in a tumor-associated antigen level is preferred, any decrease in the tumor-associated antigen level is evidence of treatment of a cancer in a patient by the inventive method. For example, with respect to unresectable, locally advanced pancreatic cancer, treatment can be evidenced by at least a 10% decrease in the CA19- 9 tumor-associated antigen level at 4-12 weeks after completion of the therapeutic period as compared to the CA19-9 level before the therapeutic period. Similarly, with respect to locally advanced rectal cancer, treatment can be evidenced by at least a 10% decrease in the CEA tumor-associated antigen level at 4-12 weeks after completion of the therapeutic period as compared to the CEA level before the therapeutic period.
[00180] With respect to quality of life assessments, such as the Clinical Benefit Response Criteria, the therapeutic benefit of the treatment in accordance with the invention can be evidenced in terms of pain intensity, analgesic consumption, and/or the Karnofsky Performance Scale score. The Karnofsky Performance Scale allows patients to be classified according to their functional impairment. The Karnofsky Performance Scale is scored from 0-100. In general, a lower Karnofsky score is predictive of a poor prognosis for survival. Thus, the treatment of cancer in a human patient alternatively, or in addition, is evidenced by (a) at least a 50% decrease (e.g., at least a 60%, 70%, 80%, 90%, or 100% decrease) in pain intensity reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of treatment, as compared to the pain intensity reported by the patient before treatment, (b) at least a 50% decrease (e.g., at least a 60%, 70%, 80%, 90%, or 100% decrease) in analgesic consumption reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of treatment as compared to the analgesic consumption reported by the patient before treatment, and/or (c) at least a 20 point increase (e.g., at least a 30 point, 50 point, 70 point, or 90 point increase) in the Karnofsky Performance Scale score reported by a patient, such as for any consecutive four week period in the 12 weeks after completion of the therapeutic period as compared to the Karnofsky Performance Scale score reported by the patient before the therapeutic period.
[00181] The treatment of a proliferative disorder (e.g. cancer, whether benign or malignant) in a human patient desirably is evidenced by one or more (in any combination) of the foregoing results, although alternative or additional results of the referenced tests and/or other tests can evidence treatment efficacy.
[00182] Detection, monitoring, and rating of various cancers in a human are further described in Cancer Facts and Figures 2001, American Cancer Society, New York, N.Y., and International Patent Application WO 01/24684. Accordingly, a clinician can use standard tests to determine the efficacy of the various embodiments of the inventive method in treating cancer. However, in addition to tumor size and spread, the clinician also may consider quality of life and survival of the subject in evaluating efficacy of treatment.
[00183] In some embodiments, administration of a taxane at least 24 hours prior to the administration of a PI3 a inhibitor provides improved therapeutic efficacy over a treatment wherein the taxane and PI3 a inhibitor are administered simultaneously. Improved efficacy may be measured using any method known in the art, including but not limited to those described herein. In some embodiments, the improved therapeutic efficacy is an improvement of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, 110%, 120%, 150%, 200%, 300%, 400%, 500%, 600%, 700%, 1000%, 10000% or more, using an appropriate measure (e.g., inhibiting PI3 a signaling tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival). Improved efficacy may also be expressed as fold improvement, such as at least about 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60- fold, 70-fold, 80-fold, 90-fold, 100-fold, 1000-fold, 10000-fold, or more, using an appropriate measure (e.g. tumor size reduction, duration of tumor size stability, duration of time free from metastatic events, duration of disease-free survival).
Pharmaceutical compositions and administration
[00184] The invention provides, in one aspect, a combination treatment utilizing a PI3 a inhibitor and a taxane. The therapeutic agents (including compounds) that are provided for use in the combination therapies of the invention can be administered simultaneously or separately.
This administration in combination includes, for example, simultaneous administration of two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. For example, multiple therapeutic agents can be formulated together in the same dosage form and administered simultaneously. Alternatively multiple therapeutic agents can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa. In the separate administration protocol, a compound of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart. The term "combination treatments" also embraces the administration of the therapeutic agents as described herein in further combination with other biologically active compounds or ingredients and non-drug therapies (e.g., surgery or radiation treatment).
[00185] Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action. An effective amount of an inhibitor of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer. Sequential or substantially simultaneous administration of each inhibitor or therapeutic agent can be effected by any appropriate route as noted above and including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination (e.g. in some embodiments a taxane) selected may be administered by intravenous injection while the other therapeutic agent (a PI3 a inhibitor) of the combination may be administered orally.
Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
[00186] In some embodiments, adrninistration of the compounds of the invention can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell or tissue being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
[00187] The amount of each compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg kg/day, in single or divided doses. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g., by dividing such larger doses into several small doses for administration throughout the day. [00188] In some embodiments, a combination treatment of the invention is administered in a single dose comprising at least one PD ct inhibitor and at least one taxane. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a combination treatment of the invention may also be used for treatment of an acute condition.
[00189] The subject pharmaceutical compositions can be administered as a combination of a PI3Ka inhibitor and a taxane, or in further combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions. Where desired, the subject combinations and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
[00190] A pharmaceutical composition of the invention typically contains an active ingredient (e.g., a compound) of the present invention or a pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including but not limited to inert solid diluents and fillers, diluents, sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
[00191] Described below are non-limiting exemplary pharmaceutical compositions and methods for preparing the same.
Pharmaceutical compositions for oral administration.
[00192] In some embodiments, the invention provides a pharmaceutical composition for oral administration containing a compound of the invention, and a pharmaceutical excipient suitable for oral administration.
[00193] In some embodiments, the invention provides a solid pharmaceutical composition for oral administration containing: (i) a compound which is a PI3Ka inhibitor; (ii) a second compound which is a taxane; and (iii) a pharmaceutical excipient suitable for oral
administration. In some embodiments, the composition further contains: (iv) a third agent or even a fourth agent. In some embodiments, each compound or agent is present in a
therapeutically effective amount. In other embodiments, one or more compounds or agents is present in a sub-therapeutic amount, and the compounds or agents act synergistically to provide a therapeutically effective pharmaceutical composition.
[00194] In some embodiments, the invention provides for a pharmaceutical composition comprising a combination of a PI3Ka inhibitor and a taxane. The PI3-kinase a inhibitor and the taxane can be packaged as a single oral dosage form. In other embodiments, the PI3Ka inhibitor and the taxane inhibitor can be packaged as separate dosage forms, such as a tablet. [00195] In one embodiment, the present invention provides an oral dosage form comprising 100 mg to 1.5g of an inhibitor of the invention. The oral dosage form can be a tablet, formulated in form of liquid, in immediate or sustained release format.
[00196] In some embodiments, the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption. Pharmaceutical compositions of the invention suitable for oral adniinistration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or nonaqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion. Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation. For example, a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients.
Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[00197] This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds. For example, water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and or storage is expected. An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs. [00198] An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration. In preparing the compositions for an oral dosage form, any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro- crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose. For example, suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
[00199] Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, com starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
[00200] Examples of suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
[00201] Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art. About 0.5 to about 15 weight percent of disintegrant, or about 1 to about
5 weight percent of disintegrant, may be used in the pharmaceutical composition. Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, hydroxypropyl cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
[00202] Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof. Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, colloidal silicon dioxide, or mixtures thereof. A lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
[00203] When aqueous suspensions and/or elixirs are desired for oral administration, the active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
[00204] The tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
[00205] Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
[00206] A suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10.
An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance ("HLB" value).
Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions. Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable. Similarly, lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10. However, HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
[00207] Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids,
oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acyl lactylates; mono- and di-acetylated tartaric acid esters of mono- and di- glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
[00208] Within the aforementioned group, ionic surfactants include, by way of example:
lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di- glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
[00209] Ionic surfactants may be the ionized forms of lecithin, lysolecithin,
phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine,
lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG- phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate, caprate, laurate, myristate, palmitate, oleate, ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl sulfate, docusate, lauroyl carnitines, palmitoyl carnitines, myristoyl carnitines, and salts and mixtures thereof.
[00210] Hydrophilic non-ionic surfactants may include, but are not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterols, derivatives, and analogues thereof; polyoxyethylated vitamins and derivatives thereof; polyoxyethylene-polyoxypropylene block copolymers; and mixtures thereof; polyethylene glycol sorbitan fatty acid esters and hydrophilic transesterification products of a polyol with at least one member of the group consisting of triglycerides, vegetable oils, and hydrogenated vegetable oils. The polyol may be glycerol, ethylene glycol, polyethylene glycol, sorbitol, propylene glycol, pentaerythritol, or a saccharide.
[00211] Other hydrophilic-non-ionic surfactants include, without limitation, PEG- 10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG- 15 stearate, PEG-32 distearate, PEG-40 stearate, PEG- 100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl laurate, PEG-40 glyceryl laurate, PEG-40 palm kernel oil, PEG-50 hydrogenated castor oil, PEG-40 castor oil, PEG-35 castor oil, PEG-60 castor oil, PEG-40 hydrogenated castor oil, PEG-60 hydrogenated castor oil, PEG-60 corn oil, PEG-6 caprate caprylate glycerides, PEG-8 caprate/caprylate glycerides, polyglyceryl-10 laurate, PEG-30 cholesterol, PEG-25 phyto sterol, PEG-30 soya sterol, PEG-20 trioleate, PEG-40 sorbitan oleate, PEG-80 sorbitan laurate, polysorbate 20, polysorbate 80, POE-9 lauryl ether, POE-23 lauryl ether, POE-10 oleyl ether, POE-20 oleyl ether, POE-20 stearyl ether, tocopheryl PEG- 100 succinate, PEG-24 cholesterol, polyglyceryl-lOoleate, Tween 40, Tween 60, sucrose monostearate, sucrose monolaurate, sucrose monopalmitate, PEG 10-100 nonyl phenol series, PEG 15-100 octyl phenol series, and poloxamers.
[00212] Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil- soluble vitarnms/vitamin derivatives; and mixtures thereof. Within this group, preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
[00213] In one embodiment, the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection. A solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
[00214] Examples of suitable solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol,
hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofiirfuryl alcohol PEG ether (glycofurol) or methoxy PEG ; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone, ε-caprolactam,
N-alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide and polyvinylpyrrolidone; esters such as ethyl propionate, tributylcitrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, ε-caprolactone and isomers thereof, δ-valerolactone and isomers thereof, β-butyrolactone and isomers thereof; and other solubilizers known in the art, such as dimethyl acetamide, dimethyl isosorbide, N-methyl pyrrolidones, monooctanoin, diethylene glycol monoethyl ether, and water.
[00215] Mixtures of solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone,
N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose,
hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
[00216] The amount of solubilizer that can be included is not particularly limited. The amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art. In some circumstances, it may be advantageous to include amounts of solubilizers far in excess of bioacceptable amounts, for example to maximize the concentration of the drug, with excess solubilizer removed prior to providing the composition to a subject using conventional techniques, such as distillation or evaporation. Thus, if present, the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients. If desired, very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less. Typically, the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
[00217] The composition can further include one or more pharmaceutically acceptable additives and excipients. Such additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
[00218] In addition, an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons. Examples of pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like. Also suitable are bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like. Salts of polyprotic acids, such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used. When the base is a salt, the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like. Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
[00219] Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like. Examples of suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid and the like.
Pharmaceutical compositions for injection.
[00220] In some embodiments, the invention provides a pharmaceutical composition for injection containing at least one compound of the present invention and a pharmaceutical excipient suitable for injection. For example a pharmaceutical composition for injection is provided comprising at least one PI3 ct inhibitor and at least one taxane. Also provided are pharmaceutical compositions comprising a PI3Ka inhibitor, and pharmaceutical compositions comprising a taxane, where the PI3Ka inhibitor is administered separately or together with the taxane. The PI3 a inhibitor and the taxane may be formulated separately, and may further include a third therapeutic agent. Components and amounts of agents in the compositions are as described herein. The forms in which the compositions of the present invention may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
[00221] Aqueous solutions in saline are also conventionally used for injection. Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Pharmaceutical compositions for topical (e.g.. transdermal) delivery [00222] In some embodiments, the invention provides a pharmaceutical composition for transdermal delivery containing at least one compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery. For example a pharmaceutical composition for topical delivery is provided comprising at least one PI3Ka inhibitor and at least one taxane. Also provided are pharmaceutical compositions for topical delivery comprising a PI3 a inhibitor, and pharmaceutical compositions for topical delivery comprising a taxane, where the PB a inhibitor is administered separately or together with the taxane. The PI3 a inhibitor and the taxane may be formulated separately, and may further include a third therapeutic agent.
[00223] Compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical acUninistration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions. In general, carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients. In contrast, a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
[00224] The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin. There are many of these penetration-enhancing molecules known to those trained in the art of topical formulation. Examples of such carriers and excipients include, but are not limited to, humectants (e.g., urea), glycols (e.g., propylene glycol), alcohols (e.g., ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), pyrrolidones, glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
[00225] Another exemplary formulation for use in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts, either with or without another agent.
[00226] The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Pharmaceutical compositions for inhalation
[00227] Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner. For example a pharmaceutical composition for respiratory delivery is provided comprising at least one PI3Ka inhibitor and a taxane. Also provided are
pharmaceutical compositions for respiratory delivery comprising a PI3 a inhibitor, and pharmaceutical compositions for respiratory delivery comprising a taxane, where the PI3Ka inhibitor is administered separately or together with the taxane. Compositions comprising a PI3 a inhibitor and a taxane may be formulated separately, and may further include a third therapeutic agent.
Other pharmaceutical compositions
[00228] Pharmaceutical compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art. See, e.g., See, e.g., Anderson, Philip O.; noben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, New York, 1990; atzung, ed., Basic and Clinical
Pharmacology, Ninth Edition, McGraw Hill, 20037ybg; Goodman and Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw Hill, 2001; Remingtons Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000; Martindale, The Extra Pharmacopoeia, Thirty-Second Edition (The Pharmaceutical Press, London, 1999); all of which are incorporated by reference herein in their entirety.
[00229] Administration of the compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g. transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also abe administered intraadiposally or intrathecally.
[00230] In some embodiments, a compound of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a compound of the invention may also be used for treatment of an acute condition.
[00231] In some embodiments, a compound of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
[00232] Administration of the compounds of the invention may continue as long as necessary. In some embodiments, a compound of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, a compound of the invention is aa'ministered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a compound of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[00233] An effective amount of a compound of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
[00234] The compounds of the invention may be aaministered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
[00235] When a compound of the invention is administered in a composition that comprises one or more agents, and the agent has a shorter half-life than the compound of the invention unit dose forms of the agent and the compound of the invention may be adjusted accordingly.
[00236] The subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
[00237] Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
[00238] The invention also provides kits. The kits include the compounds of the present invention as described herein, in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like. Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. The kit may further contain another agent. In some embodiments, the compounds of the present invention and the agent are provided as separate compositions in separate containers within the kit. In some embodiments, the compound of the present invention and the agent are provided as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like) are known in the art and may be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
Further Combination therapies
[00239] The present invention also provides methods for further combination therapies in which, in addition to an PDKct inhibitor and a taxane, one or more agents known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes is used or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In one aspect, such therapy includes but is not limited to the combination of the composition comprising at least one PBKa inhibitor and at least one taxane, as described herein, with one or more additional therapeutic agents such as anticancer agents,
chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide, where desired, a synergistic or additive therapeutic effect. Pathways that may be targeted by administering another agent include, but are not limited to, MAP kinase, Akt, NFkB, WNT, RAS/ RAF7MEK/ERK, JNK/SAP , p38 MAPK, Src Family Kinases, JAK/STAT and/or PKC signaling pathways. Other agents may target one or more members of one or more signaling pathways. Representative members of the nuclear factor-kappaB (NFkB) pathway include but are not limited to RelA (p65), RelB, c-Rel, p50/pl05 (NF-κΒ 1), p52/p 100 (NF-KB2), IkB, and IkB kinase. Non-limiting examples of receptor tyrosine kinases that are members of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway that may be targeted by one or more agents include FLT3 LIGAND, EGFR, IGF-1R, HER2/neu, VEGFR, and PDGFR. Downstream members of the PI3K/AKT pathway that may be targeted by agents according to the methods of the invention include, but are not limited to, forkhead box O transcription factors, Bad, GSK-3P, I-KB, mTOR, MDM-2, and S6 ribosomal subunit.
[00240] This invention further relates to a method for using the compounds or pharmaceutical composition in combination with other tumor treatment approaches, including surgery, ionizing radiation, photodynamic therapy, or implants, e.g., with corticosteroids, hormones, or used as radiosensitizers.
[00241] One such approach may be, for example, radiation therapy in inhibiting abnormal cell growth or treating the proliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compounds of the invention in this combination therapy can be determined as described herein.
[00242] Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term "brachytherapy," as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g., At-211, 1-131, 1-125, Y-90,
Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as 1-125, 1-131,
Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
[00243] Without being limited by any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal the combination of a taxane and a PI3 a inhibitor of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which combined amounts are effective in sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
[00244] Photodynamic therapy includes therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as e.g., VISUDYNE and porfuner sodium. Angiostatic steroids include compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortexolone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
[00245] Implants containing corticosteroids include compounds, such as e.g., fluocinolone and dexamethasone. Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
EXAMPLES
Example 1. Evaluation of the Combination of Docetaxel and Compound A in NCI-H1048 and cells in vitro
[00246] The objective of the study: to determine the pharmacodynamic effects of compound A in combination with docetaxel in NCI-H1048 cells in vitro and compare pre-dosing effect of both drugs dosed on three different schedules: concomitant simultaneous vs 24hr pre-dose of either compound A or docetaxel. [00247] Compound A was prepared according to methods disclosed in WO 2011/022439 and WO 2013/071272. Compound A was dissolved in DMSO at 10 mM concentration, and docetaxel was dissolved in DMSO at 10 mM concentration.
[00248] NCI-H1048 cells were plated in 6-well plates with 750,000 cells/well in 1ml of HITES growth media supplemented with 5% of fetal bovine serum (FBS) and allowed to adhere overnight. Cells were treated with 3 μΜ compound A, lOnM docetaxel or ΙμΣ, DMSO for controls and allowed to incubate for 2 hr, 24 hr, or 48 hr. After the timecourse, media was removed by aspiration and cells were washed with 1 mL ice cold DPBS. 120μΙ, complete lysis buffer (M-PER lysis buffer supplemented with 25 μΜ NaF, 1 μΜ NaOrtho Vanadate, 25 μΜ β- GP, and protease inhibitor cocktail tablet) was then added to cells. Cells were scraped and the supernate was transferred to a chilled 1.5 mL eppendorf tube. Cells were placed at -80 °C overnight. Cell lysates were thawed and spun at 14000RPM for 10 minutes at 4 °C. The supernatant was then transferred to a new 1.5 mL eppendorf tube. The supernatants were assayed for protein concentration using BCA Protein Assay kit (Thermo Scientific). Samples were mixed with 4X NuPAGE LDS Sample Buffer (Life Technologies ) and lOXNuPAGE Sample Reducing Agent (Life Technologies), heated at 70 °C for 10 minutes, and 7 μg of proteins were loaded onto a 12% Tris-Glycine SDS-PAGE Midi gels (Life Technologies). Standard semi-dry blotting techniques were used to transfer proteins to the PVDF membranes. Membranes were incubated with primary antibody (Cell Signaling Technologies: pAKT (T308), pAKT (S473), pS6 (S240/244), p4EBPl (S65), Cleaved PARP and Cleaved Lamin A at 4 °C. Blots were washed with TBST and incubated with fluorescently-labeled secondary antibody AlexaFluor 680 Goat Anti-Rabbit IgG (H+L) (Life Technologies) for 1 hr at RT. Membranes were dried and imaged using Odyssey LI-COR Infrared Imager /scanner) (LI-COR Inc). Li-cor Odyssey Software 2.1 was used to quantitate the proteins/PD markers on the Western blots. Western blot analysis of tumor samples
[00249] NCI-H1048 tumors approximately 400 -500 mm3 in size were collected at 2, 24 and 48 h post last dose and frozen in liquid nitrogen. Tumor chunks were homogenized in 700 Lof M- PER lysis buffer supplemented with 25 μΜ NaF, 1 μΜ NaOrtho Vanadate, 25 μΜ β-GP, and protease inhibitor cocktail tablets. The supernatants were assayed for protein concentration using a BCA Protein Assay kit (Thermo Scientific). Samples were mixed with 4X NuPAGE LDS Sample Buffer (Life Technologies ) and lOXNuPAGE Sample Reducing Agent (Life Technologies), heated at 70 °C for 10 minutes, and 20 μg of proteins were loaded onto a 12% Tris-Glycine SDS-PAGE Midi gels (Life Technologies.) Standard semi-dry blotting techniques were used to transfer the protein to the PVDF membranes. Membranes were incubated with primary antibody (Cell Signaling Technologies: pAKT (T308), pAKT (S473), pS6 (S240/244), p4EBPl (S65), Cleaved PARP and Cleaved Lamin A overnight at 4°C overnight. Blots were washed with TBST and incubated with fluorescently-labeled secondary antibody AlexaFluor 680 Goat Anti-Rabbit IgG (H+L) (Life Technologies) for 1 hr at RT. Membranes were imaged using Odyssey LI-COR Infrared Imager /scanner) (LI-COR Inc). Li-cor Odyssey Software 2.1 was used to quantitate the proteins/PD markers on the Western blots.
Example 2. Evaluation of the Docetaxel in the Gastric human tumor xenografts Hs746T
[00250] The objective of the study was to determine the pharmacodynamic effect of docetaxel in Hs746T gastric xenograft tumor samples.
[00251] Hs746T tumors tumors approximately 600 mm3 in size were collected at 2, 7, 24 and 48 h post last dose and frozen in liquid nitrogen. Tumor chunks were homogenized in ~ 800 μίοί M-PER lysis buffer supplemented with 25 μΜ NaF, 1 μΜ NaOrtho Vanadate, 25 μΜ β- GP, and protease inhibitor cocktail tablets. The supernatants were assayed for protein concentration using a BCA Protein Assay kit (Thermo Scientific). Samples were mixed with 4X NuPAGE LDS Sample Buffer (Life Technologies ) and lOXNuPAGE Sample Reducing Agent (Life Technologies), heated at 70 °C for 10 minutes, and 20 μg of proteins were loaded onto a 12% Tris-Glycine SDS-PAGE Midi gels (Life Technologies.). Standard semi-dry blotting techniques were used to transfer the protein to the PVDF membranes. Membranes were incubated with primary antibody (Cell Signaling Technologies: pAKT (T308), pAKT (S473), pS6 (S240/244), p4EBPl (S65) at 4 °C overnight. Blots were washed with TBST and incubated with fluorescently-labeled secondary antibody AlexaFluor 680 Goat Anti-Rabbit IgG (H+L) (Life Technologies) for 1 hr at RT. Membranes were imaged using Odyssey LI-COR Infrared Imager /scanner) (LI-COR Inc). Li-cor Odyssey Software 2.1 was used to quantitate the proteins/PD markers on the Western blots.
Example 3. Evaluation of the Combination of Docetaxei and Compound A in NCI-H1048 and GA0098 xenograft models in vivo
[00252] Objectives of this study: to determine the in vivo antitumor activity of compound A in combination with docetaxel in female Balb/c Nude mice bearing NCI-H1048 human lung cell carcinoma xenografts and in female Balb/c Nude mice bearing GA0098 human gastric carcinoma xenografts.
[00253] Compound A was prepared according to methods disclosed in WO 2011/022439 and WO 2013/071272. Compound A was formulated in 100% PEG400 and stored at approximately 25 °C, shielded from light. The control/vehicle used in this study was 100% PEG400 + 0.9% saline.
[00254] NCI-H1048 cells were grown in HITES Medium supplemented with 5% fetal bovine serum (FBS) until approximately 80% confluence was reached. Prior to injection, the cells were detached with Trypsin (Invitrogen), washed twice with phosphate buffered saline (PBS), and re- suspended in KITES media (Invitrogen,) without supplements. NCI-H1048 cells were resuspended to a final concentration of 4.0 x 107cells/mL in HITES media without supplements, with 50% Matrigel (BD Biosciences).
Eight week old female Balb/c Nude mice were inoculated SC in right flank (cell suspension in serum free HITES and 50% Matrigel) with 4.0 x 106 NCI-H1048 cells. Tumor growth was monitored with vernier calipers. The mean TV (MTV) was calculated using the formula V = W2 x L /2
[00255] Body weight and the tumor growth were monitored twice a week. Tumor size was measured to the nearest 0.1 mm using vernier caliper and applying the formula V = W2 x 172, where V = volume, W = width, and L = length for the tumor xenograft. Xenografts were allowed to grow until they reached an average size of approximately 170 mm3 after 8 days. Mice bearing the proper size xenograft were randomly assigned into one of the treatment groups (n=7/group) shown in Table 1 and began to be treated with their assigned test materials, either vehicle (100% PEG400 + 0.9% saline), compound A (at 140 mg kg on QDX3 schedule via oral gavage, PO), docetaxel (at 5mg/kg, on QW schedule, intravenously IV), or combination of compound A with docetaxel over a 21 day treatment period. The percentage of tumor growth inhibition (TGI) and percent body weight (BW) change were determined on Day 21 (see Table 1 for details). Statistical comparisons of tumor growth between the treatment and vehicle groups were conducted using a linear mixed effects regression analysis on the change in area under the tumor volume (TV)-versus— time curves (AAUC) values (see Table 1 for details). All p values less than 0.05 were considered significant.
Table 1 Study Design and Noteworthy Findings for Compound A and Docetaxel
Figure imgf000071_0001
Figure imgf000072_0001
(24-hr pre- Nude Maximum 4.5% (Day dose) Days 1 , 8, 15 Mean 14)
%BWLC
AAUC = change in areas under the tumor volume-versus-time curves; BIW = twice weekly; BWL body wieght loss; IP =
aDose volumes for PO and IP administration were 5 mL kg body weight. Dose volume for IV administation was 10 mL/kg body
bTGI values were calculated on Day 21 post treatment initiation.
cMaximum mean percent BWL between Day 0 to Day 21
dAAUC=Statistical analysis was performed with a linear mixed effects regresssion model. A p value of <0.05 was considered.
Synergistic analysis: p>0.05=additive; p<0.05 and score <0=synergistic; p<0.05, score >0, and the combination growth rate is lower than both the single agent growth rates = subadditive; p<0.05, score >0, and the combination growth rate is higher than at least one of the single agent growth rates = antagonistic. P values <0.05 were considered statistically significant. Calculated on Day 21 post treatment initiation.
[00256] The results of this study are summarized by the following figures. Figures 5A and 5B show the increased apoptosis observed in vivo in NCI-H1048 when pre-dosing docetaxel in combination with compound A. Figure 5A shows quantified Cleaved lamin A and Figure 5B shows quantified cleaved PARP.
[00257] Figures 6A and 6B show the effect that pre-dosing docetaxel in combination with compound A has on in vivo antitumor activity compared to simultaneous combination dosing in NCI-H1048 cells. Figure 6A shows the effect of pre-dosing docetaxel at low clinically relevant doses while Figure 6B shows the effect of pre-dosing docetaxel at 10 mg/kg. Pre-dosing docetaxel in combination with compound A results in improved or equal antitumor activity compared to simultaneous combination dosing. Furthermore, Figure 6A shows that the pre- dosing effect of docetaxel is more pronounced at low clinically relevant doses for docetaxel. These results demonstrate that administration of compound A on an intermittent schedule in combination with 5mg kg of docetaxel is efficacious in reducing tumor size independently of order of aoministration of both drugs. The concomitant combination of compound A with docetaxel resulted in additive efficacy, while pre-dosing with 5mg/kg of docetaxel for 24 hr followed by compound A on intermittent schedule (QDx3) showed more robust anti-tumor activity and strong synergistic effect (see Table 2 for details).
[00258] Figure 7 shows the effect that pre-dosing docetaxel in combination with compound A has on enhanced tumor growth delay (compound A: 140 mg/kg QD3; docetaxel: 10 mg/kg QW). Figure 7 shows the effect on NCI-H1048 cells where tumor regrowth is suppressed for at least 20, 30 or even 40 days. Combination Comparisons (Log-Transformed) for Compound A and
Docetaxel
Figure imgf000074_0001
IV - intravenously; QD = daily; QW = weekly; PO = orally; SEM - standard error of the mean.
Synergistic analysis: p > 0.05 = additive; p < 0.05 and score < 0 = synergistic; p < 0.05, score
> 0, and the combination growth rate is lower than both the single agent growth rates
= subadditive; p < 0.05, score > 0, and the combination growth rate is higher than at least 1 of the single agent growth rates = antagonistic. A p value < 0.05 was considered statistically significant.
[00259] 4-6 week female BALB/C Nude mice were inoculated subcutaneously in flank (trocar) with 3mm x 3mm GA0098 Gastric Cancer tumor fragments. Tumor growth was monitored with vernier calipers and the mean tumor volume (MTV) was calculated using the formula (0.5 x [length x width2]). When the MTV reached approximately 193mm3, animals were randomized into treatment groups (n =8/group) and dosed according to study design shown in Table 3.
[00260] Body weight and the tumor growth were monitored twice a week. On day 1 mice began treatment with assigned test materials, either vehicle (100% PEG400 + 0.9% saline), compound A (at 140 mg/kg on QD x3 schedule via oral gavage, PO), docetaxel (at 10 mg/kg, on QW schedule, intravenously IV), or a combination of compound A with docetaxel over a 21 day treatment period. The percentage of tumor growth inhibition (TGI) and percent body weight (BW) change were determined on Day 21. Statistical comparisons of tumor growth between the treatment and vehicle groups were conducted using a linear mixed effects regression analysis on the change in area under the tumor volume (TV)-versus- time curves (AAUC) values (see Table 3 for details). All p values less than 0.05 were considered significant. Table 3: Study Design and Noteworthy Findings for Compound A and Docetaxel in GA0098 model
Method of Sex/Numb
Administratio er Per Noteworthy
Treatment Dose n/ Frequency Group Species/ Endpoints Findings
(mg kg
Group ) Strain
PEG400 + NA PO/(QDx21) Female/8 Mus TG N/A
0.9%saline NA rV/(QWx3), musculu Maximum 0%, (Day 0) days 1, 8 ,15 s Mean
%BWLb
Compound A 140 PO/(QDx3) Female/8 Mus TGI 38.20%
Days 1-3, 8- musculu AAUCC 45.2; p 10, 15-17 s <0.001
Maximum 0%, (Day 0)
Mean
%BWL
Docetaxel 10 rV/(QWx3) Female/8 Mus TGI 48.20% days 1, 8, 15 musculu AAUC 32.2;
s PO.001
Maximum 6.5%, (Day
Mean 21)
%BWL
Compound A 140 PO/(QDx3) Female/8 Mus TGI 65.80%
+ Days 1-3, 8- musculu AAUC 69.1;
10, 15-17 s P<0.001
Docetaxel; 10 rV/(QWx3) Synergy Additive analysis'1
Concomitant Days 1,8,15 Maximum 9.4%, (Day dosing Mean 17)
%BWL
Compound A 140 PO/(QDx3) Female/8 Mus TGI 79.80%
+ Days 2-4, 9- musculu AAUC 100.2;
11, 16-18 s PO.001
Docetaxel (24 10 IV/(QWx3) Synergy Additive hr predosed); analysis'1
Sequential Maximum 8.8%, (Day dosing Mean 10)
Days 1,8,15 %BWL
AAUC = change in areas under the tumor volume-versus-time curves; BWL body wieght loss; aTGI values were calculated on Day 21 post treatment initiation.
bMaximum mean percent BWL between Day 0 to Day 21.
cAAUC=Statistical analysis was performed with a linear mixed effects regresssion model. A p value of <0.05 was considered. Synergistic analysis: pX).05=additive; p<0.05 and score <0=synergistic; p<0.05, score >0, and the combination growth rate is lower than both the single agent growth rates = subadditive; p<0.05, score >0, and the combination growth rate is higher than at least one of the single agent growth rates = antagonistic. P values <0.05 were considered statistically significant. Calculated on Day 21 post treatment initiation.
[00261] The results of this study are summarized in Figure 8 which shows the effect that pre- dosing docetaxel for 24h in combination with compound A has on in vivo antitumor activity compared to simultaneous combination dosing in GA0098 xenografts. These results demonstrate that administration of compound A on an intermittent schedule in combination with 10 mg kg of docetaxel is efficacious in reducing tumor size in both concomitant and sequential combination treatment groups. Furthermore, pre-dosing docetaxel in combination with compound A results in significantly improved antitumor activity compared to simultaneous combination dosing (TGI 79.8% vs 65.8%). The concomitant and sequential combination of compound A with docetaxel resulted in additive efficacy in this model (see Table 4 for details).
Table 4 Combination Comparisons (Log-Transformed) for Compound A and Docetaxel
Figure imgf000076_0001
IV = intravenously; PO = orally; SEM = standard error of the mean.
Synergistic analysis: p > 0.05 = additive; p < 0.05 and score < 0 = synergistic; p < 0.05, score
> 0, and the combination growth rate is lower than both the single agent growth rates
= subadditive; p < 0.05, score > 0, and the combination growth rate is higher than at least 1 of the single agent growth rates = antagonistic. A p value < 0.05 was considered statistically significant.
Example 4. Compound A with or without Docetaxel in Participants with Locally Advanced or Metastatic Non-small Cell Lung Cancer
[00262] The purpose of this study is to determine the recommended phase 2 dose (RP2D) of Compound A when administered in combination with docetaxel in patients with non-small cell lung cancer (NSCLC) and to evaluate efficacy, safety, and tolerability of Compound A administered alone and in combination with docetaxel at the RP2D in patients with locally advanced or metastatic non-small cell lung cancer. [00263] This study consists of 2 phases: Phase lb - dose escalation phase; Phase 2 - expansion phase.
[00264] Approximately 15 patients with NSCLC who have been treated with multiple prior lines of therapies are enrolled for Phase lb. The participants receive docetaxel (36 mg/m2) intravenous (IV) and Compound A tablets, orally administered, once daily in 21 -day dosing cycles. Compound A dose is escalated until recommended Phase 2 dose (RP2D) is determined. Up to 140 adults with locally advanced or metastatic NSCLC refractory or resistant to 1 prior line platinum-based, non-docetaxel containing systemic chemotherapy are enrolled for Phase 2 (dose expansion). Phase 2 expansion study uses a sequential, multistage Bayesian adaptive design and consists of up to 3 parts evaluating the following patient populations:
[00265] Part 1 : NSCLC (inclusive of both squamous and nonsquamous histology and all PIK3CA genotypes).
[00266] Part 2: Histology-specific NSCLC (either squamous or nonsquamous).
[00267] Part 3: Histology- and genotype-specific NSCLC (PD 3CA MUT/AMP positive squamous NSCLC if squamous histology is selected in Part 2).
[00268] Each part of the adaptive Phase 2 study is designed as a stand-alone, randomized study evaluating Progression-Free Survival (PFS) as the primary efficacy measure in a total of 60 patients between the 2 treatment arms: Compound A plus docetaxel versus docetaxel alone. An event-driven analysis of PFS is performed after each part of the Phase 2 study. On the basis of the PFS analysis of the preceding part of the study, the study may be stopped for efficacy or futility, or proceed to the next part.
[00269] PFS in Phase 2 is is evaluated for approximately 6 months after last dose of study drug. PFS is defined as the time from the date of randomization to the date of first documentation of PD or death due to any cause, whichever occurs first.
[00270] Study drug is administered in 21 -day dosing cycles. During each phase of the study, patients are treated with a maximum of 9 cycles of either docetaxel alone or docetaxel plus Compound A. Patients treated with docetaxel plus Compound A may continue to receive Compound A monotherapy until progression of disease, occurrence of unacceptable toxicities or death. The maximum duration of treatment for patients is 12 months unless it is determined that a patient would derive benefit from continued treatment beyond 12 months. Patients are followed after discontinuation of study drug to collect PFS and OS data. Patients may withdraw from therapy at any time. The overall time to participate in this study is up to 24 months.
[00271] The number of participants experiencing Dose-Limiting Toxicities (DLT) in Phase lb is evaluated for approximately 9 months. Toxicity is evaluated according to the NCI CTCAE version 4.03. DLT is defined as any of the events specified in the protocol that are considered by the investigator to be at least possibly related to therapy with Compound A plus docetaxel.
[00272] Maximum Tolerated Dose (MTD) in Phase lb is evaluated for approximately 9 months. MTD is defined as the highest dose level of Compound A when administered with weekly docetaxel at which no more than 1 of the 6 patients experiences a DLT during the first cycle (21 days) of therapy in Phase lb.
[00273] Recommended Phase 2 Dose in Phase lb (RP2D) of Compound A plus docetaxel is evaluated for approximately 9 months. RP2D is determined in Phase lb based on safety, tolerability, PK and other observations in both Cycle 1 and Cycle 2 and beyond. The RP2D does not exceed the MTD.
[00274] Secondary Outcome Measures:
[00275] Clinically significant change from baseline in vital sign measures is assessed up to 30 days from the last dose date. Vital sign measurements include measurements of diastolic and systolic blood pressure, heart rate, and temperature.
[00276] The percentage of patients with clinically significant changes from baseline in participant physical examination findings is assessed up to 30 days from the last dose date.
[00277] Change from baseline in 12-lead electrocardiogram (ECG) is assessed over the duration of the study. Clinically significant changes from baseline in ECGs are tabulated by time point including any unscheduled measurements.
[00278] Change from Baseline in Clinical laboratory test results is assessed up to 30 days from the last dose date. Shift tables summarizing the number of patients with each baseline NCI CTCAE grade and changes to the worst NCI CTCAE grade are produced for selected laboratory parameters.
[00279] The percentage of adverse events (AE) is assessed from signing of informed consent up to 30 days after last dose date. Treatment-emergent AEs that occur after administration of the first dose of study drug and through 30 days after the last dose of study drug are tabulated.
An Adverse Event (AE) is defined as any untoward medical occurrence in a clinical investigation participant administered a drug; it does not necessarily have to have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign
(eg, a clinically significant abnormal laboratory finding), symptom, or disease temporally associated with the use of a drug, whether or not it is considered related to the drug.
[00280] The percentage of participants experiencing Serious adverse events is assessed from signing of informed consent up to 30 days after last dose date. A serious adverse event (SAE) is any untoward medical occurrence or effect that at any dose results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability / incapacity, is a congenital anomaly / birth defect or is medically important due to other reasons than the above mentioned criteria.
[00281] Response Rate is assessed approximately 12 months. Response rate is defined as complete response + partial response.
[00282] Disease Control Rate is assessed at the end of the Cycle 2 , 4, and 6, and then every 3 cycles thereafter until the patient discontinues study drug due to disease progression, unacceptable toxicity, or death (approximately 12 months). Disease control rate is defined as complete response [CR] + partial response [PR] +stable disease [SD].
[00283] Duration of Response is assessed from the date of first documented response up to the first documentation of progression of disease (approximately 12 months). The duration of response is defined as the time from the date of first documentation of a response to the date of first documentation of progression of disease.
[00284] Time to Progression is assessed from the date of randomization to the date of first documentation of progressive disease (approximately 24 months). Time to progression is defined as the time from the date of randomization to the date of first documentation of progression of disease.
[00285] Overall Survival in Phase 2 is assessed every 3 months from the last dose of the study drug for up to one year. Overall survival is defined as the time from the date of randomization to the date of death.
[00286] Cmax: Single dose Maximum (peak) plasma concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. Maximum observed plasma concentration
(Cmax) is the peak plasma concentration of a drug after administration, obtained directly from the plasma concentration-time curve.
[00287] Tmax: single-dose first time of occurrence of maximum (peak) concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. Tmax is defined as time to reach the maximum plasma concentration (Cmax).
[00288] AUC,a„: Area under the concentration time curve from time 0 to the next dose in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. AUC,aU is defined as area under the plasma concentration versus time curve from zero to next dose.
[00289] AUC(iast) Area Under the Plasma Concentration-Time Curve From Time 0 to the Time of the Last Quantifiable Concentration in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. AUC(iaSi) is defined as area under the plasma concentration versus time curve from zero to the time of the last quantifiable concentration. [00290] CL F: Apparent total body clearance in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. CL/F is apparent clearance of the drug after extravascular administration.
[00291] Ύι '. Terminal disposition half-life in Phase lb is assessed Cycle 1 Day 1 pre-dose and up to 24 hours post-dose. Ti/2 is the time required for half of the drug to be eliminated from the plasma.
[00292] Compound A plasma concentrations when administered 1 day after docetaxel is 1 day post docetaxel dose. The plasma concentration of Compound A was evaluated one day following docetaxel dosing.
Figure imgf000080_0001
[00293] The age Eligible for the study is 18 Years and older. Both male and female genders are eligible for the study.
[00294] The following inclusion criteria are used: Has a histologically and/or cytologically confirmed diagnosis of NSCLC (squamous or nonsquamous). For Phase 2, has a diagnosis of mixed squamous and nonsquamous (or adenosquamous) NSLC. Has locally advanced or metastatic disease (Stage Illb or Stage IV) with radiographically or clinically evaluable lesions. Has experienced failure of at least 1 prior chemotherapy regimen. For Phase 2, has received 1 prior platinum-based chemotherapy regimen (excluding a docetaxel-containing regimen) for advanced or metastatic (Stage Illb or Stage IV) disease followed by documented progressive disease. For Phase lb, has experienced failure of multiple lines of prior chemotherapy. For Phase 2, has archived or fresh tumor biopsy samples obtained during screening sufficient for genotyping. Has adequate organ function, before the first dose of study drug. Has Eastern Cooperative Oncology Group (ECOG) Performance Status of 0 or 1. Female participants who are postmenopausal for at least 1 year before the screening visit or are surgically sterile, or are of childbearing potential, agree to practice 2 effective methods of contraception, at the same time, from the time of signing the informed consent through 30 days after the last dose of study drug, or agree to practice true abstinence. Male participants agree to practice effective barrier contraception during the entire study treatment period and through 30 days after the last dose of study drug, or agree to practice true abstinence. Has suitable venous access for the study- required blood sampling. Has recovered (ie, <= Grade 1 toxicity or eligibility per this protocol is met) from the reversible effects of prior anticancer therapy.
[00295] The following exclusion criteria are used: Previous treatment with a PI3 or AKT inhibitor. Prior cancer therapy or other investigational therapy within 2 weeks before the first administration of study drug or failed to recover from the reversible effects of prior anticancer therapies. For prior therapies with a half-life longer than 3 days, the interval must be at least 28 days before the first administration of study drug, and the patient must have documented progressive disease. Has poorly controlled diabetes mellitus defined as HbAlc > 6.5%. Has taken strong inhibitors or strong inducers of CYP3A4 within 14 days before the first dose of study drug. Has taken histamine-H2 receptor antagonists and/or neutralizing antacids within 24 hours before the first administration of study drug. Has taken proton pump inhibitors within 7 days before the first administration of study drug. Has any clinically significant co-morbidities.
Has acute myocardial infarction within 6 months before starting study drug, current or history of
New York Heart Association Class III or IV heart failure, Evidence of current uncontrolled cardiovascular conditions including cardiac arrhythmias, angina, pulmonary hypertension, or
ECG evidence of acute ischemia or active conduction system abnormalities, Fridericia's corrected QT interval > 475 milliseconds (msec) (males) or > 450 msec (females) on a 12-lead
ECG during the Screening period, or abnormalities on 12-lead ECG including, but not limited to, changes in rhythm and intervals that in the opinion of the investigator are considered to be clinically significant. Has known, previously diagnosed human immunodeficiency virus infection or active chronic hepatitis B or C. Has brain metastasis, except for those patients who have completed definitive therapy, are not on steroids, have a stable neurologic status for at least 2 weeks after completion of the definitive therapy and steroids, and do not have neurologic dysfunction that would confound the evaluation of neurologic and other adverse events. Has active secondary malignancy that requires treatment. Has any serious medical or psychiatric illness, including drug or alcohol abuse. Male participants who intend to donate sperm during the course of this study or 12 weeks after receiving their last dose of study drug. Female participants who are lactating and breastfeeding or have a positive serum pregnancy test during the Screening period or a positive urine pregnancy test on Day 1 before administration of the first dose of study drug.
Example 5. Compound A with or without Docetaxel in Participants with Locallv Advanced and Metastatic Gastric or Gastroesophageal Adenocarcinoma
[00296] This is an open-label, multicenter, phase lb study of Compound A in combination with Compound B (6-((( 1 R,2S)-2-aminocyclohexyl)amino)-7-fluoro-4-( 1 -methyl- 1 H-pyrazol-4-yl)- lH-pyrrolo[3,4-c]pyridin-3(2H)-one citrate), alisertib (MLN8237), paclitaxel, or docetaxel in adult patients with locally advanced and metastatic gastric or gastroesophageal adenocarcinoma. The study consists of a dose escalation phase (Part 1) and a dose expansion phase (Part 2). The statistical design for the dose expansion consists of equal randomization and adaptive randomization phases.
[00297] The primary objective in Part 1 (dose escalation) is to determine dose-limiting toxicity (DLT) and the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) for Compound A when administered with each of the combination partners.
[00298] The primary objective in Part 2 (dose expansion) is to evaluate the overall response rate (ORR) as the primary efficacy measure of Compound A in combination with each of the combination partners in patients with gastric or gastroesophageal adenocarcinoma.
[00299] The secondary objectives are to evaluate the safety and tolerability of Compound A in combination with each of the combination partners, to evaluate additional efficacy measures, such as progression-free survival (PFS), disease control rate, response duration, time to progression (TTP), and overall survival (OS) of Compound A in combination with each of the combination partners in patients with gastric or gastroesophageal adenocarcinoma, and to evaluate the pharmacokinetics (PK) of Compound A when dosed in combination with alisertib, Compound B, docetaxel, or paclitaxel, and to evaluate the PK of alisertib and Compound B when these agents are dosed in combination with Compound A. [00300] Inclusion criteria: Male and female patients aged 18 years or older at the time of consent. In Part 1 (dose escalation), patients must have a histologically confirmed diagnosis of advanced solid tumor, including but not limited to gastric or gastroesophageal adenocarcinoma, and are refractory to or relapsed after prior line(s) of therapy with no effective therapeutic options available. In Part 2 (dose expansion), patients must have a histologically confirmed diagnosis of metastatic or locally advanced adenocarcinoma of the stomach or gastroesophageal junction (Stage Illb or IV), with measurable lesions per modified RECIST, Version 1.1 by radiographic techniques (CT or MRI), and have received 1 prior systemic chemotherapy regimen for advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction with documented progressive disease.
[00301] Exclusion criteria: Patients who have received prior systemic anticancer therapies, or other investigational agents or radiotherapy within 2 weeks before first dose of study drug; are receiving treatment with P-glycoprotein (P gp) inhibitors/inducers (Compound A + Compound B arm only); have received strong cytochrome P 450 (CYP) 3A4 inducers/inhibitors or proton pump inhibitors (PPIs) within 7 days before the first administration of study drug or have conditions that require the concomitant use of CYP3A4 inducers/inhibitors or PPIs during the course of the study; have poorly controlled diabetes mellitus; have signs of peripheral neuropathy >NCI CTCAE Grade 2; have symptomatic brain metastases or brain metastases with a stable neurologic status for <2 weeks after completion of the definitive therapy and steroids. In Part 2 (dose expansion), prior treatment with any of the following: Aurora A-targeted agent (excluding the Compound A + Compound B arm); taxane-containing regimen (excluding the Compound A + Compound B arm); spleen tyrosine kinase (SYK) inhibitor (Compound A + Compound B arm only); or phosphoinositide 3-kinase (PI3K) or serine/threonine kinase, also known as protein kinase B or PKB (AKT) inhibitor.
[00302] Part 1 (Dose Escalation)
[00303] During Part 1 , the dose of Compound A will be escalated (planned doses of 300 mg, 600 mg, and 900 mg) according to a 3+3 dose escalation scheme, while Compound B, alisertib, paclitaxel, and docetaxel will be administered at a fixed dose and regimen until the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) is determined.
[00304] Part 2 (Dose Expansion)
[00305] Compound A will be administered PO QD for 3 days on (i.e., Days 2, 3, 4; 9, 10, 11;
16, 17, and 18) and 4 days off per week in each 21 -day cycle. The RP2D of Compound A when administrated concomitantly with docetaxel will be determined in the Part 1 dose escalation phase and used in the Part 2 dose expansion phase. Docetaxel will be administered IV at 75 mg/m2 on Day 1 once every 3 weeks in a 21 -day cycle.
Figure imgf000084_0001
Docetaxel X

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of treating a neoplastic condition in a subject in need thereof comprising administering simultaneously or sequentially a therapeutically effective amount of a
combination of a PI3K-kinase a (PI3 a) inhibitor and a taxane, wherein the taxane is not paclitaxel, and wherein the PI3Koc inhibitor is a compound of the following formula:
Figure imgf000085_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
R3 is amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
2. The method of claim 1 , wherein the PI3Ka inhibitor
Figure imgf000085_0002
or a pharmaceutically acceptable salt thereof.
3. The method of claim 1 , wherein the taxane is docetaxel.
4. The method of any one of claims 1 -3, wherein the neoplastic condition is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
5. The method of claim 4, wherein the neoplastic condition is non-small cell lung cancer.
6. The method of claim 5, wherein the non-small cell lung cancer is squamous non- small cell lung cancer.
7. The method of claim 5, wherein the non-small cell lung cancer is non-squamous non- small cell lung cancer
8. The method of any one of claims 1 -7, wherein the subject has a PIK3CA mutation and/or amplification.
9. The method of any one of claims 1-8, wherein the taxane is administered to the subject prior to administration of the PI3Ka inhibitor.
10. The method of claim 9, wherein the taxane is administered to the subject about 12 to about 36 hours prior to administration of the PBKot inhibitor.
11. The method of claim 9, wherein the taxane is administered to the subject about 24 hours prior to administration of the PI3Ka inhibitor.
12. The method of any one of claims 1 -8, wherein the PI3K a inhibitor and the taxane are administered simultaneously.
13. The method of any one claims 1-11, wherein the PI3Ka inhibitor is ao!ministered according to an intermittent regimen.
14. The method of claim 13 wherein the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6, or 7 consecutive days, followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
15. The method of claim 13, wherein the PDKa inhibitor is administered for 3 non- consecutive days of a 7-day cycle.
16. The method of claim 13 wherein the PI3K.cc inhibitor is administered for 3 consecutive days of a 7-day cycle followed by an intermission of at least 1 day.
17. The method of claim 13, wherein the PI3 a inhibitor is administered on 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle.
18. The method of any one of claims 1-17, wherein a dose of the PDKa inhibitor is about 100 mg to about 2100 mg.
19. The method of any one of claims 1-17, wherein a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, about 1200, about 1500 mg, about 1800 mg or about 2100 mg.
20. The method of any one of claims 1-17, wherein the amount of PDKa inhibitor adniinistered is about 300 mg to about 6300 mg in a 7-day cycle.
21. The method of any one of claims 1-20, wherein the taxane is administered at a dose of about 25 mg/m2 to about 100 mg/m2 in a single administration.
22. The method of claim 21, wherein the taxane is administered at a dose of about 36 mg/rrf in a single administratioa
23. A pharmaceutical regimen for the treatment of a disorder mediated by PI3-kinase a (PI3Ka), wherein the regimen comprises simultaneous or sequential administration of at least one taxane and at least one PI3Kct inhibitor to a human subject in need thereof, wherein the taxane is not paclitaxel; and wherein the PI3Ka inhibitor is a compound of the following formula:
Figure imgf000087_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
R3 is amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic; or wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4-, 5-, 6-, or 7-membered ring.
24. The pharmaceutical regimen of 23 wherein the PI3Ka
Figure imgf000087_0002
or a pharmaceutically acceptable salt thereof.
25. The pharmaceutical regimen of claim 23, wherein the taxane is docetaxel.
26. The pharmaceutical regimen of any one of claims 23-25, wherein the regimen comprises at least one cycle wherein the taxane is administered before the PI3Ka inhibitor and wherein the taxane and the PI3Ka inhibitor are not administered on the same day.
27. The pharmaceutical regimen of any one of claims 23-26, wherein the PI3Ka inhibitor is administered according to an intermittent regimen.
28. The pharmaceutical regimen of claim 27, wherein the PI3 a inhibitor is
administered for 2, 3, 4, 5, 6, or 7 consecutive days, followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
29. The pharmaceutical regimen of claim 27, wherein the PI3Ka inhibitor is
administered for 3 non-consecutive days in a 7-day cycle.
30. The pharmaceutical regimen of claim 27, wherein the PI3Ka inhibitor is
aaministered for 3 consecutive days of a 7-day cycle followed by an intermission of at least 1 day.
31. The pharmaceutical regimen of claim 27, wherein the PI3Kot inhibitor is
administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle.
32. The pharmaceutical regimen of claim 27, wherein the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 2, 3, and 4 of a 7-day cycle.
33. The pharmaceutical regimen of claim 27, wherein the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 2, 4, and 6 of a 7-day cycle.
34. The pharmaceutical regimen of claim 27, wherein the taxane is administered on Day 1 and the PI3Ka inhibitor is administered on Days 3, 4, and 5 of a 7-day cycle.
35. The pharmaceutical regimen of any one of claims 23-34, wherein the PI3Ka inhibitor is administered once daily on each of the days that the PI3Ka inhibitor is administered.
36. The pharmaceutical regimen of any one of claims 23-35, wherein a dose of PI3Ka inhibitor is about 100 mg to about 2100 mg.
37. The pharmaceutical regimen of any one of claims 23-35 wherein a dose of the PI3Ka inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg.
38. The pharmaceutical regimen of any one of claims 23-35, wherein the amount of the PI3Ka inhibitor administered is about 300 mg to about 6300 mg in 7-day cycle.
39. The pharmaceutical regimen of any one of claims 23-38, wherein the taxane is administered at a dose of about 25 mg/m2 to about 100 mg m2 in a single administration.
40. The pharmaceutical regimen of claim 39, wherein the taxane is administered at a dose of about 36 mg/m2 in a single administration.
41. The pharmaceutical regimen of any one of claims 23-40, wherein the disorder is a cancer selected from the group consisting of non-small cell lung cancer, small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, breast cancer, ovarian cancer, renal cell carcinoma, prostate cancer, neuroendocrine cancer, gastric cancer, bladder cancer, colon cancer, cervical cancer and endometrial cancer.
42. The pharmaceutical regimen of claim 41, wherein the disorder is non-small cell lung cancer.
43. The pharmaceutical regimen of claim 42, wherein the non-small cell lung cancer is squamous non-small cell lung cancer.
44. The pharmaceutical regimen of claim 42, wherein the non-small cell lung cancer is non-squamous non-small cell lung cancer
45. The pharmaceutical regimen of any one of claims 23-44, wherein the subject has a PDGCA mutation or amplification.
46. A method of enhancing apoptosis in cancer cells comprising administering to the cells simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the PDKct inhibitor is a compound of the following formula:
Figure imgf000089_0001
or a pharmaceutically acceptable salt thereof, wherein
W1 is CR3;
R is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or N 'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
amido of formula -C(0)N(R)2 or-NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic;
wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4 or 7-membered ring.
47. The method of claim 46, wherein the PI3Ka inhibitor is
Figure imgf000089_0002
or a pharmaceutically acceptable salt thereof.
48. The method of claim 46, wherein the taxane is docetaxel.
49. The method of any one of claims 46-48, wherein the administration takes place in vitro.
50. The method of any one of claims 46-48, wherein the administration takes place in vivo.
51. The method of any one of claims 46-50, wherein the taxane is administered to the cell prior to administration of the PI3Ka inhibitor.
52. The method of claim 51, wherein the taxane is administered to the cell about 12 hours to about 36 hours prior to administration of the PI3Ka inhibitor.
53. The method of claim 51, wherein the taxane is administered to the cell about 24 hours prior to administration of the PI3 a inhibitor.
54. The method of any one of claims 46-53, wherein the PI3Ka inhibitor is administered according to an intermittent regimen.
55. The method of claim 54, wherein the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6, or 7 consecutive days, followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
56. The method of claim 54, wherein the PDKa inhibitor is administered for 3 non- consecutive days of a 7-day cycle.
57. The method of claim 54, wherein the PI3Ka inhibitor is administered for 3 consecutive days of a 7-day cycle followed by an intermission of at least 1 day.
58. The method of claim 54, wherein the PDKct inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle.
59. A method of suppressing tumor regrowth in a subject in need thereof comprising administering to the subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase a (PI3 a) inhibitor and a taxane, wherein the Pl3Koc inhibitor is a compound of the following formula:
Figure imgf000090_0001
or a pharmaceutically acceptable salt thereof, wherein
Wl is CR3;
R1 is hydrogen;
R2 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR'R" wherein R' and R" are taken together with nitrogen to form a cyclic moiety; and
amido of formula -C(0)N(R)2 or -NHC(0)R, wherein R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic;
wherein the (R)2 groups taken together with the nitrogen to which it is attached form a 4 or 7-membered ring.
60. The method of claim 59, wherein the PDKa inhibitor is
Figure imgf000091_0001
or a pharmaceutically acceptable salt thereof.
61. The method of claim 59, wherein the taxane is docetaxel.
62. The method of any one of claims 59-61, wherein the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the PI3 a inhibitor or the taxane.
63. The method of any one of claims 59-61, wherein the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the PI3 a inhibitor or the taxane.
64. The method of any one of claims 59-63, wherein the taxane is administered to the subject prior to administration of the PI3Ka inhibitor.
65. The method of claim 64, wherein the taxane is administered to the subject about 12 to about 36 hours prior to administration of the PI3Ka inhibitor.
66. The method of claim 64, wherein the taxane is administered to the subject about 24 hours prior to administration of the PDKcc inhibitor.
67. The method of any one of claims 59-66, wherein the PI3Ka inhibitor is administered according to an intermittent regimen.
68. The method of claim 67, wherein the PI3Ka inhibitor is administered for 2, 3, 4, 5, 6, or 7 consecutive days, followed by an intermission of at least 1, 2, 3, 4, 5, or 6 consecutive days.
69. The method of claim 67, wherein the PI3Ka inhibitor is administered for 3 non- consecutive days of a 7-day cycle.
70. The method of claim 67, wherein the PI3Ka inhibitor is ao^ninistered for 3 consecutive days of a 7-day cycle followed by an intermission of at least 1 day.
71. The method of claim 67, wherein the PI3Koc inhibitor is administered for 3 consecutive days followed by an intermission of 4 consecutive days per 7-day cycle.
72. The method of any one of claims 59-71, wherein a dose of PDKoc inhibitor administered is about 100 mg to about 2100 mg.
73. The method of any one of claims 59-71, wherein a dose of the PDKoc inhibitor is about 100 mg, about 300 mg, about 600 mg, about 900 mg, about 1200 mg, about 1500 mg, about 1800 mg or about 2100 mg.
74. The method of any one of claims 59-71, wherein the amount of PDKoc inhibitor administered is about 300 mg to about 6300 mg in a 7-day cycle.
75. The method of any one of claims 59-61, wherein the subject does not exhibit tumor regrowth for at least 20 days from the first date of administration of the taxane.
76. The method of any one of claims 59-61, wherein the subject does not exhibit tumor regrowth for at least 35 days from the first date of administration of the taxane.
PCT/US2015/051387 2014-09-24 2015-09-22 COMBINATION TREATMENT WITH PI3Kα INHIBITORS AND TAXANES WO2016048982A2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201462054905P 2014-09-24 2014-09-24
US62/054,905 2014-09-24
US201562132931P 2015-03-13 2015-03-13
US62/132,931 2015-03-13
US201562195479P 2015-07-22 2015-07-22
US62/195,479 2015-07-22

Publications (1)

Publication Number Publication Date
WO2016048982A2 true WO2016048982A2 (en) 2016-03-31

Family

ID=55582237

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/051387 WO2016048982A2 (en) 2014-09-24 2015-09-22 COMBINATION TREATMENT WITH PI3Kα INHIBITORS AND TAXANES

Country Status (2)

Country Link
TW (1) TW201625263A (en)
WO (1) WO2016048982A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160176869A1 (en) * 2014-12-18 2016-06-23 Takeda Pharmaceutical Company Limited Solid State Forms of Fused Heteroaromatic Pyrrolidinones

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160176869A1 (en) * 2014-12-18 2016-06-23 Takeda Pharmaceutical Company Limited Solid State Forms of Fused Heteroaromatic Pyrrolidinones
US10676473B2 (en) * 2014-12-18 2020-06-09 Takeda Pharmaceutical Company Limited Solid state forms of fused heteroaromatic pyrrolidinones
US11352355B2 (en) 2014-12-18 2022-06-07 Calithera Biosciences, Inc. Solid state forms of fused heteroaromatic pyrrolidinones

Also Published As

Publication number Publication date
TW201625263A (en) 2016-07-16

Similar Documents

Publication Publication Date Title
US9669032B2 (en) Enhanced treatment regimens using mTOR inhibitors
JP6114296B2 (en) Treatment regimen with multiple drugs
JP6584952B2 (en) Treatment of cancer using PI3 kinase isoform modulator
ES2588197T3 (en) Benzoxazole kinase inhibitors and methods of use
EP3442529B1 (en) Combination therapy with notch and cdk4/6 inhibitors for the treatment of lung cancer
US20180162807A1 (en) Aromatic compounds and uses thereof
US10040767B2 (en) Benzimidazole derivatives and uses thereof
JP2016522212A (en) Treatment of cancer using PI3 kinase isoform modulator
EP3268362A1 (en) Substituted pyridines and uses thereof
EP2914295A1 (en) Treatment of rheumatoid arthritis and asthma using p13 kinase inhibitors
WO2014151147A1 (en) Combination of kinase inhibitors and uses thereof
WO2012154695A2 (en) Treatment of polycystic disease
US20210253614A1 (en) Compositions and methods for inhibiting cd73
WO2016048982A2 (en) COMBINATION TREATMENT WITH PI3Kα INHIBITORS AND TAXANES
US20160287601A1 (en) Enhanced treatment regimens using pi3k inhibitors
WO2010096732A1 (en) Combination therapy to improve drug efficiency
EP2928488A1 (en) Combinations of a pi3k/akt inhibitor compound with an her3/egfr inhibitor compound and use thereof in the treatment of a hyperproliferative disorder
Melichar et al. Serum Uric Acid and C-reactive Protein in Patients with Metastatic Colorectal Carcinoma During Combination Therapy with High-dose Folinic Acid
NZ786604A (en) Combination therapy with notch and cdk4/6 inhibitors for the treatment of

Legal Events

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

Ref document number: 15844225

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15844225

Country of ref document: EP

Kind code of ref document: A2