NZ617245B2 - Combinations of akt inhibitor compounds and chemotherapeutic agents, and methods of use - Google Patents

Combinations of akt inhibitor compounds and chemotherapeutic agents, and methods of use Download PDF

Info

Publication number
NZ617245B2
NZ617245B2 NZ617245A NZ61724512A NZ617245B2 NZ 617245 B2 NZ617245 B2 NZ 617245B2 NZ 617245 A NZ617245 A NZ 617245A NZ 61724512 A NZ61724512 A NZ 61724512A NZ 617245 B2 NZ617245 B2 NZ 617245B2
Authority
NZ
New Zealand
Prior art keywords
cancer
compound
acceptable salt
combination
formula
Prior art date
Application number
NZ617245A
Other versions
NZ617245A (en
Inventor
Kui Lin
Michelle Nannini
Premal Patel
Elizabeth Punnoose
Deepak Sampath
Jeffrey Wallin
Original Assignee
Genentech 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 Genentech Inc filed Critical Genentech Inc
Priority claimed from PCT/US2012/031720 external-priority patent/WO2012135781A1/en
Publication of NZ617245A publication Critical patent/NZ617245A/en
Publication of NZ617245B2 publication Critical patent/NZ617245B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41661,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • 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
    • A61K31/436Heterocyclic 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 six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes

Abstract

Disclosed herein is a combination comprising a) compound of formula Ia 2-(4-chlorophenyl)-3-(isopropylamino)-1-(4-((R)-5-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)piperazin-1-yl)propan-1-one (GDC-0068/ipatasertib) or a pharmaceutically acceptable salt thereof and 5-FU (fluorouracil) and/or capecitabine. Also disclosed is the use of the combinations for treating hyperproliferative disorders, such as cancer. apecitabine. Also disclosed is the use of the combinations for treating hyperproliferative disorders, such as cancer.

Description

COMBINATIONS OF AKT INHIBITOR COMPOUNDS AND CHEMOTHERAPEUTIC AGENTS, AND METHODS OF USE PRIORITY OF INVENTION This application claims priority to United States Provisional Application Number 61/470,803 that was filed on April 1, 2011, and to United States Provisional Application Number ,624 that was filed on April 1, 2011. The entire content of these provisional ations are hereby incorporated herein by reference.
FIELD OF THE ION The invention relates generally to pharmaceutical combinations of compounds with activity against hyperproliferative disorders such as cancer and which include compounds that inhibit AKT kinase activity. Also described are methods of using the combinations for in vitro , in situ, and in vivo diagnosis or treatment of mammalian cells, or associated pathological conditions.
OUND OF THE INVENTION Protein kinases (PK) are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine and threonine es of proteins by transfer of the terminal ) ate from ATP. Through signal transduction pathways, these enzymes modulate cell growth, differentiation and proliferation, i.e., virtually all aspects of cell life in one way or r depend on PK activity (Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Book. I and II, Academic Press, San Diego, CA). Furthermore, abnormal PK activity has been related to a host of disorders, ranging from vely non-life threatening diseases such as psoriasis to extremely virulent diseases such as astoma (brain cancer). Protein kinases are an important target class for therapeutic modulation (Cohen, P. (2002) Nature Rev. Drug Discovery 1:309).
International Patent Application Publication Number discusses a series of inhibitors of AKT of formula I: R1 N R5 R2O N R10 (I).
Currently, there remains a need for improved s and/or compositions that can be used to treat hyperproliferative diseases such as cancer. The t invention addresses at least one aspect of this need; and/or at least provides the public with a useful choice.
In this ication where reference has been made to patent specifications, other external documents, or other sources of information, this is generally for the purpose of providing a context for discussing the features of the invention. Unless specifically stated otherwise, reference to such external documents is not to be construed as an admission that such documents, or such sources of information, in any jurisdiction, are prior art, or form part of the common general knowledge in the art.
In the description in this specification reference may be made to subject matter that is not within the scope of the claims of the current application. That subject matter should be readily fiable by a person skilled in the art and may assist in putting into practice the invention as defined in the claims of this application.
SUMMARY OF THE INVENTION The present invention provides the combination of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof and one or more agents selected from 5-FU, and capecitabine, or a ceutically acceptable salt thereof.
The invention also es a kit comprising a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof; and one or more of 5-FU and capecitabine or a pharmaceutically acceptable salt thereof.
The ion also provides a product comprising a compound of formula Ia (Ia) or a pharmaceutically acceptable salt f; and one or more of 5-FU and capecitabine or a pharmaceutically acceptable salt thereof, as a combined ation for simultaneous, separate or sequential use in the therapeutic treatment of a hyperproliferative disorder.
The invention also relates to a use of a compound of formula Ia: (Ia) or a pharmaceutically able salt thereof and one or more agents selected from 5-FU, and capecitabine, or a ceutically acceptable salt thereof, in the manufacture of a medicament for the therapeutic treatment of a hyperproliferative disorder.
The invention also relates to the use of a compound of formula Ia HO (Ia) or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for therapeutically treating a hyperproliferative er in a subject, in combination with one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof.
The invention also relates to the use of a nd of formula Ia: (Ia) or a pharmaceutically acceptable salt thereof, and an agent selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the therapeutic use for improving the quality of life of a patient treated for a hyperproliferative disorder.
The invention also relates to the use of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof, and one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a ment for use in the eutic treatment of a hyperproliferative disorder in a mammal.
The invention also relates to the use of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof, and one or more agents selected from 5-FU, and tabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of mesothelioma, endometrial, , lung, ovarian, prostate, pancreatic, melanoma, gastric, colon, glioma, or head and neck cancer in a mammal.
The invention also relates to the use of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof, and one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically able salt thereof, in the manufacture of a medicament for use in the treatment or prevention of chemotherapy-resistant cancer.
BRIEF DESCRIPTION It has been determined that additive or synergistic effects in inhibiting the growth of cancer cells in vitro and in vivo can be achieved by administering a compound of formula I or a pharmaceutically acceptable salt thereof in combination with certain other ic herapeutic agents. The combinations and methods may be useful in the treatment of hyperproliferative disorders such as cancer.
Described is a method for treating a hyperproliferative disorder in a mammal comprising, administering to the mammal, a) a compound of a I: R1 N R5 R2O N R10 (I) or a pharmaceutically acceptable salt thereof; and b) one or more agents selected from 5-FU, a platinum agent (carboplatin, cisplatnin, oxaliplatin, etc.) irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, nib, PD-0325901, axel, bevacizumab, pertuzumab, fen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and GDC-0973.
Also described is a combination of, a) a compound of formula I: R1 N R5 R2O N R10 (I) or a pharmaceutically acceptable salt thereof; and b) one or more agents selected from 5-FU, a platinum agent, leucovorin, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, paclitaxel, zumab, pertuzumab, fen, rapamycin and lapatinib, or a pharmaceutically acceptable salt thereof for the prophylactic or therapeutic treatment of a hyperproliferative disorder. In one example, the formula I compound is GDC- 0068 or a salt thereof.
The compound of formula I or the pharmaceutically acceptable salt thereof and the chemotherapeutic agent may be mulated for administration in a combination as a pharmaceutical ition or they may be administered separately in alternation (sequentially) as a therapeutic combination.
Described is a method for treating a disease or condition modulated by AKT kinase in a mammal comprising, administering to the mammal, a) a compound of formula I or a pharmaceutically able salt thereof; and b) one or more agents selected from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, lomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, 32, MDV3100, abiraterone, and GDC-0973.
Also described is a combination of a) a compound of formula I or a pharmaceutically acceptable salt thereof; and b) one or more agents ed from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, erlotinib, 5901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and GDC-0973 for treating a hyperproliferative disorder.
Also described is a combination of a) a compound of formula I or a pharmaceutically acceptable salt thereof; and b) one or more agents selected from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, abine, SN-38, capecitabine, temozolomide, nib, PD-0325901, paclitaxel, bevacizumab, umab, tamoxifen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and 73 for treating a e or condition modulated by AKT kinase.
Also described is the use of a compound of formula I or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of a hyperproliferative disorder in a mammal, wherein one or more agents ed from 5-FU, a platinum agent, irinotecan, docetaxel, bicin, abine, SN-38, capecitabine, temozolomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, PLX-4032, 0, abiraterone, and GDC-0973 are administered to the mammal.
Also described is the use of a compound of formula I or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of a e or condition modulated by AKT kinase in a mammal, wherein one or more agents selected from -FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and 73 are administered to the mammal.
Also described is a kit comprising a compound of formula I or a pharmaceutically acceptable salt thereof, a container, and a package insert or label indicating the administration of the compound of formula I or a ceutically acceptable salt thereof with one or more agents selected from 5-FU, a um agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, nib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and GDC- 0973 for treating a hyperproliferative disorder.
Also described is a product comprising a compound having formula I or a pharmaceutically acceptable salt thereof, and a chemotherapeutic agent selected from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, erlotinib, 5901, paclitaxel, zumab, pertuzumab, fen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and GDC-0973; as a combined preparation for separate, simultaneous or sequential use in the treatment of a hyperproliferative disorder.
In addition to providing improved treatment for a given hyperproliferative disorder, administration of certain combinations ding those of the invention) may improve the quality of life for a patient compared to the quality of life experienced by the same patient receiving a different treatment. For example, administration of a combination of a compound of formula I or a pharmaceutically able salt thereof, and a chemotherapeutic agent as described herein to a patient may provide an improved quality of life compared to the quality of life the same t would experience if they received only the herapeutic agent as y. For example, the combined therapy with the combination described herein may lower the dose of chemo agents needed, thereby lessening the side-effects associated with high-dose chemotherapeutic agents (e.g., nausea, vomiting, hair loss, rash, decreased appetite, weight loss, etc.). The combination may also cause d tumor burden and the associated adverse , such as pain, organ dysfunction, weight loss, etc. Accordingly, bed is a compound of formula I or a pharmaceutically acceptable salt thereof, for therapeutic use for improving the quality of life of a patient treated for a hyperproliferative disorder with an agent selected from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, PLX-4032, MDV3100, abiraterone, and GDC- 0973. Accordingly, also bed is a compound of formula I or a pharmaceutically acceptable salt thereof, for therapeutic use for improving the quality of life of a patient treated for a hyperproliferative disorder with an agent selected from 5-FU, a platinum agent, leucovorin, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, capecitabine, temozolomide, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin and nib.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 illustrates results from Example 15 for the compound of Example 2 and docetaxel in LuCap35V primary prostate tumors with HScore of 200.
Figure 2 illustrates results from Example 15 for the compound of Example 2 dosed intermittently either PO or IP and docetaxel in PC3-NCI prostate .
Figure 3 illustrates results from Example 15 for the compound of Example 2 dosed PO and docetaxel in PC3-NCI prostate tumors.
Figure 4 illustrates s from Example 15 for the compound of Example 2 dosed IP intermittently and docetaxel in MCF7-neo/HER2 tumors.
Figure 5 rates results from Example 15 for the compound of Example 2 dosed PO and docetaxel in MCF7-neo/HER2 breast .
Figure 6 illustrates results from Example 15 for the compound of Example 2 and docetaxel in MAXF401 y tumors.
Figure 7 illustrates results from Example 15 for the compound of e 2 and docetaxel in SKOV3 ovarian tumors.
Figure 8 illustrates results for the compound of Example 2 and cisplatnin in SKOV3 n tumors.
Figure 9 illustrates results from e 15 for the compound of Example 2 dosed PO and latin in IGROV-1 ovarian tumors.
Figure 10 illustrates results from Example 15 for the compound of Example 2 and 0 in LuCap35V cells.
Figure 11 illustrates results of the combination of GDC-0068 and B20-4.1.1 (murine antiVEGF dy) in a breast cancer model.
Figure 12 illustrates data from Example 14 that shows that representative combinations provide additive or synergistic activity against a number of cancer types.
Figure 13 rates data from Example 14 showing the activity of Example 2 plus 5- FU/Cisplatin is associated with AKT pathway activation, particularly in gastric and head and neck squamous cell carcinoma. Additive effects were observed for the combination of GDC- 0068 plus 5-FU/cisplatin, and are associated with PTEN (low or null), pAKT xpression) and PI3K mutation and amplification.
Figure 14 illustrates BLISS score data from Example 14 g the activity of Example 2 (GDC-0068) plus 5-FU/Cisplatin (“chemo”) combinations in Gastric cell lines.
Synergy is trated in the combination in NUGC3 cell lines (Gastric cancer) where PTEN status is low and pAKT is overexpressed. Additionally, this particular cell line (NUGC3) shows additive effects at vel doses of 5-Fu/Cisplatin and high doses of GDC-0068.
Figure 15 illustrates data from Example 14 showing that Example 2 plus Docetaxel combinations show maximum effect in PTEN null line which had minimal single agent response to Example 2.
Figure 16 illustrates data from Example 14 that shows that Example 2 plus Docetaxel combinations are weaker in PTEN normal cell lines.
Figure 17 shows data for the sequencing combination of Akt inhibitor Formula 1A (GDC-0068) with DTX in the LuCap145.2 PTEN null primary prostate cancer xenograft model.
Figure 18 shows data for Formula 1A 068) dosed PO + docetaxel in MCF-7 breast tumors.
Figure 19 shows data for Formula 1A 068) dosed PO + carboplatin in OVCAR3 ovarian tumors.
Figure 20 shows data for single agent GDC-0068 in HGC-27 (Her2(-) & PTEN null) gastric tumor xenograft.
Figure 21 shows the PET scan responses for breast cancer patients treated with GDC- 0068 single agent therapy.
Figure 22 shows PET and tumor marker response for one breast cancer patient treated with GDC-0068 single agent therapy.
Figure 23 shows results of one patient having Akt1 E17K mutation breast cancer with partial se from one cycle of ent with GDC-0068 in combination with xel after failing multiple other chemotherapy ents.
Figure 24 shows results of a treatment of GDC-0068 in combination with FOLFOX with partial response where patient suffered from PIK3CA mutant squamous carcinoma of cervix, after failing prior treatments.
Figure 25 shows results of a treatment of GDC-0068 in combination with FOLFOX with partial response where patient suffered from PTEN-Loss (Hscore 40), KRAS-Wild-Type Colorectal Cancer, after g prior treatments.
Figure 26 shows Western Blot data showing PD response in LuCaP35V tumors treated with GDC-0068 in combination with MDV3100 for 3 and 8 hours.
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS AND DEFINITIONS The words "comprise," "comprising," de," "including," and "includes" when used in this specification and claims are intended to specify the presence of stated features, integers, components, or steps, but they do not preclude the ce or addition of one or more other features, integers, components, steps, or groups thereof.
The term "alkyl" as used herein refers to a saturated linear or branched-chain lent hydrocarbon radical of one to twelve carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, - CH 2CH 3), 1-propyl (n-Pr, n-propyl, -CH2CH 2CH 3), yl (i-Pr, i-propyl, -CH(CH3)2), 1- butyl (n-Bu, n-butyl, -CH2CH 2CH 2CH 3), 2-methylpropyl (i-Bu, i-butyl, -CH2CH(CH 3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH 2CH 3), 2-methylpropyl (t-Bu, l, -C(CH3)3), 1- pentyl (n-pentyl, -CH2CH 2CH 2CH 2CH 3), 2-pentyl (-CH(CH3)CH 2CH 2CH 3), 3-pentyl (- CH(CH 2CH 3)2), 2-methylbutyl (-C(CH3)2CH 2CH 3), 3-methylbutyl (- CH(CH 3)CH(CH 3)2), 3-methylbutyl (-CH2CH 2CH(CH 3)2), ylbutyl (- CH 2CH(CH 3)CH 2CH 3), 1-hexyl (-CH2CH 2CH 2CH 2CH 2CH 3), 2-hexyl (- CH(CH 3)CH 2CH 2CH 2CH 3), 3-hexyl (-CH(CH2CH 3)(CH 2CH 2CH 3)), ylpentyl (- C(CH 3)2CH 2CH 2CH3), 3-methylpentyl (-CH(CH3)CH(CH 3)CH 2CH 3), 4-methylpentyl (- CH(CH 3)CH 2CH(CH 3)2), 3-methylpentyl (-C(CH3)(CH 2CH 3)2), 2-methylpentyl (- CH(CH 2CH 3)CH(CH 3)2), 2,3-dimethylbutyl (-C(CH3)2CH(CH 3)2), 3,3-dimethylbutyl (- CH(CH 3)C(CH 3)3, 1-heptyl, 1-octyl, and the like.
The term "alkenyl" refers to linear or branched-chain monovalent hydrocarbon radical of two to twelve carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp2 double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents bed herein, and includes radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" ations. Examples include, but are not limited to, ethylenyl or vinyl (-CH=CH 2), allyl (-CH2CH =CH 2), and the like.
The term "alkynyl" refers to a linear or branched lent hydrocarbon radical of two to twelve carbon atoms with at least one site of ration, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynyl (-C≡CH), propynyl (propargyl, -CH2C≡CH), and the like.
The terms cycle", cyclyl", "carbocyclic ring" and "cycloalkyl" refer to a monovalent non-aromatic, saturated or lly unsaturated ring having 3 to 12 carbon atoms as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring. Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as d systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Examples of monocyclic ycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentenyl, 1-cyclopentenyl, 1-cyclopent enyl, cyclohexyl, 1-cyclohexenyl, 1-cyclohexenyl, 1-cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
"Aryl" means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring . Some aryl groups are represented in the exemplary structures as "Ar". Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Typical aryl groups include, but are not limited to, ls derived from benzene (phenyl), substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1,2-dihydronapthalene, 1,2,3,4- tetrahydronapthyl, and the like. Aryl groups are optionally substituted independently with one or more substituents bed .
The terms "heterocycle," "hetercyclyl" and "heterocyclic ring" are used interchangeably herein and refer to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) carbocyclic radical of 3 to 20 ring atoms in which at least one ring atom is a heteroatom selected from nitrogen, oxygen and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described below. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 6 atoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are bed in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem.
Soc . (1960) 82:5566. The term "heterocycle" includes heterocycloalkoxy. "Heterocyclyl" also includes ls where cycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, ydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, nyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, yl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl, 3-azabicyco[3.1.0]hexanyl, 3- azabicyclo[4.1.0]heptanyl, yclo[2.2.2]hexanyl, 3H-indolyl izinyl and N-pyridyl ureas. Spiro moieties are also ed within the scope of this tion. Examples of a heterocyclic group wherein 2 ring carbon atoms are substituted with oxo (=O) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described .
The term "heteroaryl" refers to a monovalent aromatic radical of 5-, 6-, or 7- membered rings, and includes fused ring systems (at least one of which is aromatic) of 5-20 atoms, containing one or more heteroatoms independently ed from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups are pyridinyl (including, for example, 2- hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4- hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, uranyl, inyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, l, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. Heteroaryl groups are optionally tuted independently with one or more substituents described herein.
The heterocycle or heteroaryl groups may be carbon (carbon-linked), nitrogen gen-linked) or oxygen n-linked) attached where such is le. By way of example and not limitation, carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, ene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, on 2, 3, or 4 of an ine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
By way of example and not limitation, nitrogen bonded heterocycles or aryls are bonded at position 1 of an ine, azetidine, pyrrole, pyrrolidine, oline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, dine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or βcarboline.
The terms "treat" and "treatment" refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the growth, development or spread of cancer. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not ing) state of disease, delay or g of disease progression, amelioration or palliation of the disease state, and ion (whether partial or total), whether detectable or ctable.
"Treatment" can also mean prolonging survival as compared to expected survival if not ing treatment. Those in need of ent include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
The phrase "therapeutically effective amount" means an amount of a compound useful in the present invention that (i) treats the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or ates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. In the case of cancer, the therapeutically ive amount of the drug may reduce the number of cancer cells; reduce the tumor size; t (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
The terms r" and rous" refer to or describe the physiological condition in s that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells. es of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the neum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic , astoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, ma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, te cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer. Gastric cancer, as used herein, includes stomach cancer, which can develop in any part of the h and may spread throughout the stomach and to other organs; particularly the esophagus, lungs, lymph nodes, and the liver.
A "chemotherapeutic agent" is a biological (large molecule) or al (small molecule) compound useful in the treatment of cancer, regardless of mechanism of action.
Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase tors, ns, antibodies, ensitizers, and kinase inhibitors. Chemotherapeutic agents include compounds used in “targeted y” and non-targeted conventional chemotherapy.
The term "mammal" includes, but is not limited to, humans, mice, rats, guinea pigs, monkeys, dogs, cats, horses, cows, pigs, sheep, and poultry.
The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, indications and/or gs concerning the use of such therapeutic products.
The phrase "pharmaceutically acceptable salt" as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound useful in the invention.
Exemplary salts include, but are not limited, to e, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, e, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate “mesylate”, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1'-methylene-bis -(2-hydroxynaphthoate)) salts. A pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion. The counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically able salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ion.
If the compound is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for e, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, ic acid, nitric acid, methanesulfonic acid, oric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a sidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as ic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like. Acids which are generally considered suitable for the formation of pharmaceutically useful or acceptable salts from basic pharmaceutical compounds are sed, for e, by P.
Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al, l of ceutical Sciences (1977) 66(1) 1 19; P. Gould, ational J. of Pharmaceutics (1986) 33 201 217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; Remington’s Pharmaceutical Sciences, 18th ed., (1995) Mack hing Co., Easton PA; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference thereto.
If the compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative es of suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, y, secondary, and tertiary amines, and cyclic amines, such as dine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
The phrase "pharmaceutically able" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
A "solvate" refers to a physical association or complex of one or more solvent molecules and a compound useful in the invention. The compounds may exist in unsolvated as well as solvated forms. Examples of solvents that form es include, but are not limited to, water, panol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to the complex where the solvent molecule is water.
This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the lline solid. Preparation of solvates is generally known, for example, M. Caira et al, J.
Pharmaceutical Sci., 93(3), 601 611 (2004). Similar preparations of solvates, hemisolvate, hydrates and the like are described by E. C. van Tonder et al, AAPS PharmSciTech., 5(1), article 12 (2004); and A. L. Bingham et al, Chem. Commun., 603 604 (2001). A typical, nonlimiting , process es ving the inventive compound in desired amounts of the desired solvent (organic or water or es f) at a higher than ambient temperature, and cooling the solution at a rate sufficient to form crystals which are then ed by standard methods. Analytical techniques such as, for example I.R. spectroscopy, show the presence of the solvent (or water) in the crystals as a solvate (or hydrate).
The term "synergistic" as used herein refers to a therapeutic combination which is more ive than the additive effects of the two or more single agents. A determination of a synergistic interaction between a compound of formula I or a pharmaceutically able salt thereof and one or more chemotherapeutic agent may be based on the results obtained from the assays described herein. The results of these assays can be analyzed using the Chou and Talalay combination method and Dose-Effect Analysis with CalcuSyn software in order to obtain a Combination Index (Chou and Talalay, 1984, Adv. Enzyme Regul. 55).
The combinations provided by this invention have been evaluated in several assay systems, and the data can be analyzed ing a standard program for quantifying synergism, additivism, and antagonism among anticancer agents. The program utilized, for example in Figure 12, is that described by Chou and Talalay, in "New Avenues in Developmental Cancer Chemotherapy," Academic Press, 1987, Chapter 2. Combination Index values less than 0.8 indicates synergy, values greater than 1.2 te nism and values between 0.8 to 1.2 indicate additive effects. The combination therapy may provide "synergy" and prove "synergistic", i.e., the effect ed when the active ingredients used together is greater than the sum of the effects that s from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different ions in separate es. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination y, effective dosages of two or more active ingredients are administered together. In some examples, Combination effects were evaluated using both the BLISS ndence model and the t single agent (HSA) model (Lehár et al. 2007, Molecular Systems Biology 3:80). BLISS scores quantify degree of potentiation from single agents and a BLISS score > 0 suggests greater than simple vity. An HSA score > 0 suggests a combination effect greater than the maximum of the single agent responses at corresponding concentrations. se Evaluation Criteria in Solid Tumors, n 1.1 (RECIST v1.1), were used to evaluate tumor responses in certain human clinical trials. This section provides the definitions of the ia used to determine objective tumor se for target lesions. ete response” (CR) is used to mean disappearance of all able target lesions with pathological lymph nodes (whether target or non-target) having reduction in short axis to less than about 10 mm. “Partial se” (PR) is used to mean at least about a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum of diameters. “Progressive disease” (PD) is used to mean at least about a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (nadir), including baseline. In addition to the relative increase of about 20%, the sum also demonstrates an absolute increase of at least about 5 mm. In one e, the ance of one or more new lesions is considered PD. “Stable disease” (SD) is used to mean neither ient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum on study.
Adverse Event Grading (Severity) Scale is used to evaluate safety and tolerability with Grade 1 is mild (intervention not indicated), Grade 2 is moderate (minimal, local, or noninvasive intervention indicated), Grade 3 is severe (severe or medically significant but not immediately life threatening; hospitalization or prolongation of hospitalization indicated), Grade 4 is very severe, life threatening or disabling, urgent intervention indicated, and Grade is death related to the adverse event.
Described is a method for ng the hyperproliferative disorder wherein administration of the compound of formula I or the salt thereof and the one or more agents selected from 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN- 38, capecitabine, temozolomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin, lapatinib, 32, MDV3100, abiraterone, and GDC- 0973 provides a synergistic effect in treating the hyperproliferative disorder. In a further aspect, the synergistic effect has a ation Index value of less than about 0.8.
In preclinical models, GDC-0068 administration resulted in a dose-dependent se in plasma e levels. Grade 1 or 2 hyperglycemia events were observed in a Phase Ia human clinical trial of GDC-0068 among fasted patients and were relieved with a combination of oral anti-diabetic therapy and diet. ore, bed is a method of treating a hyperproliferative disease, such as cancer, in a patient suffering therefrom comprising administering a compound of formula I (for example, GDC-0068) in combination with an anti-diabetic compound (for example metformin). The combination of anti-diabetic therapies prevents, treats or reverses hyperglycemia ffects of the formula Ia compound treatments. In one example, GDC-0068 is administered on an empty stomach (fasting), optionally in ation with anti-diabetic therapies, and in combination with the chemotherapeutic agents described herein. In other embodiments, chemotherapeutic agents (further sed herein, for example docetaxel and folfox) are also stered as part of the combination.
FORMULA I COMPOUNDS Formula I compounds include a nd of formula I: R1 N R5 R2O N R10 I, and pharmaceutically acceptable salts thereof, wherein: R1 is H, Me, Et, vinyl, CF3, CHF2 or CH2F; R2 is H or Me; R5 is H, Me, Et, or CF3; R6 R7 (CRcRd)n (CH2)m (CRaRb)p O A is ; G is phenyl optionally substituted by one to four R9 groups or a 5-6 membered heteroaryl optionally substituted by a halogen; R6 and R7 are independently H, OCH3, (C3-C6 lkyl)-(CH2), (C3-C6 cycloalkyl)- (CH2CH2), V-(CH2)0-1 wherein V is a 5-6 membered heteroaryl having from one to two ring heteroatoms independently selected from N, O and S, W-(CH2)1-2 wherein W is phenyl optionally substituted with F, Cl, Br, I, OMe, CF3 or Me, C3-C6-cycloalkyl optionally substituted with C1-C3 alkyl or O(C1-C3 alkyl), hydroxy-(C3-C6-cycloalkyl), fluoro-(C3-C6- cycloalkyl), CH(CH3)CH(OH)phenyl, 4-6 membered heterocycle optionally substituted with F, OH, alkyl, cyclopropylmethyl or C(=O)(C1-C3 alkyl), or C1-C6-alkyl optionally substituted with one or more groups independently selected from OH, oxo, O(C1-C6-alkyl), CN, F, NH2, NH(C1-C6-alkyl), N(C1-C6-alkyl)2, cyclopropyl, phenyl, imidazolyl, piperidinyl, pyrrolidinyl, morpholinyl, tetrahydrofuranyl, oxetanyl, or tetrahydropyranyl, or R6 and R7 together with the nitrogen to which they are attached form a 4-7 membered heterocyclic ring, wherein said heterocyclic ring is optionally substituted with one or more groups ndently selected from OH, halogen, oxo, CF3, CH2CF3, CH2CH2OH, O(C1-C3 alkyl), C(=O)CH3, NH2, NHMe, N(Me)2, S(O)2CH3, cyclopropylmethyl and C1-C3 alkyl; Ra and Rb are H, or Ra is H, and Rb and R6 together with the atoms to which they are attached form a 5- 6 membered heterocyclic ring having one or two ring nitrogen atoms; Rc and Rd are H or Me, or Rc and Rd er with the atom to which they are attached from a cyclopropyl ring; R8 is H, Me, F or OH, or R8 and R6 together with the atoms to which they are attached form a 5-6 membered heterocyclic ring having one or two ring nitrogen atoms; each R9 is independently halogen, C1-C6-alkyl, C3-C6-cycloalkyl, O-(C1-C6-alkyl), CF3, OCF3, 6-alkyl), CN, OCH2-phenyl, CH2O-phenyl, NH2, NH-(C1-C6-alkyl), N- (C1-C6-alkyl)2, piperidine, pyrrolidine, CH2F, CHF2, OCH2F, OCHF2, OH, SO2(C1-C6-alkyl), C(O)NH2, (C1-C6-alkyl), and C(O)N(C1-C6-alkyl)2; R10 is H or Me; and m, n and p are independently 0 or 1.
A specific compound of a I is a nd wherein A is Rc N R7 A specific compound of Formula I is a compound Formula Ia: HO Ia, or a pharmaceutically acceptable salt thereof.
In one embodiment described the compound of formula I excludes the compound (S)- 2-(4-chlorophenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinyl)(isopropylamino)propanone Formula Ia: HO Ia, and pharmaceutically acceptable salts thereof (this compound may also be ed to as GDC-0068).
PREPARATION OF FORMULA I COMPOUNDS nds useful in this invention may be synthesized by synthetic routes that include processes analogous to those well known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by s generally described in Louis F. Fieser and Mary , ts for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed.
Springer-Verlag, Berlin, including supplements).
Compounds of Formula I may be prepared singly or as compound ies comprising at least 2, for e 5 to 1,000 compounds, or 10 to 100 compounds. Libraries of nds of Formula I may be prepared by a combinatorial 'split and mix' approach or by multiple parallel ses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art. Thus also described is a nd library comprising at least 2 compounds of Formula I, or salts thereof.
For illustrative purposes, s 1-4 and Schemes A-J shows a general method for preparing the compounds useful in the present ion as well as key intermediates. For a more detailed description of the individual on steps, see the Examples section below.
Those d in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific ng materials and reagents are depicted in the s and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
S OH OH Cl MeOOC Reduction H2N NH2 N Chlorination N N O HS N N N 1 2 3 4 Boc Boc N N Cl O O Cl N N SNAr Oxidation N O Hydrolysis N N N N N N N OAc OAc O OH 6 7 8 R O N N 1. Acylation 2. HCl N N N N R6 R7 N N OH OH (CRcRd)n 9 10 (CH2)m (CRaRb)p G R8 Scheme 1 Scheme 1 shows a method of preparing compound 10 of Formula I wherein R1 is H, R2 is H and R5 is H. Formation of pyrimidine 2 can be accomplished by the reaction of the keto ester 1 with thiourea in the presence of a base such as KOH in an appropriate solvent, such as ethanol. After reduction of the mercapto group of compound 2 under standard reducing conditions (e.g., Raney Ni and NH4OH) to provide compound 3, the hydroxypyrimidine 3 can be chlorinated under standard conditions (e.g., POCl3 in DIEA/DCE) to provide compound 4. Compound 4 is then oxidized under standard conditions (e.g., MCPBA in an appropriate solvent such as CHCl3) to give the pyrimidineoxide . Treatment of the pyrimidine-oxide with acetic anhydride gives the rearrangement product 6. nd 7 is obtained by reacting compound 6 with an appropriately substituted piperidine under rd SNAr reaction conditions to provide compound 7.
Compound 7 is hydrolyzed to e compound 8, which is then deprotected to yield the intermediate 9. Acylation of the piperazinyl cyclopenta[d]pyrimidine 9 with an appropriated amino acid in the presence of a coupling reagent such as HBTU, followed by deprotection if necessary, gives nd 10 of Formula I.
O O 2O Br COOEt O3 COOEt O H2N NH2 11 12 13 14 (+)-pulegone OH OH Cl Acetic chlorination N anhydride N ion N oxidation HS N N N N 16 17 18 R O Boc Boc 1. HCl N N 2. Acylation N LiOH 3. HCl N N N N N N N N N Cl OAc OH OH N 20 21 22 Boc Boc 1.HCl R O 19 2 Acylation N LiOH N 3. HCl N N N N N N N N N N OAc OH OH 23 24 25 R O Boc 1.HCl N 2 Acylation N R = R6 R7 3. HCl (CRcRd)n N N N N (CH2)m (CRaRb)p N N G OMe OMe 26 27 Scheme 2 Scheme 2 shows a method of preparing compounds 22, 25 and 27 of Formula I wherein R1, R2 and R5 are methyl. According to Scheme 2, bromination of (+)-pulegone 11 with bromine gives the dibromide 12. The treatment of the dibromide 12 with a base such as sodium ethoxide provides the pulegenate 13. Ozonolysis of the pulegenate 13 gives the ketoester 14. Treatment of the keto ester 14 with ea in the presence of a base such as KOH in ethanol, followed by reduction of the mercapto group under standard conditions (e.g., Raney Ni catalyst in ammonia) affords the hydroxypyrimidine 16. nation of the hydroxypyrimidine 16 under standard conditions (e.g., POCl3) provides the 4- chloropyrimidine 17. The oxidation of the 4-chloropyrimidine 17 with an oxidizing agent such as MCPBA or hydrogen peroxide provides the N-oxide 18. Rearrangement of the N- oxide 18 with acetic anhydride yields the intermediate 19. Compound 19 is reacted with the desired zine according to the procedure described in Scheme 1 to e nd 20 where R5 is H and 23 where R5 is Me. Compounds 20 and 23 are ted to chiral separation using HPLC with chiral stationary and then hydrolyzed upon treatment with a base such as lithium hydroxide to provide compounds 21 and 24, respectively. After deprotection, compounds 21 and 24 are then reacted with the appropriate amino acid to provide compounds 22 and 25, respectively.
Alternatively, the 7-hydroxy group of compound 24 may be alkylated with an alkylation reagent such as an alkyl halide in the presence of a base such as NaH or KOH to provide compound 26 where R2 is Me. After deprotection, nd 26 is then reacted with the appropriate amino acid to provide compound 27.
O NH4+ OH NH4OAc -O O Halogenation O O N O H2N O N 14 63 N Boc Boc N N R5 H Oxidation Ac2O N R5 N N R5 N N 65 N 66 67 O Boc Boc Boc N N N N R5 Hydrolysis N R5 ion N R5 Asymmetric Reduction N N N N N N AcO HO O 68 69 70 R O Boc R O N Boc N 1. HCl N OR N 2. Acylation OR N R5 N R5 3. Functionalisation N R5 N R5 N N HO N HO N 72 73 HO 71 74 R = R6 R7 (CRcRd)n R5= H, Me, Et, CF3 (CH2)m (CRaRb)p Scheme 3 Scheme 3 shows an ative method of preparing compounds 73 and 74.
According to Scheme 3, ion of 14 using an ammonia synthon gives 63. Pyrimidine ion using, for example, ammonium formate in the presence of formamide at 50°C- 250°C and/or at high pressure gives the bicyclic unit 64. Activation of 64 using, for example, POCl3 or SOCl2 gives the activated pyrimidine 65. Displacement of this g group, using a suitable protected/substituted piperazine at 0°C to 150°C gives the piperazine 66.
Oxidation, using, for example, m-chloroperoxybenzoic acid (“MCPBA” or “m-CPBA”) or Oxone® at -20°C to 50°C gives the N-oxide 67. Treatment with an acylating agent (e.g., acetic anhydride) followed by heating (40°C to 200°C) causes rearrangement to give 68.
Hydrolysis, using, for example LiOH or NaOH at 0°C to 50°C gives the alcohol 69.
Oxidation, using for example, Swern conditions, MnO4 or pyridine-SO3 complex at appropriate temperatures gives the ketone 70. Asymmetric reduction using, for e, a catalytic chiral catalyst in the presence of hydrogen, the CBS catalyst or a borohydride reducing agent in the presence of a chiral ligand gives rise to either the (R) or the (S) stereochemistry at the alcohol 71 or 72. atively, a non-chiral reducing agent could be used (e.g., H2, Pd/C), allowing the methyl group on the cyclopentane unit to provide facial ivity and ultimately diastereoselectivity. If the reduction gives a lower diastereoselctivity, the diastereomers could be separated by (for example) chromatography, llization or derivitization. Finally deprotection of the Boc-group, using, for example, acid at 0°C to 50°C, acylation using an appropriately functionalized amino acid and final functionalization of the amine of this amino acid (e.g., removal of any protecting group, alkylation, reductive amination or acylation to introduce new substituents) gives rise to the final compounds 73 and 74.
R' X Acylation R' Lewis Acid NBoc Saponification S O S HO2C R X R' N O (2) O S (1) (3) O OH N R' Scheme 4 Introduction of a chiral auxiliary (e.g., Evans oxazolidinone, etc.) to nd 1 may be accomplished by standard acylation procedures to give the ate 2. For example, treatment of the acid with an activating agent (e.g., COCl2) or mixed anhydride ion (e.g., 2,2-dimethylpropanoyl chloride) in the presence of an amine base at -20°C to 100°C followed by treatment with the appropriate chiral auxiliary (X)δ gives nd 2. The stereochemistry and choice of the chiral auxiliary may determine the stereochemistry of the newly created chiral center and the reoselectivity. Treatment of compound 2 with a Lewis acid (e.g., TiCl4) at low temperature (e.g., -20°C to -100°C) and an amine base (e.g., Hunig’s base) followed by the use of an appropriately substituted imminium ion sor 3 at low temperature then gives rise to compound 4. The temperature, Lewis acid and chiral ary may all be expected to influence the diastereoselectivity of the addition adduct.
Finally, saponification under mild conditions (e.g., LiOH/H2O at -10°C to 30°C) gives rise to the desired acid 5.
Also bed is a method of preparing a compound of Formula I, comprising: reacting a compound having the formula: R5 N R1 N R10 wherein R1, R2, R5 and R10 are as defined herein, with an amino acid having the formula: R6 R7 (CRcRd)n (CH2)m (CRaRb)p O OH wherein R6, R7, Ra, Rb, Rc, Rd, G, m, n and p are as defined herein.
The amino acids used in the synthesis of compounds of Formula I as illustrated in Schemes 1-4 and in the Examples are either commercially available or may be prepared according to the methods disclosed herein. For example, in certain embodiments the amino acids used to e compounds of Formula I include β-phenylglycine amino acids having the Formula 1A, γ-phenylglycine amino acids having the Formula 2A, β-phenylalanine amino acids having the Formula 3A, and γ-phenylalanine amino acids having the a R6 R7 R7 N R6 N Rc Rc N R7 R6 Rb R7 R8 Rd Ra N O G O O G O R8 R8 OH OH R8 OH OH 1A 2A 3A 4A Methods of preparing amino acids of Formulas 1A-4A are shown in Schemes A-J.
CO2H CO2R' Hydroxylmethylation CO2R' (R9)t (R9)t (R9)t 21 22 1. Activation 2. Elimination NPg 1. Addition of primary amine CO2R' (R9)t 2. Protection CO2R' (R9)t of amine (Pg) 23 24 1. Addition of secondary amine Acid formation 2. Acid formation R6 R6 NPg N CO2H CO2H (R9)t (R9)t 26 Scheme A Scheme A illustrates a method of preparing optionally substituted β-phenylglycine amino acids 25 and 26 of the Formula 1A wherein R8 is H, and R6, and R9 and are as defined herein, t is 0 to 4, and R7 is H or an amine ting group. ing to Scheme A, the acid 20 is converted to an ester 21 wherein R' is alkyl using standard conditions such as treatment with an appropriate alcohol (e.g., MeOH) in the presence of a catalytic amount of an acid such as concentrated H2SO4 or a coupling agent such as DCC/DMAP; or alternatively by treatment with an appropriate electrophile (e.g., MeI, EtBr, BnBr) in the presence of a base such as NEt3/DMAP at an appropriate temperature (e.g., -20°C to 100°C). The appropriate choice of ester is determined by the conditions required to reform the acid at the end of the sis, with many appropriate examples and ions being listed in ‘Protective Groups in Organic sis’ by Greene and Wuts, Wiley-Interscience, third n, Chapter 5. Introduction of the hydroxymethyl group to provide compound 22 may be performed by treatment with an appropriate aldehyde (e.g., formaldehyde) in the presence of base such as NaOEt at an appropriate temperature (e.g., -20°C to room temperature).
Activation of the alcohol group of compound 22 to form a leaving group (e.g., a mesylate, tosylate, halide) may be accomplished by treatment with, for example, methanesulphonyl chloride in the presence of excess base such as NEt3, DIPEA, or DBU at an appropriate temperature (e.g., -20°C to room temperature). In many cases the olefin 24 can be isolated directly from this ure, in other cases warming (30°C to 100°C) or additional base (e.g., DBU in the case of halide) may be required to complete the elimination to provide compound 24. The activated olefin 24 may be d with the desired primary amine (e.g., ethylamine) in a suitable solvent, such as THF, at an appropriate temperature (e.g., -20°C to reflux) to generate the amino ester intermediate. In the case wherein compound 24 has an electron rich aromatic ring or on poor/bulky primary amine, g (e.g., 30-240°C in a sealed tube) or microwave chemistry may be required. Protection of the amine group (for e as Boc-group) may be accomplished using Boc2O under standard conditions to provide compound 23 wherein Pg is a protecting group. ative protecting groups may be used, and many appropriate examples are listed in ‘Protective Groups in Organic Synthesis’ by Greene and Wuts, Wiley-Interscience, third edition, Chapter 7. Saponification of the ester 23 to form the protected amino acid 25 may be accomplished using ions appropriate for the ester (e.g., aqueous LiOH for methyl , hydrogenation for benzyl esters, acid for t- butyl esters).
Alternatively, the activated olefin 24 may be treated with a secondary amine (e.g., diethylamine) in a suitable solvent such as THF at an appropriate temperature (e.g., -20°C to reflux) to generate the aminoester intermediate (not shown). In the case wherein compound 24 has an electron rich ic ring or electron poor/bulky ary amine, heating (e.g., 30-240°C in a sealed tube) or microwave chemistry may be required. Saponification of the ester to form the amino acid 26 may be accomplished using conditions appropriate for the ester (e.g., aqueous LiOH for methyl esters, hydrogenation for benzyl esters, acid for t-butyl esters, etc.).
In an alternative to Scheme A, Pg may be tuted with R7 in compounds 23 and 1. Addition of CO2R' N (R9)t secondary amine R7 2. Acid formation CO2H 24 (R9)t Scheme A1 Scheme A1 shows an alternative to Scheme 1, wherein the activated olefin 24 is reacted to form the amino acid 26A.
O N Oxidant 1. R6NH2 HO Pg CO2R' CO2R' (R9)t (R9)t CO2R' 2. Protection (R9)t 24 28 1. R6R7NH 2. ection Deprotection R6 R6 N N HO R7 HO Pg CO2H (R9)t CO2H (R9)t 31 Scheme B Scheme B shows a method of preparing optionally substituted β-phenylglycine amino acids 30 and 31 of Formula 1A wherein R8 is OH, and R6, and R9 are as d herein, t is 0 to 4, and R7 is as defined herein or an amine protecting group. Oxidation of the unsaturated ester 24 (prepared according to Scheme A), n t is 0-4 and R' is alkyl, using a rd oxidizing agent such as MCPBA at an appropriate temperature (room temperature to reflux) provides the epoxide intermediate 28. Intermediate 28 may be treated with an appropriate amine, typically at high temperature (e.g., 50-300°C) and high pressure (e.g., in a sealed tube or a bomb) to give the amino alcohol 29 or 30. If a secondary amine is used (such as in the preparation of compound 30), then deprotection of the ester using conditions listed in ctive Groups in Organic Synthesis’ by Greene and Wuts, Wiley-Interscience, third edition, Chapter 5 may be used (e.g., LiOH for a methyl ester, hydrogenation for a benzyl ester, etc). When a primary amine is used (such as in the preparation of compound 29), protection of the amine (e.g., as a oup using Boc anhydride) followed by deprotection of the ester (using the above conditions) provide the ylated amino acid 31.
CO2tBu CO2H 1. Base R8 Deprotection R8 CO2R''' CO2R''' CO2R''' (R9)t (R9)t (R9)t 2. Br CO2tBu 32 33 34 Curtius NHPg NHPg Deprotection R8 R8 CO2H CO2R''' (R9)t (R9)t 36 35 Scheme C Scheme C shows a method of preparing optionally substituted β-phenylglycine amino acids 36 of the a 1A wherein R8 is methyl, R6 is H, R7 is an amine protecting group t is 0 to 4, and R9 is as defined herein. The ester 32, wherein R''' is alkyl, can be d with a base (e.g., NaOtBu) at an appropriate temperature (e.g., 0°C to reflux) to form the anion, followed by addition of an electrophile (e.g., utyl 2-bromoacetate) at an appropriate temperature (e.g., -78°C to room temperature) to give the homologated ester 33. Removal of the t-butyl ester of compound 33 using an appropriate acid such as TFA or HCl at an appropriate temperature (e.g, 0°C to reflux) es compound 34. A Curtius rearrangement of compound 34 using, for example, DPPA in the presence of mild base such as NEt3 at an appropriate temperature (e.g., 0°C to ), followed by treatment of the ve intermediate with an alcohol (e.g., t-BuOH), optionally in the presence of a Lewis acid (e.g., SnCl2) at higher temperature (e.g., 40-200°C) provides compound 35 wherein Pg is an amine protecting group. The choice of alcohol used to prepare compound 35 determines the amine protecting group (e.g., t-BuOH es the Boc-amine). Deprotection of the ester group of compound 35 using standard conditions (e.g., with LiOH when the protecting group is a methyl ester, hydrogenation for a benzyl ester, etc.) gives the acid compound 36.
In one alternative of Scheme C, R8 may be methyl, H or F.
In another alternative of Scheme C, Pg may be substituted with R7 in compounds 35 and 36.
NO2 Rc RcRdCHNO2 Rc Reduction CO2R' Rd NH (R9)t Base CO2R' (R9)t (R9)t 37 38 Protection NHBoc Rc Rd Rd Hydrolysis NBoc CO2H (R9)t (R9)t 40 39 Scheme D Scheme D shows a method of preparing optionally substituted γ-phenylglycine amino acids 40 of Formula 2A wherein Rc, Rd, and R9 are as defined herein t is 0 to 4, R6 is H, and R7 is an amine ting group such as Boc. The starting unsaturated ester 24, prepared according to Scheme A, can be treated with a substituted nitromethane tive (e.g., nitroethane) in the presence of a base such as DBU at an appropriate temperature (e.g., 0°C to room ature) to give the homologated adduct 37. The nitro group of compound 37 can be reduced using standard conditions (e.g., hydrogenation, Zn/acid, etc.) at an appropriate temperature (e.g., room temperature to reflux), and the resulting intermediate can be cyclized to give the lactam intermediate 38. Protection of the amine, for example with a Boc-group to provide compound 39, may be accomplished using Boc2O under standard conditions.
Alternative ting groups may be used, and many appropriate examples are listed in ‘Protective Groups in c sis’ by Greene and Wuts, Wiley-Interscience, third edition, Chapter 7. Treatment of compound 39 with an s base such as LiOH or KOH at an appropriate temperature (e.g., 0 to 100°C) effects ring opening of the lactam to give the appropriately substituted protected amino acid compound 40.
In one alternative of Scheme D, Boc may be replaced with R7 in compounds 39 and NHR7 NHR7 NHR7 NHR7 Introduce Rc Rc Separation Rc Rc chiral auxilary Rd Rd Rd Rd O O O CO2H (R9)t (R9)t (R9)t (R9)t Χ Χ Χ 40 40a 40b 40c Chiral ry Chiral Separation cleavage NHR7 NHR7 Rc Rc Rd Rd O O (R9)t (R9)t OH OH 40d 40e Scheme D1 Scheme D1 shows representative methods of forming the single enantionmers of the gamma amino acids 40d and 40e, wherein Rc, Rd, and R9 are as defined herein, t is 0 to 4, R6 is H, and R7 is an amine protecting group such as Boc. In one possible method, the racemic amino acid is subject to chiral chromatographic separation using a chiral stationary phase.
Alternatively, a diastereomeric mixture may be prepared which could be separated by conventional chromatographic techniques. For example, activation of compound 40 (e.g., COCl2, base) and introduction of a chiral ary (e.g., an Evans’ idinone) in the presence of a basic amine (e.g., Hunig’s base) at -20°C to 50°C gives the diastereomeric mixture of compounds 40b and 40c. This mixture may be separated using standard conditions (e.g., column chromatography, HPLC, SFC, etc.) to give the individual diastereomers. These may be converted to the desired acids by cleavage of the chiral auxiliary (in the case of an Evans’ auxiliary, by using (for example) OOH at -15°C to room temperature) to give the compounds 40d and 40e. The temperature may need to be kept low so as to t racemisation of the newly separated chiral center.
CO2tBu CO2H CO2tBu R8 Deprotection R8 CO2R''' CO2R''' CO2R''' (R9)t Base (R9)t (R9)t 32 41 42 NHPg NHPg Deprotection R8 R8 CO2H CO2R''' (R9)t (R9)t 44 43 Scheme E Scheme E shows a method of making optionally substituted γ-phenylglycine amino acids 44 of a 2A wherein R8 is , R6 is H, R7 is an amine protecting group, t is 0 to 4, and R9 is as defined herein. The ester 32, wherein R''' is alkyl and t is 0-4, can be treated with a le base such as KOtBu at an appropriate temperature (e.g., 0°C to reflux) to form the anion, followed by addition of an acrylate unit (e.g., t-butylacrylate) at a temperature g from -78°C to room temperature to give the homologated ester 41.
Saponification of the t-butyl ester of compound 41 by treatment with a suitable acid such as TFA or HCl at an appropriate temperature (e.g, 0°C to reflux) provides compound 42. A Curtius rearrangement of compound 42 using, for example, DPPA in the presence of mild base such as NEt3 at an appropriate temperature (e.g., 0°C to reflux), followed by treatment of the reactive intermediate with an appropriate alcohol (e.g., tBuOH), optionally in the presence of a Lewis acid (e.g., SnCl2) at elevated temperatures (e.g., 40-200°C) provides compound 43. The choice of alcohol determines the amine protecting group of compound 43 (e.g., tBuOH provides the Boc-amine). ection of the ester of compound 43 under standard conditions (e.g., LiOH for a methyl ester, hydrogenation for a benzyl ester, etc.) gives the acid 44.
In one alternative to Scheme E, Pg may be substituted with R7 in compounds 43 and CHO NC CO2R''' (R9)t (R9)t Base CO2R''' 45 46 Reduction R6 1. Substitution R6 2. Substitution 1. tution N NH2 2. Protection NPg R7 3. fication (R9)t (R9)t 3. Saponification (R9)t CO2H CO2R''' CO2H 48 47 49 1. Protection 2. Saponification NHPg (R9)t CO2H 50 Scheme F Scheme F shows a method of ing optionally substituted β-phenylalanine amino acids 48, 49 and 50 of Formula 3A wherein R6 is H, R7 is an amine protecting group, t is 0 to 4, and R9 is as defined herein. An appropriately substituted aldehyde 45 can be treated with a cetate of the formula CN-CH2CO2R''' wherein R''' is alkyl (e.g., ethyl 2- cyanoacetate) in the presence of a suitable base such as piperidine at an riate ature (e.g., room temperature to reflux) to give the unsaturated ester 46. Reduction of the olefin and the nitrile groups of compound 46 to provide compound 47 may be accomplished in a number of ways. For example, the olefin may be reduced with any agent known to effect ductions, such as NaBH4. The nitrile may be reduced using agents such as LiAlH4 or NaBH4 in the presence of a Lewis acid such as BF3.OEt2 or TFA. A number of alternative reducing agents may be used, such as those listed in ‘Reductions in Organic Chemistry’ by Hudlicky, ACS monograph, 2nd n, Chapter 18. If desired, the primary amine 47 can be kylated or bisalkylated at this stage using standard conditions (e.g., ive amination using an appropriate aldehyde, Lewis acid and reducing agent) to provide intermediates (not shown) en route to compounds 48 and 49. To prepare primary and secondary amines, protection may be accomplished using any number of protecting groups (e.g., ‘Protective Groups in Organic Synthesis’ by Greene and Wuts, Wiley-Interscience, third edition, Chapter 7), for example as a Boc-group using Boc anhydride at 0 °C to room ature. Cleavage of the ester group to form the amino acid 48, 49 or 50 may be accomplished using an aqueous bases such as LiOH or KOH, or any of the alternative reagents listed in the aforementioned ‘Protecting Groups’ text (e.g., hydrogenation for a benzyl .
In one alternative to Scheme F, Pg may be substituted with R7 in compounds 49 or 50.
Reduction (R9)t 1. Activation (R9)t CO2H OH 2. Base 51 52 R'O2C NHPg NHPg ection (R9)t NHPg (R9)t CO2R' CO2H 53 54 Scheme G Scheme G shows a method of preparing optionally substituted α-phenylalanine amino acids 54 of Formula 4A, wherein R6 is H, R7 is an amine protecting group, t is 0 to 4, and R9 is as defined herein. An appropriately substituted acid 51 may be reduced to the benzyl alcohol 52 using for example LiAlH4 at a ature ranging from room temperature to reflux. The l group of compound 52 can be activated as a leaving group (e.g., halide, mesylate, etc.) using, for example, PBr3, MsCl/NEt3, etc. Displacement of this leaving group using a ted glycine derivative such as ethyl 2-(diphenylmethyleneamino)acetate in the ce of strong base such as LDA, nBuLi provides the amino ester intermediate 53 wherein R1 is alkyl and Pg is a protecting group. riate protecting groups are listed in ‘Protective Groups in Organic Synthesis’ by Greene and Wuts, Wiley-Interscience). The amine protecting group may be changed at this stage, for example to uce a Boc-group. uent deprotection of the ester 53 (e.g., using 3N HCl, LiOH, hydrogenation for a benzyl ester, etc.) at an appropriate temperature (e.g., 0°C to reflux) provides the desired N- protected amino acid 54.
In one alternative to Scheme G, Pg may be substituted with R7 in compound 54 after the deprotection of nd 53.
Bn 1. Deprotection CO2R' 2. Reprotection (R9)t BnHN CO2R' 3. Cleavage of ester CO2R' formaldehyde (R9)t CO2H (R9)t Scheme H Scheme H shows a method of preparing optionally substituted γ-phenylglycine amino acids 56 of Formula 2A wherein R6 and R8 er with the atoms to which they are attached form a spirocyclic heterocyclic ring, R7 is an amine protecting group, t is 0 to 4, and R9 is as defined herein. According to Scheme H, the unsaturated ester 24 can be treated with a suitably protected glycine derivative (e.g., benzylglycine) and formaldehyde under dry conditions (e.g., with addition of molecular sieves) at an appropriate temperature (e.g., room temperature to reflux) to generate compound 55. Cleavage of the benzyl group using standard conditions (e.g., via hydrogenation, 1-chloroethylformate, etc.) followed by addition of an amine protecting group such as a Boc-group and cleavage of the ester under standard conditions (e.g., LiOH for a methyl ester, acid for a l ester, etc., at 0°C to ) provides the N-protected amino acid 56.
In one alternative to Scheme H, Pg may be substituted with R7 in compound 56. [3+2] NR'' CO2H Esterification CO2R' cycloaddition (R9)t (R9)t (R9)t CO2R' 57 58 Deprotection NBoc 1. Amine protection (R9)t (R9)t CO2H 2. Ester cleavage CO2R' 1. Amine onalization 2. Ester deprotection (R9)t CO2H Scheme I Scheme I shows a method of preparing optionally substituted β-phenylalanine amino acids 61 and 62 of Formula 3A wherein R6 and Rb together with the atoms to which they are attached form a heterocyclic ring, and R7 and R9 are as defined herein and t is 0 to 4. The acid 57 is converted to an ester 58 using standard conditions such as ent with an appropriate alcohol (e.g., MeOH) in the ce of either catalytic acid (e.g., concentrated H2SO4 or TMSCl) or a coupling agent (e.g., DCC/DMAP); or alternatively by treatment with an appropriate electrophile (e.g., MeI, EtBr, BnBr) in the presence of a suitable base such as NEt3/DMAP at appropriate temperatures (e.g., -20°C to 100°C). The appropriate choice of ester is determined by the conditions required to reform the acid at the end of the synthesis, such as described in ‘Protective Groups in Organic Synthesis’ by Greene and Wuts, Wiley- Interscience, third edition, Chapter 5. Cyclization of compound 58 to provide compound 59 may be achieved using, for example, N-(methoxymethyl)(phenyl)-N- ((trimethylsilyl)methyl)methanamine in the presence of TFA. This particular set of reagents generates the benzylamine, which can be cleaved to provide compound 60 under standard ions such as such as hydrogenation at -20°C to 50°C or any other standard conditions such as those listed in ‘Protective Groups in Organic Synthesis’ by Greene and Wuts, Wiley- Interscience, third edition, Chapter 7. Protection of the free amine of compound 60 with an alternative protecting group (e.g., Boc) using reagents listed in the aforementioned text, such as Boc-anhydride, followed by cleavage of the ester using standard conditions appropriate for the ester (e.g., aqueous LiOH for methyl esters, hydrogenation for benzyl , acid for tbutyl esters) es the acid compound 61. Alternatively, the free amine can be functionalized further (e.g., using alkylation, reductive amination, or acylation conditions), followed by ester cleavage to te the tertiary amino acid compound 62.
R6 R6 N OMe R6 Boc Boc Hydrolysis N COR* (R9)t COR* (R9)t CO2H (R9)t Optional N deprotection CO2H (R9)t Scheme J Either enantiomer of the b-amino acids may be prepared using a procedure such as that shown in Scheme J. A ylacetate d with an appropriate chiral auxillary (R*) (for example, an Evans’ auxiliary or a Sultam) with the riate stereochemistry to te the d chemistry at the b-position of the amino acid may be treated with an imine or iminium ion synthon (e.g., prepared in situ by the presence of a Lewis acid (e.g., TiCl4) and an appropriately substituted alkoxymethanamine or N- (alkoxymethyl)amide/carbamate at -100°C to 50°C). The asymmetric addition may require the presence of Lewis acids (e.g., TiCl4), amine bases (e.g., Hunig’s base) and lower temperatures (e.g., -100°C to 0°C) to generate the best levels of stereochemical induction. If the de is lower than required, the separate diastereomers may be separated at this stage by (for example) chromatography or llization. Cleavage of the chiral auxillary, using methods known to cleave the chosen auxillary (e.g., LiOH/H2O2 at -50°C to 50°C for the Evans auxillary) then leads to the desired N-protected b-amino acid with the d stereochemistry at the b-position. Additionally, if R6 is also a protecting group (e.g., 2,4- dimethoxybenzyl), it may be removed in the presence of the Boc-group (e.g., hydrogenation or DDQ, etc.) to give the Boc-amino acid, which upon removal of the Boc-group would provide the primary amine, which may be further functionalized by alkylation, acylation or reductive amination (either prior to or after coupling with the pyrimidine-piperazine unit).
In preparing compounds of Formula I, protection of remote functionalities (e.g., y or secondary amines, etc.) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the ation methods. le amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), oxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T.
W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
S OF SEPARATION In any of the synthetic methods for preparing compounds of a I, it may be advantageous to te reaction products from one another and/or from starting materials.
The d products of each step or series of steps is separated and/or purified to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low re liquid chromatography s and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
Another class of separation s involves treatment of a reaction mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, g reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction ts (LIX), or the like.
Selection of appropriate methods of separation depends on the nature of the materials involved. For example, boiling point and molecular weight in distillation and sublimation, presence or e of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply ques most likely to achieve the desired separation.
Diastereomeric mixtures can be ted into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by on with an appropriate lly active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds useful in the present invention may be somers (e.g., substituted biaryls) and are considered as useful in this invention. Enantiomers can also be separated by use of a chiral HPLC column.
A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be ed by resolution of the c mixture using a method such as formation of diastereomers using lly active resolving agents (Eliel, E. and Wilen, S.
"Stereochemistry of Organic Compounds," John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., (1975) 113 (3):283-302). Racemic mixtures of chiral nds useful in the invention can be separated and isolated by any suitable method, including: (1) ion of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric nds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the ntially pure or enriched stereoisomers directly under chiral ions. See: "Drug Stereochemistry, ical Methods and Pharmacology," Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, α-methyl-βphenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by onal crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (E. and Wilen, S.
"Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., 1994, p. 322).
Diastereomeric nds can be formed by ng asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-)menthyl chloroformate in the presence of base, or Mosher ester, α-methoxy-α- (trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem., (1982) 47 :4165), of the racemic mixture, and analyzing the 1H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers. Stable diastereomers of atropisomeric nds can be separated and ed by normal- and reverse-phase chromatography ing methods for tion of atropisomeric naphthyl-isoquinolines (WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral nary phase ("Chiral Liquid Chromatography" (1989) W. J. Lough, Ed., Chapman and Hall, New York; Okamoto, J. of Chromatogr., (1990) 513 :375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
CHEMOTHERAPEUTIC AGENTS Certain chemotherapeutic agents have demonstrated surprising and unexpected properties in ation with a compound of formula I or a pharmaceutically acceptable salt f in inhibiting cellular proliferation in vitro and in vivo. Such chemotherapeutic agents include: 5-FU, a platinum agent, irinotecan, docetaxel, doxorubicin, gemcitabine, SN-38, tabine, lomide, erlotinib, PD-0325901, paclitaxel, bevacizumab, pertuzumab, fen, rapamycin, lapatinib, PLX-4032, 0, abiraterone, and GDC-0973. 5-FU (fluorouracil, 5-fluorouracil, CAS Reg. No. 8) is a thymidylate se inhibitor and has been used for decades in the treatment of cancer, including colorectal and pancreatic cancer (US 2802005; US 2885396; Duschinsky et al (1957) J. Am. chem. Soc. 79:4559; Hansen, R.M. (1991) Cancer Invest. 9:637-642). 5-FU is named as 5-fluoro-1H- pyrimidine-2,4-dione.
Folinic acid (INN) or leucovorin (USAN) ((2S){[4-[(2-aminoformyloxo- ,6,7,8-tetrahydro-1H-pteridinyl)methylamino]benzoyl]amino}pentanedioic acid, CAS Reg. No. 14928), generally administered as calcium or sodium folinate (or leucovorin calcium/sodium), is used in cancer chemotherapy involving the synergistic combination with the chemotherapy agent 5-fluorouracil, and in certain embodiments with oxaliplatin, or optionally with other platins such as cisplatin, as part of the regimen FOLFOX. It has the structure: Oxaliplatin (CAS Reg. No. 631216) is a coordination complex that is used in cancer chemotherapy d States Patent Number 4,169,846). latin has been compared with other platinum compounds (Cisplatin, latin) in advanced cancers (gastric, ovarian). Oxaliplatin is lly administered with fluorouracil and leucovorin in a combination known as FOLFOX for the treatment of colorectal cancer. mFOLFOX6 (modified FOLFOX6) refers to oxaliplatin (e.g., ELOXATIN®), 5-FU (e.g., ADRUCIL®), and orin (e.g., WELLCOVORIN®).
Carboplatin (CAS Reg. No. 415754) is a chemotherapeutic drug used against ovarian carcinoma, lung, head and neck cancers (US 4140707; Calvert et al (1982) Cancer Chemother. Pharmacol. 9:140; d et al (1984) Cancer Res. 44:1693). Carboplatin is named as azanide; cyclobutane-1,1-dicarboxylic acid; platinum.
Cisplatin, cisplatinum, or cis-diamminedichloroplatinum(II) (CAS Reg. No. 15663- 27-1) is a chemotherapeutic drug used to treat various types of cancers, including sarcomas, some carcinomas (e.g., small cell lung , and ovarian cancer), lymphomas, and germ cell tumors. It was the first member of a class of platinum-containing anti-cancer drugs, which now also includes carboplatin and oxaliplatin. Cisplatin has the ure cis- NH3)2.
Irinotecan (CAS Reg. No. 976825) is a topoisomerase 1 inhibitor, which prevents DNA from unwinding. Irinotecan is activated by hydrolysis to SN-38, an inhibitor of topoisomerase I. The inhibition of topoisomerase I by the active metabolite SN-38 eventually leads to inhibition of both DNA replication and transcription. Its main use is in colon cancer, in particular, in combination with other chemotherapy agents. This includes the regimen I, which consists of onal 5-fluorouracil, leucovorin, and irinotecan.
Doxorubicin (CAS Reg. No. 92-8) is an anthracycline antibiotic. Like all anthracyclines, it works by intercalating DNA. Doxorubicin is commonly used in the treatment of a wide range of cancers, including hematological malignancies, many types of carcinoma, and soft tissue sarcomas. Doxorubicin is named as (8S,10S)(4-amino hydroxymethyl-tetrahydro-2H-pyranyloxy)-6,8,11-trihydroxy(2-hydroxyacetyl) methoxy-7,8,9,10-tetrahydrotetracene-5,12-dione.
Docetaxel (CAS Reg. No. 1149775) is a taxane used to treat breast, ovarian, and NSCLC cancers (US 0; US 5438072; US 5698582; US 5714512; US 5750561; Mangatal et al (1989) Tetrahedron 45:4177; Ringel et al (1991) J. Natl. Cancer Inst. 83:288; Bissery et al (1991) Cancer Res. 51:4845; Herbst et al (2003) Cancer Treat. Rev. 29:407-415; Davies et al (2003) Expert. Opin. cother. 4:553-565). xel is named as (2R,3 S)- N-carboxyphenylisoserine, N-tert-butyl ester, 13-ester with 5, 20-epoxy-1, 2, 4, 7, 10, 13- hexahydroxytaxenone 4-acetate 2-benzoate, trihydrate (US 4814470; EP 253738; CAS Reg. No. 1149775).
Gemcitabine (CAS Reg. No. 950584) is a nucleoside analog which blocks DNA replication, is used to treat various carcinomas including pancreatic, breast, NSCLC, and lymphomas (US 4808614; US 5464826; Hertel et al (1988) J. Org. Chem. 53:2406; Hertel et al (1990) Cancer Res. 50:4417; Lund et al (1993) Cancer Treat. Rev. 19:45-55). Gemcitabine is named as 4-amino[3,3-difluorohydroxy (hydroxymethyl) tetrahydrofuranyl]- 1H-pyrimidin- 2-one.
SN-38 (CAS Reg. No. 866393) is the active lite of irinotecan (see above).
It is 200 times more active than irinotecan itself. It has the name 7-ethylhydroxycamptothecin.
Capecitabine (CAS Reg. No. 1543619) is an orally-administered herapeutic agent used in the treatment of metastatic breast and colorectal cancers.
Capecitabine is a prodrug, that is enzymatically converted to 5-fluorouracil in the tumor, where it ts DNA synthesis and slows growth of tumor tissue. The activation of capecitabine follows a pathway with three enzymatic steps and two intermediary metabolites, '-deoxyfluorocytidine (5'-DFCR) and 5'-deoxyfluorouridine (5'-DFUR), to form 5- fluorouracil. Capecitabine has the name pentyl[1-(3,4-dihydroxymethyl-tetrahydrofuran- 2-yl)- rooxo-1H-pyrimidin- 4-yl]aminomethanoate.
Temozolomide (CAS Reg. No. 856221) is an alkylating agent which can be used for the treatment of Grade IV astrocytoma, also known as glioblastoma multiforme as well as Melanoma, a form of skin . Temozolomide has the name yloxo- 2,3,4,6,8- pentazabicyclo [4.3.0] nona-2,7,9-triene- 9-carboxamide.
Erlotinib (CAS Reg. No. 1833216, TARCEVA®, OSI-774, Genentech) is used to treat non-small cell lung cancer ), lung cancer, pancreatic cancer and several other types of cancer by ically targeting the epidermal growth factor receptor (EGFR) tyrosine kinase (US 5747498; US 6900221; Moyer et al (1997) Cancer Res. 57:4838; Pollack et al (1999) J. Pharmcol. Exp. Ther. 291:739; Perez-Soler et al (2004) J. Clin. Oncol. 22:3238; Kim et al (2002) Curr. Opin. Invest. Drugs 3:1385-1395; Blackhall et al (2005) Expert Opin. Pharmacother. 6:995-1002). Erlotinib is named as thynylphenyl)-6,7- bis(methoxymethoxy)quinazolinamine (CAS Reg. No. 1833216) and has the structure: O O N O O PD-0325901 (CAS Reg. No. 3912109, Pfizer) is a second-generation, non-ATP competitive, allosteric MEK inhibitor for the potential oral tablet treatment of cancer (US 6960614; US 6972298; US 2004/147478; US 2005/085550). Phase II clinical trials have been conducted for the potential ent of breast tumors, colon tumors, and melanoma.
PD-0325901 is named as (R)-N-(2,3-dihydroxypropoxy)-3,4-difluoro(2-fluoro iodophenylamino)benzamide, and has the structure: N O HO O H F OH N F I Paclitaxel (CAS Reg. No. 330694, TAXOL®, l-Myers Squibb Oncology, Princeton NJ) is isolated the compound from the bark of the c yew tree, Taxus brevifolia, and used to treat lung, ovarian, breast cancer, and advanced forms of Kaposi's sarcoma (Wani et al (1971) J. Am. Chem. Soc. 93:2325; Mekhail et al (2002) Expert. Opin.
Pharmacother. 3:755-766). Paclitaxel is named as β-(benzoylamino)-α-hydroxy-,6,12b-bis (acetyloxy)(benzoyloxy)-2a,3,4,4a,5,6,9,10,11,12,12a,12b-dodecahydro-4,11-dihydroxy- 3,13-tetramethyloxo-7,11-methano-1H-cyclodeca(3,4)benz(1,2-b) oxet ylester,(2aR-(2a-α,4-β,4a-β,6-β,9-α β-S*),11-α,12-α,12a-α,2b-α))-benzenepropanoic acid, and has the structure: Bevacizumab (CAS Reg. No. 2169743, AVASTIN, Genentech, Inc.) is a recombinant zed monoclonal antibody against VEGF, ar elial growth factor (US 6054297; Presta et al (1997) Cancer Res. 57:4593-4599). It is used in the treatment of cancer, where it inhibits tumor growth by blocking the ion of new blood vessels. Bevacizumab was the first clinically available angiogenesis inhibitor in the United States, approved by the FDA in 2004 for use in combination with standard chemotherapy in the treatment of metastatic colon cancer and most forms of metastatic non-small cell lung cancer. Several late-stage al studies are ay to determine its safety and effectiveness for ts with: adjuvant / non-metastatic colon cancer, atic breast cancer, metastatic renal cell carcinoma, metastatic glioblastoma multiforme, metastatic ovarian cancer, metastatic hormone-refractory prostate cancer, and atic or unresectable locally advanced pancreatic cancer (Ferrara et al (2004) Nat. Rev. Drug Disc. 3:391-400).
Bevacizumab has a molecular mass of about 149,000 daltons and is glycosylated.
Bevacizumab and other humanized anti-VEGF antibodies are further described in US 6884879. Additional anti-VEGF antibodies include the G6 or B20 series antibodies, e.g., G6- 31, B20-4.1, (; ; US 7060269; US 6582959; US 0; US 6054297; WO 98/45332; WO 46; WO 94/10202; EP 0666868B1; US 2006/009360; US 2005/0186208; US 2003/0206899; US 2003/0190317; US 2003/0203409; 20050112126; Popkov et al (2004) Journal of Immunological Methods 288:149-164. A “B20 series antibody” is an anti-VEGF antibody that is derived from a sequence of the B20 antibody or a B20-derived antibody according to any one of Figures 27-29 of WO 2005/012359, the entire disclosure of which is expressly incorporated herein by reference. In one embodiment, the B20 series antibody binds to a functional epitope on human VEGF comprising residues F17, M18, D19, Y21, Y25, Q89, I91, K101, E103, and C104. Other anti-VEGF antibodies e those that bind to a functional epitope on human VEGF comprising residues F17, M18, D19, Y21, Y25, Q89, I91, K101, E103, and C104 or, alternatively, comprising residues F17, Y21, Q22, Y25, D63, I83 and Q89.
Trastuzumab PTIN, huMAb4D5-8, rhuMAb HER2, Genentech) is a recombinant DNA-derived humanized, IgG1 kappa, monoclonal antibody n of the murine HER2 antibody which selectively binds with high affinity in a cell-based assay (Kd = nM) to the extracellular domain of the human mal growth factor receptor2 protein, HER2 ) (US 5821337; US 6054297; US 6407213; US 6639055; Coussens L, et al (1985) Science 230:1132-9; Slamon DJ, et al (1989) Science 244:707-12). Trastuzumab contains human framework regions with the complementarity-determining regions of a murine antibody (4D5) that binds to HER2. Trastuzumab binds to the HER2 antigen and thus inhibits the growth of cancerous cells. Trastuzumab has been shown, in both in vitro assays and in animals, to inhibit the proliferation of human tumor cells that overexpress HER2 (Hudziak RM, et al (1989) Mol Cell Biol 9:1165-72; Lewis GD, et al (1993) Cancer Immunol ther; 37:255-63; Baselga J, et al (1998) Cancer Res. 58:2825-2831). Trastuzumab is a mediator of antibody-dependent cellular cytotoxicity, ADCC ing TE, et al (1996) [abstract]. Proc. Annual Meeting Am Assoc Cancer Res; 37:471; Pegram MD, et al (1997) [abstract]. Proc Am Assoc Cancer Res; 38:602; Sliwkowski et al (1999) Seminars in Oncology 26(4), Suppl 12:60-70; Yarden Y. and Sliwkowski, M. (2001) Nature Reviews: Molecular Cell Biology, Macmillan Magazines, Ltd., Vol. 2:127-137). HERCEPTIN was approved in 1998 for the treatment of patients with ErbB2-overexpressing metastatic breast cancers (Baselga et al, (1996) J. Clin. Oncol. 14:737-744). The FDA approved HERCEPTIN  in 2006 as part of a treatment regimen containing doxorubicin, cyclophosphamide and paclitaxel for the adjuvant treatment of patients with HER2-positive, node-positive breast cancer. There is a significant clinical need for developing further HER2- directed cancer therapies for those patients with verexpressing tumors or other diseases associated with HER2 expression that do not respond, or respond poorly, to TIN  treatment.
Pertuzumab ARG™, rhuMab 2C4, Genentech) is a al stage, humanized antibody and the first in a new class of agents known as HER dimerization inhibitors (HDIs) which block the ability of the HER2 receptor to orate with other HER receptor family members, i.e. HER1/EGFR, HER3, and HER4 (US 6949245; Agus et al (2002) Cancer Cell 2:127–37; Jackson et al (2004) Cancer Res 1–9; Takai et al (2005) Cancer 104:2701– 8). In cancer cells, interfering with HER2's ability to collaborate with other HER family ors blocks cell signaling and may ultimately lead to cancer cell growth inhibition and death of the cancer cell. HDIs, because of their unique mode of action, have the potential to work in a wide variety of tumors, including those that do not press HER2 (Mullen et al (2007) Molecular Cancer eutics 00).
Temozolomide, (CAS Reg. No. 856221, TEMODAR®, TEMODAL®, Schering Plough) is a oral chemotherapy drug ed by the FDA for the treatment of anaplastic astrocytoma, and has been studied for other brain tumor types such as glioblastoma multiforme (US 5260291; Stevens et al (1984) J. Med. Chem. 27:196; Newlands et al (1997) Cancer Treat. Rev. 23:35-61; Danson et al (2001) Expert Rev. Anticancer Ther. 1:13-19).
Temozolomide is named as (4-methyloxo- 2,3,4,6,8-pentazabicyclo [4.3.0] nona-2,7,9- - oxamide or 3,4-dihydromethyloxoimidazo [5,1-d]-as-tetrazine carboxamide (US 5260291, CAS No. 856221), and has the structure: N N Tamoxifen (CAS Reg. No. 105401, NOLVADEX®, ISTUBAL®, VALODEX®) is an orally active, selective estrogen receptor modulator (SERM) which is used in the treatment of breast cancer and is currently the s largest selling drug for this indication.
Tamoxifen (Nolvadex®) was first approved by the FDA (ICI Pharmaceuticals, now AstraZeneca) in 1977 for treatment of metastatic breast cancer (Jordan VC (2006) Br J Pharmacol 147 (Suppl 1): S269-76). Tamoxifen is currently used for the treatment of both early and advanced estrogen receptor (ER) positive breast cancer in pre- and post-menopausal women (Jordan VC (1993) Br J Pharmacol 110 (2): 507-17). It is also approved by the FDA for the prevention of breast cancer in women at high risk of developing the disease and for the reduction of contralateral (in the opposite breast) breast cancer. Tamoxifen is named as [4-(1,2-diphenylbutenyl)phenoxy]-N,N-dimethyl-ethanamine, (CAS Reg. No. 105401) and has the structure: Rapamycin (CAS Reg. No. 531239, sirolimus, RAPAMUNE®) is an immunosuppressant drug used to prevent rejection in organ transplantation, and is ally useful in kidney transplants. Rapamycin is a macrolide antibiotic ("-mycin" ) first discovered as a product of the bacterium Streptomyces copicus in a soil sample from an island called Rapa Nui, better known as Easter Island (Pritchard DI (2005). Drug Discovery Today (10): 688–691). Rapamycin inhibits the response to interleukin-2 (IL-2) and thereby blocks activation of T- and B-cells. The mode of action of rapamycin is to bind the cytosolic protein FK-binding protein 12 (FKBP12). The cin-FKBP12 complex inhibits the ian target of rapamycin (mTOR) pathway h directly g the mTOR Complex1 (mTORC1). mTOR is also called FRAP (FKBP-rapamycin associated protein) or RAFT (rapamycin and FKBP target). Rapamycin is named as (3 S,6 R,7 E,9 R,10 R,12 R,14 S,15 E,17 E,19 E,21 S,23 S,26 R,27 R,34a S)- 9,10,12,13,14,21,22,23,24,25,26,27,32,33,34,34a-hexadecahydro-9,27-dihydroxy[(1 R) [(1 S,3 R,4 R)hydroxymethoxycyclohexyl]methylethyl]-10,21-dimethoxy- 6,8,12,14,20,26-hexamethyl-23,27-epoxy-3H-pyrido[2,1-c][1,4]-oxaazacyclohentriacontine- 1,5,11,28,29(4 H,6 H,31 H)-pentone (CAS Reg. No. 531239), and has the structure: Lapatinib (CAS Reg. No. 3880828, TYKERB®, GW572016, Glaxo line) has been approved for use in combination with capecitabine (XELODA®, Roche) for the treatment of patients with advanced or metastatic breast cancer whose tumors over-express HER2 (ErbB2) and who have ed prior therapy including an anthracycline, a taxane and trastuzumab. Lapatinib is an ATP-competitive epidermal growth factor (EGFR) and HER2/neu (ErbB-2) dual tyrosine kinase inhibitor (US 6727256; US 6713485; US 7109333; US 6933299; US 7; US 7157466; US 6) which inhibits receptor autophosphorylation and activation by binding to the ATP-binding pocket of the EGFR/HER2 protein kinase domain. Lapatinib is named as hloro(3- benzyloxy)phenyl)(5-((2-(methylsulfonyl)ethylamino)methyl)furanyl)quinazolin- 4-amine, and has the structure: O HN Cl S N O H N N .
Vemurafenib 4, PLX-4032, CAS Reg. No. 10298725) has been shown to cause programmed cell death in various cancer call lines, for example melanoma cell lines.
Vemurafenib interrupts the B-Raf/MEK step on the B-Raf/MEK/ERK pathway − if the B-Raf has the common V600E mutation. Vemurafenib works in patients, for example in melanoma patients as approved by the FDA, whose cancer has a V600E BRAF on (that is, at amino acid position number 600 on the B-RAF protein, the normal valine is replaced by glutamic acid). About 60% of melanomas have the V600E BRAF mutation. The V600E mutation is present in a variety of other cancers, including lymphoma, colon cancer, melanoma, thyroid cancer and lung cancer. Vemurafenib has the following structure: F O Cl O N N H .
ZELBORAF® (vemurafenib) (Genentech, Inc.) is a drug product approved in the U.S. and indicated for treatment of patients with unresectable or metastatic melanoma with BRAF V600E on as detected by an FDA-approved test. AF® (vemurafenib) is not recommended for use in melanoma patients who lack the BRAF V600E mutation (wild-type BRAF ma).
MDV3100 (CAS Reg. No. 9150871) is an androgen receptor antagonist drug developed for the treatment of hormone-refractory te cancer. Up to an 89% decrease in prostate specific antigen serum levels has been reported after a month of taking the ne.
As opposed to bicalutamide, MDV3100 does not promote translocation of AR to the nucleus and in addition ts g of AR to DNA and AR to coactivator proteins. MDV 3100 was found clinically active for metastatic castration-resistant prostate cancer patients in ongoing phase I and II trials. MDV3100 has the name 4-(3-(4-cyano uoromethyl)phenyl)-5,5-dimethyloxothioxoimidazolidinyl)fluoro-N- methylbenzamide.
Abiraterone (CAS Reg. No. 1542293; see United States Patents 5,604,213 and ,618,807) is a drug currently under investigation for use in castration-resistant prostate cancer. It blocks the formation of testosterone by inhibiting 1 (CYP450c17), an enzyme also known as 17α-hydroxylase/17,20 lyase. This enzyme is involved in the formation of DHEA and androstenedione, which may ultimately be metabolized into testosterone. erone has the name (3S,8R,9S,10R,13S,14S)-10,13-dimethyl(pyridin- 3-yl)-2,3,4,7,8,9,10,11,12,13,14,15-dodecahydro-1H-cyclopenta[a]phenanthrenol. It may also be administered as the acetate prodrug (3S,8R,9S,10R,13S,14S)-10,13-dimethyl (pyridinyl)-2,3,4,7,8,9,10,11,12,13,14,15-dodecahydro-1H-cyclopenta[a]phenanthrenyl acetate.
ZYTIGA® (abiraterone acetate) (JOHNSON & JOHNSON Corp) is a drug product approved in the U.S. and indicated for use in combination with prednisone for the treatment of patients with metastatic castration-resistant prostate cancer who have received prior chemotherapy containing docetaxel.
GDC-0973 is a selective inhibitor of MEK, also known as n activated protein kinase kinase (MAPKK), which is a key component of the RAS/RAF/MEK/ERK pathway that is frequently activated in human tumors. opriate activation of the MEK/ERK pathway promotes cell growth in the absence of ous growth factors. A Phase I clinical trial evaluating 73 for solid tumors is ongoing. GDC-0973 can be ed as described in International Patent Application ation Number WO2007044515(A1).
GDC-0973 has the name: (S)-(3,4-difluoro(2-fluoroiodophenylamino)phenyl)(3- hydroxy(piperidinyl)azetidinyl)methanone, and the ing structure: O F F .
PHARMACEUTICAL COMPOSITIONS Pharmaceutical compositions or formulations described include combinations of Formula I compounds, a chemotherapeutic agent, and one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient.
One example includes a first formulation for oral ry of a nd of formula I, or a salt thereof, and one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient, and a second formulation for oral delivery of vemerafenib, or a salt thereof, and one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient. In one example, the first formulation comprises 68 or a salt thereof.
The Formula I compounds, and chemotherapeutic agents useful in the present invention may exist in unsolvated as well as solvated forms with ceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention e the use of both solvated and unsolvated forms.
The Formula I compounds, and chemotherapeutic agents useful in the present invention may also exist in different tautomeric forms, and all such forms are embraced for use within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier.
For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and enamine isomerizations. Valence tautomers e interconversions by reorganization of some of the bonding electrons.
Pharmaceutical compositions encompass both the bulk ition and individual dosage units comprised of more than one (e.g., two) pharmaceutically active agents including a a I compound and a chemotherapeutic agent selected from the lists of the additional agents described , along with any pharmaceutically inactive excipients, diluents, carriers, or glidants. The bulk composition and each dual dosage unit can contain fixed amounts of the aid pharmaceutically active agents. The bulk composition is material that has not yet been formed into dual dosage units. An illustrative dosage unit is an oral dosage unit such as s, pills, capsules, and the like. Similarly, the herein-described method of ng a patient by administering a pharmaceutical composition described herein is also intended to encompass the administration of the bulk composition and individual dosage units. ceutical compositions also embrace isotopically-labeled compounds useful in the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds useful in the invention, and their uses. Exemplary isotopes that can be incorporated into nds include isotopes of hydrogen, carbon, nitrogen, , orus, sulfur, fluorine, chlorine and iodine, such as 2H, 3H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 Cl, 123 I and 125 I. Certain isotopically-labeled compounds useful in the present invention (e.g., those labeled with 3H and 14 C) are useful in compound and/or substrate tissue distribution assays.
Tritiated (3H) and carbon-14 (14 C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as ium (2H) may afford certain therapeutic advantages resulting from greater metabolic ity (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15 O, 13 N, 11 C and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
Isotopically labeled compounds useful in the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
Formula I nds and herapeutic agents are formulated in accordance with standard pharmaceutical practice for use in a therapeutic combination for therapeutic treatment (including prophylactic ent) of hyperproliferative disorders in s including humans. bed is a pharmaceutical composition comprising a Formula I compound in association with one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient.
Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or ble rs, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like. The particular carrier, diluent or ent used will depend upon the means and purpose for which the compound useful in the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water. le s solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and es thereof. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound useful in the t invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
The formulations may be prepared using conventional dissolution and mixing procedures. For example, the bulk drug substance (i.e., compound useful in the present invention or stabilized form of the compound (e.g., complex with a extrin tive or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more of the excipients described above. The nd useful in the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
The pharmaceutical ition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. Generally, an article for distribution includes a container having deposited n the pharmaceutical formulation in an appropriate form. Suitable ners are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has ted thereon a label that describes the contents of the container. The label may also include appropriate warnings.
Pharmaceutical formulations of the compounds useful in the t ion may be prepared for various routes and types of administration. For example, a a I compound having the desired degree of purity may optionally be mixed with pharmaceutically able diluents, carriers, excipients or izers gton's ceutical Sciences (1995) 18th edition, Mack Publ. Co., , PA), in the form of a lyophilized formulation, milled powder, or an aqueous solution. Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed. The pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
The pharmaceutical formulation is preferably sterile. In particular, formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
The pharmaceutical formulation rily can be stored as a solid composition, a lized formulation or as an aqueous solution.
The pharmaceutical formulations will be dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to l practitioners. The "therapeutically effective amount" of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the ation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding.
As a general proposition, the initial pharmaceutically effective amount of the Formula I compound administered orally or parenterally per dose will be in the range of about 0.01- 1000 mg/kg, namely about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of nd used being 0.3 to 15 mg/kg/day. The dose of the Formula I compound and the dose of the chemotherapeutic agent to be administered may range for each from about 1 mg to about 1000 mg per unit dosage form, or from about 10 mg to about 100 mg per unit dosage form. The doses of Formula I compound and the chemotherapeutic agent may administered in a ratio of about 1:50 to about 50:1 by , or in a ratio of about 1:10 to about 10:1 by weight.
Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as ate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium de; thonium chloride; benzalkonium chloride, benzethonium chloride; , butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; ol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides and other carbohydrates ing glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic tants such as TWEEN, PLURONICS or hylene glycol (PEG). The active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by acial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery s (for example, liposomes, albumin microspheres, microemulsions, nano-particles and psules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 18th edition, (1995) Mack Publ. Co., Easton, PA.
Sustained-release preparations of Formula I compounds may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a nd of Formula I, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release es include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (US 3773919), copolymers of L-glutamic acid and gamma-ethyl-L- glutamate, non-degradable ethylene-vinyl acetate, able lactic acid-glycolic acid copolymers such as the LUPRON DEPOT (injectable microspheres composed of lactic acid-glycolic acid copolymer and lide acetate) and poly-D (-) oxybutyric acid.
The pharmaceutical formulations e those suitable for the administration routes detailed herein. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations lly are found in Remington's Pharmaceutical Sciences 18th Ed. (1995) Mack Publishing Co., Easton, PA. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In l the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Formulations of a compound of Formula I and/or chemotherapeutic agent suitable for oral administration may be prepared as discrete units such as pills, hard or soft e.g., gelatin capsules, cachets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, syrups or elixirs each containing a predetermined amount of a compound of Formula I and/or a chemotherapeutic agent. The amount of compound of Formula I and the amount of herapeutic agent may be formulated in a pill, capsule, solution or suspension as a combined formulation. Alternatively, the a I compound and the chemotherapeutic agent may be formulated separately in a pill, e, solution or suspension for administration by alternation.
Formulations may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a lowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, e active or dispersing agent. Molded tablets may be made by molding in a le machine a e of the powdered active ingredient moistened with an inert liquid diluent.
The tablets may optionally be coated or scored and ally are formulated so as to provide slow or controlled release of the active ient therefrom.
Tablet excipients of a pharmaceutical formulation may include: Filler (or diluent) to increase the bulk volume of the powdered drug making up the tablet; Disintegrants to age the tablet to break down into small fragments, ideally individual drug particles, when it is ingested and promote the rapid dissolution and absorption of drug; Binder to ensure that granules and tablets can be formed with the required mechanical strength and hold a tablet together after it has been compressed, preventing it from breaking down into its ent powders during packaging, shipping and routine handling; Glidant to improve the ility of the powder making up the tablet during tion; Lubricant to ensure that the ing powder does not adhere to the equipment used to press the tablet during manufacture. They improve the flow of the powder mixes through the presses and minimize friction and breakage as the finished tablets are d from the ent; Antiadherent with function similar to that of the glidant, reducing adhesion between the powder making up the tablet and the machine that is used to punch out the shape of the tablet during manufacture; Flavor incorporated into tablets to give them a more pleasant taste or to mask an unpleasant one, and Colorant to aid identification and patient ance.
Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for cture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and tion in the gastrointestinal tract and thereby provide a sustained action over a longer . For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
For treatment of the eye or other external tissues, e.g., mouth and skin, the ations are preferably applied as a topical nt or cream ning the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w. When formulated in an nt, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. atively, the active ingredients may be formulated in a cream with an oil-in-water cream base.
If desired, the aqueous phase of the cream base may include a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane ol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas.
Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs.
The oily phase of the emulsions useful in this invention may be constituted from known ingredients in a known manner, including a mixture of at least one fier with a fat or an oil, or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. Together, the emulsifier(s) with or without stabilizer(s) make up an emulsifying wax, and the wax together with the oil and fat comprise an emulsifying ointment base which forms the oily sed phase of cream formulations. Emulsifiers and emulsion stabilizers suitable for use in the formulation e Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl monostearate and sodium lauryl sulfate.
Aqueous suspensions of the pharmaceutical formulations contain the active materials in admixture with excipients suitable for the cture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, cellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., yethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a sation product of ethylene oxide with a l ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more ng agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
Pharmaceutical compositions may be in the form of a e injectable ation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may be a solution or a suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared from a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's on and isotonic sodium de solution. In addition, sterile fixed oils may conventionally be employed as a t or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
The amount of active ingredient that may be ed with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release ation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an s on intended for intravenous infusion may contain from about 3 to 500 µg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations suitable for parenteral administration include aqueous and non-aqueous e ion solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
Formulations suitable for topical administration to the eye also include eye drops wherein the active ient is dissolved or suspended in a suitable carrier, especially an aqueous t for the active ingredient. The active ingredient is preferably present in such formulations in a concentration of about 0.5 to 20% w/w, for example about 0.5 to 10% w/w, for example about 1.5% w/w.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and in, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range n 0.1 and 500 microns in increments s such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid tion through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations e aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder stration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or laxis disorders as described below.
Formulations suitable for l administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such rs as are known in the art to be appropriate.
The formulations may be ed in ose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the e liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are ed from sterile powders, granules and tablets of the kind previously bed. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
Further bed are veterinary itions comprising at least one active ingredient as above defined together with a veterinary carrier therefore. nary carriers are materials useful for the purpose of stering the composition and may be solid, liquid or gaseous als which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered erally, orally or by any other desired route.
COMBINATION Y The compound of formula I or a pharmaceutically acceptable salt thereof may be employed in combination with other chemotherapeutic agents or a pharmaceutically acceptable salt thereof for the treatment of a hyperproliferative disease or disorder, including , cancers, and neoplastic tissue, along with pre-malignant and non-neoplastic or nonmalignant hyperproliferative disorders. In certain embodiments, a compound of Formula I or a pharmaceutically acceptable salt thereof is combined in a dosing regimen as combination therapy, with a second compound or a pharmaceutically acceptable salt thereof that has anti- hyperproliferative properties or that is useful for treating the hyperproliferative disorder. The second compound of the dosing regimen preferably has complementary activities to the compound of formula I or a pharmaceutically acceptable salt thereof, and such that they do not adversely affect each other. Such compounds may be administered in amounts that are effective for the purpose intended. In one embodiment, the therapeutic combination is administered by a dosing regimen wherein the therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof is administered in a range from twice daily to once every three weeks (q3wk), and the therapeutically effective amount of the chemotherapeutic agent is administered in a range from twice daily to once every three weeks.
In one example, in response to the stration of herapeutic agents, for example docetaxel, cancer cells upregulate pathways, for e the PI3K/AKT y, in an attempt to circumvent the chemotherapy and become resistant to the chemotherapy. In another example, certain cancers are associated with mutations in PTEN , PI3K or AKT that render the cancers inherently resistant to chemotherapy treatments. By dosing compounds of formual I in combination with the chemotherapeutic agents, compounds of formula I inhibit the pathways that are upregulated in response to the chemotherapeutic agents, or have mutations in the PTEN status, PI3K or AKT pathways. In one embodiment, the combinations herein prevent cancer cells from becoming ant to certain chemotherapeutic therapies. In r embodiment, the combinations herein treat patients that have received chemotherapeutic agents but have become resistant to the treatment or have failed the treatment.
In another example, in response to treatment with Folfox (or one or more of 5-FU, latin or tin, and folinic acid), certain cancers, for example gastric and colon cancers, induce an increase in pAKT, which can act as a mechanism of resistance for the colon cancer in response to the treatment. In another example, certain cancers, for example gastric and colon cancers, are associated with PTEN, PI3K or AKT ons, which can act as a mechanism of resistance for the cancer in response to the treatment.
In one embodiment, GDC-0068 or a salt thereof is administered in ations with Folfox (or one or more of 5-FU, oxaliplatin or cisplatin, and folinic acid) to prevent cancer cells from becoming resistant to the treatment. In another embodiment, GDC-0068 or a salt thereof is administered in combinations with Folfox (or one or more of 5-FU, oxaliplatin or cisplatin, and folinic acid) to treat ts with cancer that have received one or more of the chemotherapeutic agents but have become resistant to the treatment or have failed the treatment. In one specific example, the cancer is gastric cancer. In another example, the cancer is colon cancer.
In another example, certain cancers, for example breast, lung (e.g., non-small lung), prostate (e.g., CRCP), gastric and head/neck cancer are associated with mutations in PTEN status, PI3K or AKT, which can act as a mechanism of resistance for the cancer in se to treatment with taxanes, such as docetaxel or paclitaxel. In one embodiment, GDC-0068 or a salt thereof is administered in combinations with a taxane (e.g., docetaxel) to prevent cancer cells from ng resistant to taxane treatment. In another embodiment, GDC-0068 or a salt thereof is administered in ations with a taxane (e.g., docetaxel) to treat patients with cancer that have received the taxane but have become resistant to the treatment or have failed the ent, in one e due to a PTEN status, PI3K or AKT mutation. In one specific example, the cancer is prostate cancer or hormone refractive prostate cancer. In another example, the cancer is tion resistant prostate cancer and the combination further ses hormone y, for example prednisone. In another example, the combination is ive at does of the taxane that are low enough to prevent harmful side effects from occurring, such as toxicity, neutropenia, hypersensitivity reactions and fluid retention, where such doses of the taxane alone would not be effective.
In another example, in response to treatment with taxanes, for example docetaxel or paclitaxel, certain s, for example prostate cancer (e.g., CRCP), induce an increase in pAKT, which can act as a mechanism of resistance for the prostate cancer in response to the treatment. In one embodiment, GDC-0068 or a salt thereof is administered in combinations with a taxane (e.g., docetaxel) to prevent cancer cells from becoming resistant to taxane treatment. In another embodiment, GDC-0068 or a salt thereof is administered in combinations with a taxane (e.g., docetaxel) to treat patients with cancer that have received the taxane but have become ant to the treatment or have failed the treatment. In one specific example, the cancer is prostate . In r example, the cancer is castration resistant prostate cancer and the combination further comprises hormone therapy, for example prednisone.
The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. The combined administration includes coadministration, using separate ation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological ties.
The compound of formula I or the pharmaceutically acceptable salt thereof can be administered for a time period of about 1 to about 10 days after administration of the one or more agents begins. Also described is, the compound of formula I or the pharmaceutically acceptable salt thereof being administered for a time period of about 1 to 10 days before administration of the combination begins. Also described is, stration of the compound of formula I or the pharmaceutically acceptable salt thereof and administration of the chemotherapeutic agent beginning on the same day.
Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments, such as to increase the therapeutic index or mitigate ty or other side-effects or consequences.
In a particular embodiment of anti-cancer therapy, a compound of formula I, or ceutically acceptable salt thereof, may be combined with a chemotherapeutic agent, as well as ed with surgical therapy and radiotherapy. The amounts of the compound of formula I or a pharmaceutically acceptable salt thereof and the other ceutically active chemotherapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
ADMINISTRATION OF PHARMACEUTICAL COMPOSITIONS The compounds may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, tion, intradermal, intrathecal, epidural, and infusion ques), transdermal, rectal, nasal, l (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal. Topical administration can also involve the use of transdermal administration such as transdermal patches or iontophoresis devices.
Formulation of drugs is discussed in Remington's ceutical Sciences, 18th Ed., (1995) Mack Publishing Co., , PA. Other examples of drug formulations can be found in Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel , Vol 3, 2nd Ed., New York, NY. For local immunosuppressive treatment, the compounds may be administered by intralesional administration, including perfusing or otherwise ting the graft with the inhibitor before transplantation. It will be appreciated that the preferred route may vary with for e the condition of the recipient. Where the compound is stered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier, glidant, or excipient. Where the compound is administered parenterally, it may be formulated with a pharmaceutically acceptable parenteral vehicle or diluent, and in a unit dosage injectable form, as detailed below.
A dose to treat human patients may range from about 20 mg to about 1600 mg per day of the compound of formula I or a pharmaceutically acceptable salt thereof. A typical dose may be about 50 mg to about 800 mg of the compound. A dose may be administered once a day (QD), twice per day (BID), or more frequently, depending on the pharmacokinetic (PK) and codynamic (PD) properties, including tion, distribution, metabolism, and excretion of the particular compound. In on, toxicity factors may influence the dosage and administration dosing regimen. When administered orally, the pill, capsule, or tablet may be ingested twice daily, daily or less frequently such as weekly or once every two or three weeks for a specified period of time. The regimen may be repeated for a number of cycles of therapy.
METHODS OF TREATMENT Therapeutic combinations of: (1) a compound of formula I or a pharmaceutically acceptable salt f, and (2) a chemotherapeutic agent are useful for treating diseases, conditions and/or disorders including, but not limited to, those modulated by AKT kinase in a mammal. s which can be treated according to the methods described include, but are not limited to, mesothelioma, endometrial, breast, lung, n, prostate (including castration resistant ce cancer “CRPC”), pancreatic, melanoma, gastric, colon, glioma, head and neck ARTICLES OF MANUFACTURE Also described is, an article of manufacture, or "kit", containing a compound of formula I or pharmaceutically acceptable salt f useful for the treatment of the diseases and disorders described above. In one embodiment, the kit comprises a container and a compound of formula I or pharmaceutically acceptable salt thereof.
The kit may further comprise a label or package insert, on or associated with the container. The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such eutic products. Suitable containers include, for example, bottles, vials, es, blister pack, etc. The container may be formed from a variety of materials such as glass or plastic.
The ner may hold a compound of formula I or pharmaceutically acceptable salt thereof, or a ation f which is effective for ng the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic ion ). At least one active agent in the composition is a compound of formula I or a pharmaceutically acceptable salt thereof. The label or package insert indicates that the composition is used for treating the condition of choice, such as cancer. In one embodiment, the label or package inserts indicates that the composition comprising a compound of formula I or pharmaceutically acceptable salt thereof can be used to treat a disorder resulting from abnormal cell growth. The label or package insert may also indicate that the composition can be used to treat other disorders.
Alternatively, or additionally, the article of manufacture may further comprise a second container comprising a ceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other als desirable from a commercial and user oint, including other buffers, diluents, filters, needles, and syringes.
The kit may further comprise directions for the administration of the compound of a compound of formula I or pharmaceutically acceptable salt thereof , and, if t, the second pharmaceutical formulation. For example, if the kit comprises a first composition comprising a nd of formula I or pharmaceutically acceptable salt thereof and a second pharmaceutical ation, the kit may further comprise directions for the aneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
In another embodiment, the kits are suitable for the delivery of solid oral forms of a compound of formula I or pharmaceutically acceptable salt thereof, such as tablets or capsules. Such a kit preferably includes a number of unit dosages. Such kits can include a card having the dosages oriented in the order of their intended use. An example of such a kit is a "blister pack". r packs are well known in the packaging industry and are widely used for ing pharmaceutical unit dosage forms. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the ent schedule in which the dosages can be administered.
According to one embodiment, a kit may comprise (a) a first ner with a compound of formula I or pharmaceutically acceptable salt thereof contained therein; and optionally (b) a second container with a second pharmaceutical formulation ned therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity. Alternatively, or additionally, the kit may further se a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, 's solution and dextrose solution. It may r include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and es.
Where the kit comprises a composition of a compound of formula I or pharmaceutically acceptable salt thereof and a second therapeutic agent, i.e. the chemotherapeutic agent, the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be ned within a single, undivided container. Typically, the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are stered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
SPECIFIC EMBODIMENTS In one embodiment described the hyperproliferative disorder is cancer.
In one embodiment described the cancer is associated with PTEN mutation.
In one embodiment described the cancer is associated with AKT mutation, overexpression or amplification.
In one embodiment described the cancer is associated with PI3K mutation.
In one embodiment described the cancer is associated with a HER2 mutation.
In one embodiment described the cancer is selected from, breast, lung, ovarian, prostate (e.g., castration resistant te cancer), melanoma, gastric, colon, renal, head and neck, and .
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and 5-FU are stered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof, 5-FU, and latin are administered to the mammal and the cancer is gastric, ovarian, or colon.
In one embodiment bed the compound of formula I or a pharmaceutically acceptable salt thereof, 5-FU, and oxaliplatin are administered to the mammal and the cancer is gastric, prostate, head or neck.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof, 5-FU, oxaliplatin, and folinic acid are administered to the mammal and the cancer is c, ovarian, or colon.
In one embodiment described the compound of formula I or a pharmaceutically able salt thereof, 5-FU, oxaliplatin, and c acid are administered to the mammal and the cancer is gastric, prostate, head or neck.
In one ment described the compound of formula I, e.g., GDC-0068, or a pharmaceutically acceptable salt thereof, and one or more of 5-FU, oxaliplatin, and folinic acid are administered to the mammal to treat cancer and the cancer is HER2 negative gastric (e.g., first line), colorectal (e.g., first line, ally in ation with a VEGF inhibitor, such as bevacizumab), SCC eck (e.g., first line), colorectal (e.g., second line), or pancreatic (e.g., second line).
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and carboplatin are administered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt f and carboplatin are administered to the mammal and the cancer is breast, lung, or prostate.
In one embodiment described the compound of a I or a pharmaceutically acceptable salt thereof and carboplatin are administered to the mammal and the cancer is breast, lung, te, head or neck.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and irinotecan are administered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and ecan are administered to the mammal and the cancer is colon.
In one embodiment described the compound of formula I or a ceutically able salt thereof and paclitaxel are administered to the mammal to treat endometrial cancer (e.g., second line).
In one embodiment described the compound of a I or a pharmaceutically acceptable salt thereof and docetaxel are administered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and docetaxel are administered to the mammal and the cancer is breast, glioma, lung, melanoma, ovarian, or prostate.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt f and docetaxel are stered to the mammal and the cancer is breast, ovarian, or prostate.
In one embodiment described the compound of formula I, e.g., GDC-0068, or a ceutically acceptable salt thereof and docetaxel are administered to the mammal and the cancer is castration resistant prostate (e.g., first line), HER2 negative breast (e.g., first line), gastric (e.g., second line), gastroesophageal junction (e.g., second line), non-small cell lung (e.g., second line), ovarian (e.g., second line), and us cell carcinoma head and neck (e.g., second line).
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and doxorubicin are administered to the .
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and doxorubicin are administered to the mammal and the cancer is breast, lung, ovarian, , or prostate.
In one embodiment bed the compound of a I or a pharmaceutically acceptable salt thereof and SN-38 are administered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and SN-38 are administered to the mammal and the cancer is colon.
In one embodiment described the nd of formula I or a ceutically acceptable salt thereof and temozolomide are administered to the mammal.
In one ment described the compound of formula I or a ceutically acceptable salt thereof and temozolomide are administered to the mammal and the cancer is glioma.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and a platinum agent are administered to the mammal.
In one embodiment bed the compound of formula I or a pharmaceutically acceptable salt thereof and a platinum agent are administered to the mammal and the cancer is n.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt f and GDC-0973 or a pharmaceutically acceptable salt thereof are administered to the mammal.
In one embodiment described the compound of a I or a pharmaceutically acceptable salt thereof and GDC-0973 or a pharmaceutically acceptable salt thereof are administered to the mammal and the cancer is pancreatic, prostate, melanoma or breast.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and PLX-4032 or a pharmaceutically acceptable salt f are administered to the mammal.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof and PLX-4032 or a pharmaceutically acceptable salt thereof are administered to the mammal and the cancer is melanoma.
In one embodiment described the compound of formula I or a pharmaceutically acceptable salt thereof is administered orally.
In one embodiment described the compound of formula I or a pharmaceutically able salt thereof is ated as a tablet.
GENERAL PREPARATIVE PROCEDURES EXAMPLES In order to illustrate the invention, the following examples are ed. However, it is to be understood that these examples do not limit the invention and are only meant to t a method of practicing the invention. Persons d in the art will recognize that the chemical reactions described may be readily adapted to prepare a number of other AKT inhibitors useful in the invention, and alternative methods for ing the compounds useful in this invention are known. For example, the synthesis of emplified compounds useful in the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by ing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for ing other nds of the invention.
Example 1 N 2HCl HO Preparation of (S)amino(4-chlorophenyl)(4-((5R,7R)hydroxymethyl-6,7- dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl)propanone dihydrochloride Step 1: To a 1 L round-bottom flask were added (R)-(+)-Pulegone (76.12 g, 0.5 mmol), anhydrous NaHCO3 (12.5 g) and anhydrous ether (500 mL). The reaction mixture was cooled with ice-bath under nitrogen. The bromine (25.62 mL, 0.5 mmol) was added dropwise over 30 minutes. The mixture was filtered and carefully added to NaOEt (21%, 412 mL, 1.11 mmol) in an ice-cooled bath. The mixture was stirred at room temperature overnight and then 1 L of 5% HCl and 300 mL of ether were added. The aqueous phase was extracted with ether (2 x 300 mL). The combined organic phase was washed with water, dried and concentrated. The residue was added to a warmed solution of semicarbazide hydrochloride (37.5 g) and NaOAc (37.5 g) in water (300 mL), and then g ethanol (300 mL) was added to give a clear solution. The mixture was refluxed for 2.5 hours and then stirred at room temperature overnight. The mixture was treated with 1 L of water and 300 mL of ether. The aqueous phase was extracted with ether (2 x 300 mL). The combined organic phase was washed with water, dried and concentrated. The e was purified by vacuum distillation (73-76°C at 0.8 mm Hg) to give (2R)-ethyl 2-methyl(propan ylidene)cyclopentanecarboxylate (63 g, 64%). 1H NMR (CDCl 3, 400 MHz) δ 4.13 (m, 2H), 3.38 (d, J = 16 Hz, 0.5H), 2.93 (m, 0.5H), 2.50-2.17 (m, 2H), 1.98 (m, 1H), 1.76 (m, 1H), 1.23 (m, 6H), 1.05 (m, 6H).
Step 2: (2R)-Ethyl 2-methyl(propanylidene)cyclopentanecarboxylate (24 g, 0.122 mol) in ethyl acetate (100 mL) was cooled to –68°C with dry ice/isopropanol.
Ozonized oxygen (5-7 ft3h-1 of O2) was bubbled through the solution for 3.5 hours. The reaction mixture was flushed with nitrogen at room temperature until the color disappeared.
The ethyl acetate was removed under vacuum and the residue was dissolved in 150 mL of acetic acid and cooled by ice water, and zinc powder (45 g) was added. The solution was stirred for 30 minutes and then filtered. The filtrate was lized with 2N NaOH (1.3 L) and NaHCO3. The aqueous phase was extracted with ether (3 x 200 mL). The organic phase was combined, washed with water, dried and trated to afford (2R)-ethyl 2-methyl oxocyclopentanecarboxylate (20 g, 96%). 1H NMR (CDCl 3, 400 MHz) δ 4.21 (m, 2H), 2.77 (d, J = 11.2 Hz, 1H), 2.60 (m, 1H), 2.50-2.10 (m, 3H), 1.42 (m, 1H), 1.33 (m, 3H), 1.23 (m, 3H).
Step 3: To a solution of a e of (2R)-ethyl 2-methyl oxocyclopentanecarboxylate (20 g, 117.5 mmol) and thiourea (9.2 g, 120.9 mmol) in ethanol (100 mL) was added KOH (8.3 g, 147.9 mmol) in water (60 mL). The mixture was refluxed for 10 hours. After g, the solvent was removed and the residue was lized with concentrated HCl (12 mL) at 0°C and then extracted with DCM (3 x 150 mL). The solvent was removed and the residue was purified by silica gel chromatography, eluting with Hexane/ethyl acetate (2:1) to give (R)mercaptomethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinol (12 g, 56%). MS (APCI+) [M+H] +183.
Step 4: To a suspension of (R)mercaptomethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinol (12 g, 65.8 mmol) in distilled water (100 mL) was added Raney Nickel (15 g) and NH4OH (20 mL). The mixture was ed for 3 hours then filtered, and the filtrate was concentrated to afford (R)methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin- 4-ol (9.89 g, 99%). MS (APCI+) [M+H] +151.
Step 5: A mixture of (R)methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinol (5.8 g, 38.62 mmol) in POCl3 (20 mL) was refluxed for 5 minutes. Excess POCl3 was removed under vacuum and the residue was dissolved in DCM (50 mL). The mixture was then added to saturated NaHCO3 (200 mL). The aqueous phase was extracted with DCM (3 x 100 mL), and the combined c phases were dried and concentrated. The residue was purified by silica gel chromatography, eluting with ethyl acetate to give (R)chloromethyl-6,7- o-5H-cyclopenta[d]pyrimidine (3.18 g, 49%). 1H NMR (CDCl3, 400 MHz) δ 8.81 (s, 1H), 3.47 (m, 1H), 3.20 (m, 1H), 3.05 (m, 1H), 2.41 (m, 1H), 1.86 (m, 3H), 1.47 (m, 3H).
Step 6: To a solution of (R)chloromethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidine (2.5 g, 14.8 mmol) in CHCl3 (60 mL) was added MCPBA (8.30 g, 37.0 mmol) in three ns. The mixture was d at room temperature for 2 days. The mixture was cooled to 0°C and to this was added dropwise Na2S2O3 (10 g) in water (60 mL), followed by Na2CO3 (6 g) in water (20 mL). The reaction mixture was stirred for 20 minutes. The aqueous phase was extracted with CHCl3 (2 x 200 mL), and the combined organic phases were concentrated at low temperature (<25°C). The residue was purified by silica gel chromatography, eluting with ethyl acetate-DCM/MeOH (20:1) to give (R) chloromethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidine-oxide (1.45 g, 53%). 1H NMR (CDCl3, 400 MHz) δ 8.66 (s, 1H), 3.50 (m, 1H), 3.20 (m, 2H), 2.44 (m, 1H), 1.90 (m, 1H), 1.37 (d, J = 7.2 Hz, 3H).
Step 7: A solution of (R)chloromethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidine-oxide (1.45 g, 7.85 mmol) in acetic anhydride (20 mL) was heated to 110°C for 2 hours. After cooling, excess t was removed under vacuum. The residue was purified by silica gel chromatography, eluting with Hexane/ethyl acetate (3:1) to give -chloromethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl acetate (1.25 g, 70%). 1H NMR , 400 MHz) δ 8.92 (m, 1H), 6.30-6.03 (m, 1H), .30 (m, 1H), 2.84 (m, 1H), 2.40-2.20 (m, 1H), 2.15 (d, J = 6 Hz, 2H), 1.75 (m, 2H), 1.47 (d, J = 6.8, 2H), 1.38 (d, J = 7.2, 1H). MS (APCI+) [M+H] +227.
Step 8: To a solution of (5R)chloromethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl acetate (0.5 g, 2.2 mmol) in NMP (10 mL) was added 1-Bocpiperazine (0.9 g, 4.8 mmol). The reaction mixture was heated to 110°C for 12 hours. After cooling, the reaction mixture was diluted with ethyl acetate (200 mL) and washed with water (6 x 100 mL). The organic phase was dried and concentrated. The residue was purified by silica gel chromatography, eluting with ethyl acetate to give tert-butyl 4-((5R)acetoxy methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinecarboxylate (0.6 g, 72%). 1H NMR (CDCl3, 400 MHz) δ 8.60 (d, 1H), 6.05-5.90 (m, 1H), 3.80-3.30 (m, 9H), 2.84 (m, 1H), 2.20- (m, 1H), 1.49 (s, 9H), 1.29-1.20 (m, 3H). MS (APCI+) [M+H] +377. The resulting mixture of the diastereomers was purified by chiral separation HPLC (Chiralcel ODH column, 250 x 20 mm, Hexane/EtOH 60:40, 21 mL/min). The first peak (RT = 3.73 min) gave the tert-butyl 4-((5R,7R)acetoxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin- iperazinecarboxylate (0.144 g, 24%). The second peak (RT = 5.66 min) gave the tert-butyl 4-((5R,7S)acetoxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate (0.172 g, 29%). MS (APCI+) [M+H] +377.
Step 9: To a solution of tert-butyl 4-((5R,7R)acetoxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate (0.144 g, 0.383 mmol) in THF (4 mL) was added LiOH (3M, 2 mL). The mixture was stirred at room ature for 6 hours and then quenched with 2N HCl (3 mL). The solvent was d and the residue was purified by silica gel tography, eluting with ethyl acetate to give tert-butyl 4-((5R,7R) hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinecarboxylate (89 mg, 70%). %). 1H NMR , 400 MHz) δ 8.52 (s, 1H), 5.48 (br, 1H), 5.14 (m, 1H), 3.82-3.40 (m, 9H), 2.20 (m, 2H), 1.49 (s, 9H), 1.19 (d, J = 6.8 Hz, 3H). MS (APCI+) [M+H] +335.
Step 10: tert-Butyl 4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate was treated with HCl (4M in dioxane, 2 mL) in DCM (5 mL) for 6 hours to give (5R,7R)methyl(piperazinyl)-6,7-dihydro- 5H-cyclopenta[d]pyrimidinol dihydrochloride. MS (APCI+) [M+H] +235.
Step 11: Tert-butyl 2,4-dimethoxybenzylcarbamate (3.96 g, 14.8 mmol) was dissolved in THF (74 mL) and cooled to -78°C. The on was treated with butyl lithium (7.44 mL, 16.3 mmol) dropwise over a five minute period to afford a pale-yellow solution.
The solution was allowed to stir for 15 minutes before the (methoxy)methane (1.35 mL, 17.8 mmol) was added dropwise (neat). The reaction was stirred at -78°C for 10 minutes, then allowed to warm slowly to ambient temperature overnight. The reaction was concentrated in vacuo to afford a yellow gel which was partitioned between half-saturated NH4Cl solution and ether. The aqueous layer was extracted once, and the organics were combined. The organic layer was washed with water, then brine, separated, dried over Na2SO4, ed, and concentrated in vacuo. 1H NMR supports the desired near-pure (>90%) tert-butyl 2,4-dimethoxybenzyl(methoxymethyl)carbamate (4.81 g, 104% yield) as a pale-yellow oil which was used t purification.
Step 12: (R)benzyl(2-(4-chlorophenyl)acetyl)oxazolidinone (3.00 g, 9.10 mmol) was dissolved in DCM (91 mL) and cooled to -78°C. A 1M toluene solution of TiCl4 (11.4 mL, 11.4 mmol) was added to the solution followed by DIEA (1.66 mL, 9.55 mmol) to afford a dark purple reaction. This was allowed to stir for 15 minutes before the tert-butyl 2,4-dimethoxybenzyl(methoxymethyl)carbamate (3.40 g, 10.9 mmol) was added as a solution in DCM (10 mL) dropwise. The reaction was d to stir for 15 minutes at -78°C, then allowed to warm to -18°C in a brine-ice bath for one hour. This reaction was allowed to warm slowly to 0°C over a 2.5 hour period. The reaction was then quenched with the addition of saturated NH4Cl on (100 mL). The layers were ted, and the c layers was extracted once with DCM. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo to afford a yellow oil. The residue was purified by chromatography (silica gel eluted with 4:1 s:ethyl acetate) to afford the pure material as a colorless oil tert-butyl 2,4-dimethoxybenzyl((S)((R)benzyl oxooxazolidinyl)(4-chlorophenyl)oxopropyl)carbamate (4.07 g, 73.5% yield). This tert-butyl 2,4-dimethoxybenzyl((S)((R)benzyloxooxazolidinyl)(4- chlorophenyl)oxopropyl)carbamate (680 mg, 1.12 mmol) was dissolved in DCM (10.6 mL) and water (560 uL; 19:1 DCM:water) at t temperature. The on was treated with DDQ (380 mg, 1.67 mmol), and the reaction was allowed to stir for one day to afford reaction completion by TLC and LCMS is. The reaction was diluted with DCM and washed twice with half saturated NaHCO3 solution. The organic layer was dried over MgSO4, filtered, and concentrated in vacuo to afford a yellow-orange oil. The residue was purified by chromatography (silica gel eluted with 9:1 hexanes:ethyl acetate) to afford a mixture of the aldehyde by-product and tert-butyl (S)((R)benzyloxooxazolidinyl)- 2-(4-chlorophenyl)oxopropylcarbamate (not separable) as a pale-yellow oil (729 mg combined mass). LC/MS (APCI+) m/z 359.1 [M-BOC+H]+.
Step 13: 35% H2O2 (0.240 mL, 2.91 mmol) was added to a solution of LiOH-H2O (0.0978 g, 2.33 mmol) in 2:1 THF:H2O (33 mL). The reaction mixture was stirred at room temperature for 35 minutes, and then cooled to 0°C. A solution containing a e of tertbutyl (S)((R)benzyloxooxazolidinyl)(4-chlorophenyl)oxopropylcarbamate (0.535 g, 1.17 mmol) and 2,4-dimethoxybenzaldehyde (0.194 g, 1.17 mmol) in THF (7 mL) was added dropwise by addition funnel. The ice bath was allowed to slowly warm, and the reaction mixture was stirred overnight. The reaction mixture was then cooled to 0°C, and 1M Na2SO3 (7 mL) was added. The e was stirred for 5 minutes, and then warmed to room temperature and stirred an additional 20 minutes. The reaction mixture was then transferred to a separatory funnel and washed with ether (3 X). The aqueous layer was acidified with KHSO4(s), and the mixture was extracted with DCM (2 X). The ed extracts were dried (Na2SO4), filtered, and concentrated to give (S)(tert-butoxycarbonylamino)(4- chlorophenyl)propanoic acid (0.329 g, 94.2% yield) as a white residue. LC/MS ) m/z 200 [M-BOC+H]+.
Step 14: 4M HCl/dioxane (5.49 ml, 22.0 mmol) was added to a on of (S) butoxycarbonylamino)(4-chlorophenyl)propanoic acid (0.329 g, 1.10 mmol) in 2:1 dioxane:DCM (10 mL). The reaction mixture was stirred at room temperature overnight (16 hours), after wihch it was concentrated to 1/3 volume. The resulting cloudy mixture was diluted with ether, and the mixture was concentrated again to 1/3 volume. The mixture was diluted again with ether (20 mL), and the solids were isolated by filtration through a medium frit funnel with nitrogen pressure, rinsed with ether (5 X 10mL), dried under nitrogen pressure, and dried in vacuo to give (S)amino(4-chlorophenyl)propanoic acid hydrochloride (0.199 g, 76.8% yield) as a white powder. HPLC >99 area% pure. LC/MS ) m/z 200.
Step 15: Boc2O (0.368 g, 1.69 mmol) was added to a solution of (S)amino(4- phenyl)propanoic acid hydrochloride (0.199 g, 0.843 mmol) and tetramethylammonium hydroxide pentahydrate (0.382 g, 2.11 mmol) in 10:1 MeCN:H2O (7.7 mL). The on mixture was stirred overnight at room temperature (12 hours), after which the MeCN was removed on a rotary evaporator. The mixture was diluted with water and washed with ether (2 X). The aqeuous layer was acidified with KHSO4(s), the mixture was extracted with DCM, and the combined extracts were dried (Na2SO4), filtered, and concentrated to give (S)(tert-butoxycarbonylamino)(4-chlorophenyl)propanoic acid (0.229 g, 90.6% yield) as a foam. HPLC >99 area% pure. LC/MS (APCI+) m/z 200 [MBOC +H]+.
Step 16: To a solution of (5R,7R)methyl(piperazinyl)-6,7-dihydro-5H- cyclopenta[d]pyrimidinol dihydrochloride (88 mg, 0.29 mmol) and (S)(tert- butoxycarbonylamino)(4-chlorophenyl)propanoic acid (86 mg, 0.29 mmol) in DCM (10 mL) and Diisopropylethylamine (0.22 mL, 1.3 mmol) was added HBTU (110 mg, 0.29 mmol). The reaction mixture was stirred at room temperature for 1 hour. The solvent was removed and the residue was ved in ethyl acetate (100 mL), washed with water (6x50ml). The organic phase was dried and concentrated to give utyl (S)(4- chlorophenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinyl)oxopropylcarbamate (116 mg, 78%). 1H NMR (CDCl3, 400 MHz) δ 8.51 (s, 1H), 7.34-7.20 (m, 4H), 5.15-5.09 (m, 2H), 4.15-4.05 (m, 1H), 3.87-3.85 (m, 2H), 3.78-3.38 (m, 7H), 3.22-3.19 (m, 1H), 2.20-2.10 (m, 2H), 1.48 (s, 9H), 1.41 (s, 9H), 1.14- 1.12 (d, z, 3H). MS (APCI+) [M+H] +516.
Step 17: Treatment of tert-butyl (S)(4-chlorophenyl)(4-((5R,7R)hydroxy methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl)oxopropylcarbamate with HCl (4M in e, 2 mL) in DCM (5 mL) for 6 hours to give (S)amino(4- chlorophenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinyl)propanone dihydrochloride. 1H NMR (D2O, 400 MHz) δ 8.38 (s, 1H), 7.37-7.35 (d, J=8.4Hz, 2H), 7.23-7.21 (d, J=8.4Hz, 2H), 5.29-5.25 (m, 1H), 4.64 (s, 9H), 4.31-4.28 (m, 1H), 4.11 (m, 1H), 3.88-3.79 (m, 2H), 3.70-3.20 (m, 10H), 2.23-2.17 (m, 1H), .99 (m, 1H), 1.22-1.20 (m, 2H), .96 (d, J = 6.8 Hz, 2H). MS ) [M+H] +416.
Example 2 (4-chlorophenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinyl)(isopropylamino)propanone Step 1: Ethyl pulegenate (130 g, 662 mmol) in EtOAc (900 mL) was cooled to -78°C using a dry ice-isopropanol bath. This mixture was subjected to ozonolysis until the reaction turned purple in color. At this point, ozone generation ceased, and the reaction was removed from the dry-ice bath. Oxygen was d through the reaction mixture until it turned yellow. The reaction mixture was concentrated under vacuum, and the resulting residue was dissolved in glacial acetic acid (400 mL). The solution was cooled to 0°C, and Zn dust (65 g, 993 mmol) was added portionwise over 30 minutes. The reaction was then allowed to stir for 2 hours, at which point the reaction mixture was filtered through a pad of celite to remove the zinc dust. The acetic acid was neutralized to pH 7 with aqueous NaOH and NaHCO3 and extracted with ether (3 X 800 mL). The combined organics were dried with brine, MgSO4 and concentrated to give (2R)-ethyl 2-methyl oxocyclopentane-carboxylate as a brown liquid (107g, 95%).
Step 2: Ammonium acetate (240.03 g, 3113.9 mmol) was added to a solution of (R)- ethyl 2-methyloxocyclopentanecarboxylate (106.0 g, 622.78 mmol) in MeOH (1.2L). The reaction mixture was stirred at room temperature under nitrogen for 20 hours, after which it was complete as judged by TLC and HPLC. The reaction mixture was concentrated to remove MeOH. The ing residue was dissolved in DCM, washed twice with H2O, once with brine, dried (Na2SO 4), ed, and concentrated to give (R)-ethyl 2-amino methylcyclopentenecarboxylate (102 g, 97% yield) as an orange oil. LC/MS (APCI+) m/z 170 [M+H]+.
Step 3: A solution containing (R)-ethyl 2-aminomethylcyclopentenecarboxylate (161.61 g, 955.024 mmol) and ammonium formate (90.3298 g, 1432.54 mmol) in formamide (303.456 ml, 7640.19 mmol) was heated to an internal temperature of 150°C and stirred for 17 hours. The reaction mixture was cooled, and transferred to a 2L single neck flask. Then excess formamidine was d by high vacuum distillation. Once formamidine stopped coming over, the remaining oil in the still pot was dissolved in DCM and washed with brine (3 X 200 mL). The ed s washes were extracted with DCM. The combined organic extracts were dried (Na2SO 4), filtered, and concentrated. The resulting brown oil was dissolved in minimal DCM, and this solution was added using a separatory funnel to a stirred solution of ether (ca. 5 vol of ether vs. DCM solution), g some brown precipitate to form. This brown precipitate was removed by filtration through a medium frit funnel which was rinsed with ether and disposed. The filtrate was concentrated, the trituration from ether repeated two more times and then dried on high vacuum line to give (R)methyl-6,7- dihydro-5H-cyclopenta[d]pyrimidinol (93.225 g, 65.00% yield) as a brown -yellow pasty solid. LC/MS ) m/z 149.2.
Step 4: Neat POCl3 (463.9 ml, 5067 mmol) was added slowly by on funnel to a 0°C solution of (R)methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinol (152.2 g, 1013 mmol) in DCE (1.2 L). After the on was complete, the reaction mixture was warmed to room temperature, then heated to reflux and stirred for 70 minutes. The reaction was complete as determined by HPLC. The reaction mixture was cooled to room temperature, and the excess POCl3 was quenched in 4 portions as follows: Reaction e transferred to separatory funnel and dripped into a beaker containing ice and saturated NaHCO3 solution cooled in an ice bath. Once the addition of each portion of the reaction mixture was completed, the ed mixture was stirred 30 minutes to ensure complete destruction of POCl 3 prior to transfer to separatory funnel. The mixture was transferred to the separatory funnel and extracted twice with DCM. The combined ts were dried (Na2SO 4), filtered, and concentrated. The crude was purified on silica gel as follows: silica gel (1 kg) was ed in 9:1 hexane:ethyl acetate onto a 3L fritted funnel, silica settled under vacuum, topped with sand. The crude was loaded with a DCM/hexane mixture, and the compound was eluted using 1L sidearm flasks under vacuum. High Rf byproducts eluted first, then (R)- 4-chloromethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidine (104.4 g, 61.09% yield) as a brown oil. Triethylamine (93.0 ml, 534 mmol) and tert-butyl piperazinecarboxylate (34.8 g, 187 mmol) was added to a solution of (R)chloromethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidine (30.0 g, 178 mmol) in n-BuOH (250 mL). The reaction mixture was heated to reflux under nitrogen and stirred ght (17 hours), after which it was concentrated on a rotavap. The resulting oil was dissolved in DCM, washed with H2O, dried (Na 2SO 4), filtered, and was concentrated. The resulting brown oil was purified on silica gel eluting first with 2:1 hexanes:ethyl acetate until product eluting cleanly, then nt 1:1 to 1:5 DCM:ethyl acetate to give (R)-tertbutyl 4-(5-methyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate (42.0 g, 74.1% yield) as a beige powder. LC/MS (APCI+) m/z 319.1 [M+H]+.
Step 5: Solid 77% max. MCPBA (23.9 g, 107 mmol) was added nwise to a 0°C solution of rt-butyl 4-(5-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin erazinecarboxylate (20.0 g, 62.8 mmol) in CHCl 3 (310 mL). The reaction e was stirred 5 for minutes, then warmed to room temperature and stirred for 90 minutes.
HPLC looked similar after 7.5 hours. The reaction mixture was cooled to 0°C, then NaHCO3 (13.2 g, 157 mmol) and another 0.5 equivalents of m-CPBA were added. The reaction mixture was stirred overnight (14 hours). The reaction mixture was cooled to 0°C, and a solution of Na2S2O3 (29.8 g, 188 mmol) in H2O (50 mL) was added dropwise by addition funnel. This was followed by a solution of Na2CO 3 (24.6 g, 232 mmol) in H2O (70 mL) by addition funnel (mixture turns neous). The reaction mixture was stirred for 30 minutes, then the mixture was extracted with CHCl3 (3 X 150 mL). The combined extracts were dried (Na2SO 4), filtered, and concentrated to give the N-oxide. LC/MS (APCI+) m/z 335.1 .
Step 6: Ac2O (77.0 ml, 816 mmol) was added to the N-oxide (21.0 g, 62.8 mmol) from Step 5. The reaction mixture was heated under nitrogen in a 90°C sand bath and stirred for 100 minutes. The reaction mixture was cooled to room temperature, and excess acetic anhydride was removed by rotary evaporation. The ing oil was dissolved in DCM, which was then poured carefully into ice saturated Na 2CO 3. The mixture was ted with DCM, and the combined ts were dried (Na2SO 4), filtered, and concentrated to give (5R)-tert-butyl 4-(7-acetoxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate (23.6g, 100%) as a brown foam. LC/MS (APCI+) m/z 377.1 [M+H]+.
Step 7: LiOH-H2O (6.577 g, 156.7 mmol) was added to a 0°C solution of (5R)-tertbutyl 4-(7-acetoxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazine carboxylate (23.6 g, 62.69 mmol) in 2:1 THF:H2O (320 mL). The reaction mixture was stirred for 10 minutes, and then warmed to room temperature. LC/MS looked the same at 3 hours and 4.5 hours. The reaction mixture was cooled to 0°C, and then saturated NH4Cl was added to the mixture. The mixture was stirred for 5 s, and most of the THF was removed by rotary evaporation. The mixture was extracted with EtOAc (3 X 250 mL), and the combined extracts were dried (Na2SO 4), ed, and concentrated. The crude was flashed on Biotage 65M: 4:1 DCM:ethyl acetate, then gradient to 1:1 to 1:4 DCM:ethyl acetate. Once the product was eluting, then ethyl acetate was flushed through the column.
Then 30:1 DCM:MeOH eluted the rest of the product (8.83 g). The mixed fractions were reflashed with Biotage 40M using the same ions to give another 2.99 g which gave a combined yield of (5R)-tert-butyl 4-(7-hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate (11.82 g, 56.38% yield) as a brown foam. LC/MS ) m/z 335.1 [M+H]+.
Step 8: A solution of DMSO (5.45 ml, 76.8 mmol) in DCM (50 mL) was added dropwise by addition funnel to a -78°C solution of oxalyl chloride (3.35 ml, 38.4 mmol) in DCM (150 mL). The reaction mixture was stirred for 35 s, and then a solution of (5R)-tert-butyl 4-(7-hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate (9.17 g, 27.4 mmol) in DCM (80 mL) was added slowly by addition funnel. The on mixture was stirred another 1 hour at -78°C, after which neat triethylamine (18.0 ml, 129 mmol) was added to the mixture. The on mixture was then allowed to warm to room temperature, and then it was stirred for 30 s. H2O was added. The e was extracted with DCM (3 X 200 mL), and the combined extracts were dried (Na2SO 4), filtered, and concentrated in vacuo. The crude was purified on silica gel (Biotage 65M): the column was flushed with ca. 800 mL 4:1 DCM:EtOAc, then gradient to 1:1 DCM:ethyl acetate until product eluting, then 1:4 DCM:EtOAc eluted product to give (R)-tert-butyl 4-(5-methyloxo-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazine carboxylate (7.5 g, 82.3% yield) as a brown foam. The foam was concentrated (3 X) from xanes, which gave a very light brown foam. HPLC >95% area. LC/MS (APCI+) m/z 333 [M+H]+.
Step 9: Triethylamine (4.33 ml, 31.1 mmol; degassed with nitrogen 30 minutes prior to use) and formic acid (1.36 ml, 36.1 mmol; degassed with nitrogen 30 minutes prior to use) were added to a solution of (R)-tert-butyl 4-(5-methyloxo-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate (9.75 g, 29.3 mmol) in DCM (210 mL; degassed with nitrogen 30 minutes prior to use). The mixture was d for 5 minutes, then a Ru catalyst (0.0933 g, 0.147 mmol) was added. The reaction was stirred under positive nitrogen pressure overnight (18 hours). The reaction e was concentrated to dryness and dried on high vacuum. The impure material was flashed on Biotage 65M loaded 1:1 hyl acetate 500 mL flushed, then 1:4 DCM:ethyl acetate until product (2nd spot), then gradient to neat ethyl acetate, then 25:1 DCM:MeOH eluted rest of product. The fractions were combined and concentrated on a rotary evaporator. The residue was concentrated again from DCM/hexanes to give a mixture of tert-butyl 4-((5R,7R)hydroxy- -methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinecarboxylate (major) and tert-butyl 4-((5R,7S)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate ) (9.35 g, 95.3% yield) as a beige foam. LC/MS (APCI+) m/z 335 [M+H]+. 1H NMR (CDCl3) shows 88% de by integration of carbinol methine.
Step 10: 4-Nitrobenzoyl chloride (4.27 g, 23.0 mmol) was added to a 0°C solution of tert-butyl 4-((5R,7R)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate (7.0 g, 20.9 mmol) and triethylamine (4.38 ml, 31.4 mmol) in DCM (110 mL). The reaction mixture was d at room temperature overnight, after which saturated NaHCO3 was added. The mixture was stirred 10 minutes, and then extracted with DCM. The combined extracts were dried (Na2SO 4), filtered, and concentrated. The crude was flashed on Biotage 65M (3:1 hexanes:ethyl acetate loaded crude, then 2:1 hexanes:ethyl acetate eluted tert-butyl 4-((5R,7R)methyl(4-nitrobenzoyloxy)-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate and a few mixed fractions). Then tertbutyl ,7S)methyl(4-nitrobenzoyloxy)-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate was eluted using 1:2 hexanes:ethyl acetate. The ons with product were concentrated by rotary evaporation to give tert-butyl 4-((5R,7R)methyl(4- nitrobenzoyloxy)-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinecarboxylate (8.55 g, 84.5% yield) as a yellow foam. LC/MS (APCI+) m/z 484 [M+H]+. 1H NMR (CDCl3) shows single diastereomer). The fractions with other diastereomer were concentrated by rotary ation to give utyl 4-((5R,7S)methyl(4- enzoyloxy)-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinecarboxylate (0.356 g, 3.52% yield) as a brown foam. LC/MS (APCI+) m/z 484 [M+H]+.
Step 11: LiOH-H2O (0.499 g, 11.9 mmol) was added to a 0°C solution of tert-butyl 4- ((5R,7R)methyl(4-nitrobenzoyloxy)-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate (2.30 g, 4.76 mmol) in 2:1 THF:H2O (40 mL). The reaction mixture was warmed to room temperature and stirred for 1 hour. The THF was removed by rotary evaporation, ted NaHCO3 was added, and the mixture was extracted with ethyl acetate. The combined extracts were washed (1 X) with saturated , dried (Na2SO 4), filtered, and concentrated to give utyl 4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinecarboxylate (1.59 g, 100.0% yield) as a yellow foam. HPLC after workup just product>98 area% pure. LC/MS (APCI+) m/z 335 [M+H]+.
The tert-butyl 4-((5R,7S)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin yl)piperazinecarboxylate was prepared using an analogous method.
Step 12: 4M oxane (11.2 ml, 44.9 mmol) was added to a solution of tert-butyl 4-((5R,7R)hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazine carboxylate (0.600 g, 1.79 mmol) in dioxane (15 mL). The reaction mixture was stirred at room temperature under nitrogen overnight (20 hours). The mixture was trated to dryness and dried on high vacuum line. The crude was suspended in ether, sonicated, and stirred for 5 minutes. The solids were isolated by filtration through a medium frit funnel with nitrogen pressure, rinsed with ether, dried under nitrogen pressure, and dried further on a hi vacuum line to give (5R,7R)methyl(piperazinyl)-6,7-dihydro-5H- enta[d]pyrimidinol dihydrochloride (0.440 g, 79.8% yield) as a yellow powder.
LC/MS (APCI+) m/z 235. The (5R,7S)methyl(piperazinyl)-6,7-dihydro-5H- cyclopenta[d]pyrimidinol dihydrochloride was prepared using an analogous method.
Step 13: Methyl 2-(4-chlorophenyl)acetate (36.7 g, 199 mmol) and paraformaldehyde (6.27 g, 209 mmol) were dissolved/suspended in DMSO (400 mL) and treated with NaOMe (537 mg, 9.94 mmol). The e was allowed to stir at room temperature for 2 hours to completion by TLC analysis of the crude. The reaction was poured into ice-cold water (700 mL; white emulsion) and neutralized with the addition of 1M HCl solution. The aqueous layer was extracted with ethyl e (3 X), and the organics were combined. The organic layer was washed with water (2 X), brine (1 X), separated, dried over MgSO4, filtered, and concentrated in vacuo to afford the crude product as a yellow oil. The residue was loaded onto a large fritted filtered with silica gel and eluted with 9:1 hexanes:ethyl acetate until the starting material/olefin were collected. The plug was then eluted with 1:1 hexanes:ethyl acetate until the pure desired product was eluted tely. The concentrated pure fractions yielded methyl 2-(4-chlorophenyl)hydroxypropanoate as a ess oil (39.4g, 92%).
Step 14: Methyl 2-(4-chlorophenyl)hydroxypropanoate (39.4 g, 184 mmol) was dissolved in DCM (500 mL) and d with TEA (64.0 mL, 459 mmol). The solution was cooled to 0°C and slowly treated with MsCl (15.6 mL, 202 mmol), then allowed to stir for 30 minutes to completion by TLC analysis. The solution was partitioned with 1N HCl on, and the s layer was extracted once with DCM. The combined organic layer was washed once more with 1N HCl solution, separated, washed with diluted NaHCO3 solution, and separated. The c layer was dried over MgSO 4, filtered, and concentrated in vacuo to afford an orange oil. The residue was loaded onto a large fritted filter with a plug of silica gel and eluted with 9:1 s:ethyl acetate affording the pure desired product by TLC analysis. The concentrated pure ons yielded the methyl 2-(4-chlorophenyl)acrylate as a colorless oil (30.8 g, 85%). This methyl 2-(4-chlorophenyl)acrylate (500 mg, 2.54 mmol) was added as a solution in THF (1.35 mL) to a ng solution of i-PrNH2 (217 uL, 2.54 mmol) in THF (5.0 mL) at 0°C. The reaction was allowed to stir at room temperature overnight to completion by LCMS analysis. The Boc2O (584 uL, 2.54 mmol) was added to the ng amine via pipet. The reaction was allowed to stir overnight to completion by LCMS and TLC analysis of the mixture. The solution was concentrated in vacuo to afford methyl 3-(tert-butoxycarbonyl(isopropyl)amino)(4-chlorophenyl)propanoate as a colorless oil (854 mg, 94%). LC/MS (APCI+) m/z 256.1 [M-Boc]+.
Step 15: Methyl t-butoxycarbonyl(isopropyl)amino)(4- chlorophenyl)propanoate (133 g, 374 mmol) was dissolved in THF (1.0 L) and treated with KOTMS (56.0 g, 392 mmol) at room temperature. The mixture was allowed to stir overnight to completion by LCMS is of the crude. The mixture was concentrated in vacuo to afford a wet foam, which was allowed to dry under vacuum overnight to afford potassium 3- (tert-butoxycarbonyl(isopropyl)amino)(4-chlorophenyl)propanoate as a white solid (148.7 g, 105%). LC/MS (APCI+) m/z 242.1 [M-Boc-K]+.
Step 16: Potassium 3-(tert-butoxycarbonyl(isopropyl)amino)(4- chlorophenyl)propanoate (77.2 g, 203 mmol) was dissolved in THF (515 mL) and treated with pivaloyl chloride (26.3 mL, 213 mmol) at room ature. The mixture was allowed to stir for 3 hours to form the mixed anhydride. (S)benzyloxazolidinone (46.1 g, 260 mmol) was dissolved in THF (600 mL) and cooled to -78°C in a separate flask. The on was treated with n-BuLi (102 mL of a 2.50M solution in hexanes, 254 mmol) and allowed to stir for one hour. The prepared anhydride solution was added to the stirring Li-oxazolidinone via cannula, and the mixture was allowed to warm to room temperature overnight. The mixture was quenched with the addition of saturated ammonium chloride solution, then partitioned between more water and ethyl acetate. The s layer was extracted several times, and the organics were combined. The organic layer was washed with water, then brine, separated, dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified/separated (diastereomers) via chromatography (silica gel eluted with 4:1 hexanes:ethyl acetate) to afford the completely separated diastereomers as viscous oils: tertbutyl (R)((S)benzyloxooxazolidinyl)(4-chlorophenyl) pyl(isopropyl)carbamate (12.16 g, 24% based on 1/2 of acid racemate) and tert-butyl (S)((S)benzyloxooxazolidinyl)(4-chlorophenyl) oxopropyl(isopropyl)carbamate (39.14 g, 77% based on 1/2 of acid racemate). LC/MS (APCI+) m/z 401.2 [M-Boc]+.
Step 17: LiOH-H2O (168 mg, 4.00 mmol) was added to a stirring solution of THF (30 mL) and water (15 mL) at room ature until it was dissolved. The mixture was treated with hydrogen peroxide (658 uL of a 35% wt. solution in water, 8.00 mmol) and allowed to stir at room ature for 10 s. The on was cooled to 0°C in an ice bath, and the tert-butyl (S)((S)benzyloxooxazolidinyl)(4-chlorophenyl) oxopropyl(isopropyl)carbamate (1.00 g, 2.00 mmol) was added se via addition funnel as a solution in THF (15 mL) over a 10 minutes. The mixture was allowed to stir overnight at room temperature to completion by LCMS analysis of the crude. The reaction was cooled to 0°C, and then treated with 1M Na2SO 3 (9.00 mL) solution via addition funnel over a ten minute period. After the addition was complete, the mixture was allowed to warm to room ature for 10 minutes. The mixture was concentrated to remove the THF, and then d with water. The aqueous layer was washed twice with ethyl e (discarded). The s layer was partitioned with ethyl acetate, then treated dropwise while stirring with 1M HCl until pH 2-3 was attained. The aqueous layer was extracted twice with ethyl acetate, and the organics were combined. The organic was washed with brine, separated, dried over MgSO 4, filtered, and concentrated in vacuo. The colorless oil product was dried under high vacuum for one hour to afford (S)(tert-butoxycarbonyl(isopropyl)amino)(4- chlorophenyl)propanoic acid as a viscous oil/foam (685 mg, 100%). LC/MS (APCI+) m/z 242.1 [M-Boc]+.
Step 18: A solution of (5R,7R)methyl(piperazinyl)-6,7-dihydro-5H- cyclopenta[d]pyrimidinol dihydrochloride (2.92 g, 9.51 mmol) and (S)(tert- butoxycarbonyl(isopropyl)amino)(4-chlorophenyl)propanoic acid (3.25 g, 9.51 mmol) in DCM (40 mL) and DIEA (5.0 mL, 28.7 mmol) was stirred at room temperature for 10 minutes. HBTU (3.61g, 9.51 mmol) was added to the mixture. The mixture was stirred at room temperature for 1 hour. The solvent was removed, and the residue was dissolved in ethyl acetate (500 mL) and washed with water (6 X 100 mL). The organic phase was dried and trated. The residue was subject to column chromatography, eluted by EtOAc- DCM/MeOH (20:1) to give tert-butyl (S)(4-chlorophenyl)(4-((5R,7R)hydroxy methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl) oxopropyl(isopropyl)carbamate (3.68g, 69%.) LC/MS (APCI+) m/z 558.2 [M+H]+.
Step 19: The tert-butyl (S)(4-chlorophenyl)(4-((5R,7R)hydroxymethyl- 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl)oxopropyl(isopropyl) ate (2.50 g, 4.48 mmol) was dissolved in dioxane (22.4 mL) and treated with 4M HCl in dioxane (22.4 mL, 89.6 mmol) at room temperature. The resulting solution was allowed to stir overnight to completion by LCMS analysis of the crude. The solution was concentrated in vacuo to afford a gel that was dissolved in a minimal amount of methanol (10 mL). The solution was transferred via pipette to stirred ether (300 mL) to afford a white precipitate of desired t. The addition was about half when the white precipitate melted into a yellow gel. The al was concentrated in vacuo to afford a yellow gel which was allowed to stand under reduced pressure overnight to yield (S)(4-chlorophenyl)(4-((5R,7R) hydroxymethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl) (isopropylamino)propanone dihydrochloride as a light yellow powder (2.14 g, 90%). 1H NMR (D 2O, 400 MHzδ δ 8.39 (s, 1H), 7.37-7.35 (d, J = 8.4 Hz, 2H), .20 (d, J = 8.4 Hz, 2H), 5.29-5.25 (m, 1H), 4.33-4.29 (m, 1H), 4.14-4.10 (m, 1H), 3.89-3.19 (m, 11H), 2.23-2.17 (m, 1H), 2.08-1.99 (m, 1H), 1.20-1.18 (m, 6H), 0.98-0.96 (d, J = 6.8 Hz, 3H). MS ) [M+H] +458.
Examples 3-9 shown in Table 1 can also be made according to the above-described methods.
Table 1 LCMS or 1H Example Structure Name O (4-chlorophenyl) 3 Cl (dimethylamino)(4-((5R,7R) hydroxymethyl-6,7-dihydro-5H- 444.1 N cyclopenta[d]pyrimidin N yl)piperazinyl)propanone (S)(3-fluoro F O (trifluoromethyl)phenyl)(4- 4 F3C ((5R,7S)hydroxymethyl-6,7- 510.3 N dihydro-5H-cyclopenta[d]pyrimidin- 4-yl)piperazinyl) N (isopropylamino)propanone O (S)(4-chlorophenyl)(4- N ((5R,7S)hydroxymethyl-6,7- Cl o-5H-cyclopenta[d]pyrimidin- 458.3 N 4-yl)piperazinyl) N (isopropylamino)propanone (R)(4-chlorophenyl)(4- Cl ((5R,7R)hydroxymethyl-6,7- N dihydro-5H-cyclopenta[d]pyrimidin- 458 4-yl)piperazinyl) N (isopropylamino)propanone HN (4-chloro F O fluorophenyl) LCMS 7 N (cyclopropylmethylamino)(4- Cl (APCI+) m/z 488, ((5R,7R)hydroxymethyl-6,7- N 490 [M+H]+ dihydro-5H-cyclopenta[d]pyrimidin- N 4-yl)piperazinyl)propanone HN (S)(4-chloro F O fluorophenyl)(4-((5R,7R) LCMS 8 hydroxymethyl-6,7-dihydro-5H- Cl ) m/z 518, cyclopenta[d]pyrimidin 520 [M+H]+ N yl)piperazinyl)(tetrahydro-2H- N pyranylamino)propanone (S)(4-chloro fluorophenyl)(4-((5R,7R) hydroxymethyl-6,7-dihydro-5H- 9 LCMS cyclopenta[d]pyrimidin (APCI+) m/z 546 yl)piperazinyl)((1r,4S) methoxycyclohexylamino)propan Example 10 (S)(4-cyclopropylphenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinyl)((S)-pyrrolidinyl)ethanone Step 1: Cyclopropylmagnesium bromide (64.0 mL, 32.00 mmol) in THF was treated with a on of zinc (II) chloride (64.00 mL, 32.00 mmol) in THF. The mixture was stirred at ambient ature for 20 minutes. 2-(4-Bromophenyl)acetonitrile (5.228 g, 26.67 mmol) and bis[tri-t-butyl phosphine]palladium (0.6814 g, 1.333 mmol) were added as a solution in THF (2 mL). The reaction was stirred at ambient temperature under nitrogen for 12 hours. The reaction was quenched with saturated NH4Cl, diluted with methylene chloride and separated. The aqueous layer was washed with methylene chloride (2 X), and then the combined organic layers were washed with water (3 X), dried over Na2SO4 and concentrated in vacuo. The crude product was subjected to chromatography on SiO2 eluting with 25:1 hexanes/ethyl e to yield 2-(4-cyclopropylphenyl)acetonitrile (2.76 g, 66%). 1H NMR (CDCl3, 400 MHz) δ7.20 (d, J = 8.2, 2H), 7.07 (d, J = 8.2, 2H), 3.70 (s, 2H), 1.94-1.85 (m, 1H), .95 (m, 2H), 0.71-0.66 (m, 2H).
Step 2: Methanol (65 mL) was cooled to 0°C and saturated with HCl (g). This solution was d with a solution of 2-(4-cyclopropylphenyl)acetonitrile (2.76 g, 17.56 mmol) in methanol (6 mL). The reaction mixture was heated to reflux ght under a drying tube containing CaSO4. The reaction was cooled and concentrated in vacuo. The crude mixture was re-suspended in ethyl acetate and water and then ted. The organic layer was washed with saturated NaHCO3, saturated NaCl, dried over Na2SO4 and concentrated in vacuo to provide methyl 2-(4-cyclopropylphenyl)acetate as an oil (3.10 g, 93%). 1H NMR (CDCl3, 400 MHz) δ 7.16 (d, J = 8.3, 2H), 7.02 (d, 2H), 3.68 (s, 3H), 3.58 (s, 2H), 1.92-1.83 (m, 1H), 0.97-0.91 (m, 2H), 0.70-0.64 (m, 2H).
Step 3: Methyl 2-(4-cyclopropylphenyl)acetate (3.10 g, 16.30 mmol) was dissolved in a mixture of THF/MeOH/water , 80 mL), and the solution was treated with lithium hydroxide hydrate (0.8548 g, 20.37 mmol). The mixture was then stirred at ambient temperature for 4 hours. The reaction mixture was neutralized to a pH of 4 with 3N HCl and concentrated in vacuo. The solids were solved in ethyl acetate and water. The pH was re-adjusted to a pH of about 3 to about 4 with 3N HCl. The layers were then separated. The aqueous layer was washed with ethyl acetate (2 X). The combined organic layers were then washed with saturated NaCl, dried over Na2SO4 and concentrated to yield 2-(4- cyclopropylphenyl)acetic acid (2.82 g, 98%). 1H NMR (CDCl3, 400 MHz) δ 7.16 (d, J = 8.2, 2H), 7.03 (d, 2H), 3.60 (s, 2H), 1.92-1.83 (m, 1H), 098-0.91 (m, 2H), 0.70-0.64 (m, 2H).
Step 4: 2-(4-Cyclopropylphenyl)acetic acid (2.82 g, 16.003 mmol) was combined with (R)benzyloxazolidinone (3.4030 g, 19.204 mmol) in toluene (14 mL). The suspension was treated with triethylamine (6.6917 mL, 48.010 mmol) and then heated to 80°C. The solution was treated dropwise with a solution of pivaloyl chloride (1.9893 mL, 16.003 mmol) in toluene (3.5 mL). The on was heated overnight at 80°C. The reaction was cooled and washed with 2N HCl and then separated. The aqueous layer was washed with toluene, and the combined organics were then washed with 2N HCl, water, saturated NaHCO3 (2 X), saturated NaCl, dried over Na2SO4 and concentrated in vacuo. The crude product was ted to chromatography on SiO2 eluting with 9:1 hexanes/ethyl acetate to yield (R)benzyl(2-(4-cyclopropylphenyl)acetyl)oxazolidinone (3.43 g, 64%). 1H NMR (CDCl3, 400 MHz) δ 7.33-7.20 (m, 5H), 7.16-7.11 (m, 2H), 7.05 (d, J = 8.2, 2H), 4.70- 4.63 (m, 1H), 4.32-4.14 (m, 4H), 3.26 (dd, J1 = 3.2, J2 = 13.3, 1H), 2.75 (dd, J1 = 9.5, J2 = 13.3, 1H), 1.93-1.85 (m, 1H), 0.98-0.92 (m, 2H), 0.72-0.66 (m, 2H).
Step 5: ((S)(tert-Butoxycarbonyl)pyrrolidinyl)(4- cyclopropylphenyl)acetic acid was prepared according to the procedure described for Example 1, using (R)benzyl(2-(4-cyclopropylphenyl)acetyl)oxazolidinone (0.287 g, 26%). MS (ESI+) [M+H] 345.7.
Step 6: (S)-tert-Butyl 2-((S)(4-cyclopropylphenyl)(4-((5R,7R)hydroxy methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl)oxoethyl)pyrrolidine- oxylate was ed ing to the procedure described for Example 3 using (S) ((S)(tert-butoxycarbonyl)pyrrolidinyl)(4-cyclopropylphenyl)acetic acid, (0.199 g, 94%). MS (ESI+) [M+H] 562.1.
Step 7: (S)(4-Cyclopropylphenyl)(4-((5R,7R)hydroxymethyl-6,7- dihydro-5H-cyclopenta[d]pyrimidinyl)piperazinyl)((S)-pyrrolidinyl)ethanone was ed according to the procedure described for Example 3 using (S)-tert-butyl 2-((S)(4- cyclopropylphenyl)(4-((5R,7R)hydroxymethyl-6,7-dihydro-5H- cyclopenta[d]pyrimidinyl)piperazinyl)oxoethyl)pyrrolidinecarboxylate (0.145 g, 77%). MS (ESI+) [M+H] 462.2. 1H NMR (CD3OD, 400 MHz) δ 8.56 (s, 1H), 7.26 (d, 2H), 7.13 (d, 2H), 5.29 (dd, 1H), 5.32-5.26 (dd, 1H), 4.32 (d, 1H), 4.29-4.18 (m, 1H), 4.12-3.95 (m, 2H), 3.88-3.61 (m, 6H), 3.51-3.38 (m, 1H), 3.35-3.30 (m, 1H), 2.32-2.24 (m, 1H), 2.22- 2.03 (m, 2H), 1.95-1.85 (m, 2H), 1.82-1.73 (m, 2H), 1.40-1.34 (m, 1H), 1.16 (d, 3H), 1.01- 0.95 (m, 2H), 0.69-0.64 (m, 2H).
Examples shown in Table 2 can also be made according to the above described methods.
Table 2 Example Structure Name LCMS or 1H NMR m/z 461.3; 1H NMR (500 MHz, DMSOD6 ) d ppm 8.65 (s, 1H), 7.85 (d, 2H), O 4-((S)(4-((5R,7R)hydroxy 7.65 (d, 2H), 5.10 (t, N methyl-6,7-dihydro-5H- 1H), 4.80 (d, 1H), cyclopenta[d]pyrimidin 4.10-3.85 (m, 5H), 11 N yl)piperazinyl)((S) 3.68 (m, 2H), 3.40 N methylpyrrolidinyl) (m, 2H), 2.90 (s, oxoethyl)benzonitrile 3H), 2.20-2.02 (m, HO 2H), 1.93 (m, 2H), 1.68 (m, 1H), 1.50 (m, 1H),1.35-1.25 (m, 11H), 1.10 (d, 3H) m/z 490.3; 1H NMR (500 MHz, DMSOD6 ) d ppm 9.18 (m, 1H), 8.85 (m, 1H), 8.57 (s, 1H), 7.78 (d, O (S)(4-((5R,7R)hydroxy 2H), 7.62 (d, 2H), -6,7-dihydro-5H- N 5.04 (t, 1H), 4.48 (d, cyclopenta[d]pyrimidin 12 F3C 1H), 4.02 (m, 2H), N yl)piperazinyl)((S)-pyrrolidin 3.95 (m, 2H), 3.75- yl)(4- N 3.50 (m, 6H), 3.42 (trifluoromethyl)phenyl)ethanone (m, 2H), .10 HO (m, 4H), 2.10-1.90 (m 3H), 1.75 (m, 1H), 1.70-1.50 (m, 2H), 1.04 (d, 3H) LCMS (apci+) 502 [M+H]+; 2.68 min; HPLC r.t.= 1.98min, >97% purity; 1H NMR (400MHz, D2O) d ppm 8.37 (s, NH 1H), 7.43 (t, J= 8.2Hz, 1H), 7.16 (d, F O (S)(4-chlorofluorophenyl) J= 9.8Hz, 1H), 7.06 N ((S)-5,5-dimethylpyrrolidinyl) (d, J= 8.2Hz, 1H), 13 Cl (4-((5R,7R)hydroxymethyl-6,7- .24 (t, J= 7.8Hz, N dihydro-5H-cyclopenta[d]pyrimidin- 1H), 4.27 (d, J= iperazinyl)ethanone N 9.4Hz, 1H), 4.22-4.02 N (m, 1H), 3.88-3.75 HO (m, 2H), 3.72-3.60 (m, 1H), 3.59-3.41 (m, 4H0, 3.37-3.22 (m, 1H), 2.24-2.11 (m, 0.5H), 2.10-1.94 (m, 0.5H), 1.89-1.71 (m, 4H), 1.36 (s, 3H), 1.30 (s, 3H), 0.96 (d, J=7.0Hz, 3H) Example 14 In Vitro Cell proliferation Assays The in vitro potency of the ations of the compound of Example 2 with certain specific chemotherapeutic agents was measured using the CellTiter-Glo® Luminescent Cell Viability Assay, cially available from Promega Corp., n, WI. This homogeneous assay method is based on the recombinant expression of tera luciferase (US 5583024; US 5674713; US 5700670) and determines the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells (Crouch et al (1993) J. Immunol. Meth. 160:81-88; US 6602677). The CellTiter-Glo® Assay was conducted in 96 or 384 well , making it amenable to automated high-throughput screening (HTS) (Cree et al (1995) AntiCancer Drugs 6:398-404). The homogeneous assay procedure involves adding the single reagent (CellTiter-Glo® Reagent) directly to cells cultured in serum-supplemented . Cell washing, removal of medium and multiple pipetting steps are not required. The system detects as few as 15 cells/well in a ll format in 10 minutes after adding reagent and mixing.
The homogeneous "add-mix-measure" format results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in culture. The CellTiter-Glo® Assay generates a "glow-type" luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on cell type and medium used. Viable cells are reflected in ve luminescence units (RLU). The substrate, Beetle Luciferin, is oxidatively decarboxylated by recombinant firefly luciferase with itant conversion of ATP to AMP and generation of photons. The extended ife eliminates the need to use reagent ors and provides flexibility for uous or batch mode processing of multiple plates. This cell proliferation assay can be used with various multiwell formats, e.g., 96 or 384 well . Data can be recorded by luminometer or CCD camera imaging device. The luminescence output is presented as ve light units (RLU), measured over time.
The anti-proliferative effects of combinations of the compound of Example 2 and certain chemotherapeutic agents were measured using the CellTiter-Glo® Assay. EC50 values were established for the tested compounds and combinations. The range of in vitro cell potency activities was about 100 nM to about 10 µM. The data in Figure 12 demonstrates that representative ations provide additive or synergistic ty against a number of cancer types. e 15 In Vivo Tumor Xenograft Efficacy The cy of representative combinations (including those of the ion) may be measured in vivo by implanting allografts or xenografts of cancer cells in rodents and treating the tumor-bearing animals with the combinations. Variable results are to be expected depending on the cell line, the presence or absence of certain mutations in the tumor cells, the sequence of administration the compound of Example 2 and chemotherapeutic agent, dosing regimen, and other factors. Subject mice were treated with drug(s) or control (Vehicle) and monitored over l weeks or more to measure the time to tumor doubling, log cell kill, and tumor inhibition.
Results for representative combinations (including those of the invention) that were tested in this model are presented in the Figures.
The data in s demonstrates that representative combinations provide improved results compared to the administration of the respective agents individually. For example, in the LuCap35V primary human prostate tumor model the ation of Example 2 and docetaxel resulted in tumor regressions while the single agent of either compound only resulted in tumor stasis (Figure 1). Additionally, the ation of e 2 and cisplatin resulted in greater tumor growth inhibition than either single agent alone in the SKOV3 ovarian human tumor model (Figure 8).
It has been determined that certain combinations ding those of the invention) provide improved effects against certain cancer ypes. For example, certain combinations (including those of the invention) provide improved effects against cancers associated with PTEN mutation, AKT mutation (e.g., overexpression or amplification), PI3K mutation, or Her2/ErbB2 amplification or mutation. Accordingly, certain combinations described herein may be particularly useful against these types of cancers. For example, in gastric cancer, PTEN-loss ts better efficacy with certain combinations ding those of the invention) (e.g., a compound of formula I with 5-FU/cisplatin), and in prostate cancer a stronger effect was seen for a combination of a compound of formula I and docetaxel in PTEN-null lines.
PTEN status may be measured by any suitable means as is known in the art. In one example, IHC is used. atively, Western blot analysis can be used. Antibodies to PTEN are commercially available (Cell Signaling Technology, Beverly, MA, Cascade Biosciences, Winchester, MA). Example procedures for IHC and Western blot analysis for PTEN status are described in Neshat, M. S. et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR, Proc. Natl Acad. Sci. USA 98, 10314–10319 (2001) and , A., et. al. Immunohistochemical Evidence of Loss of PTEN Expression in Primary Ductal Adenocarcinomas of the Breast, American l of ogy, Vol. 155, No. 4, October 1999. Additionally, cancers associated with AKT mutation, PI3K mutation, and with Her2/ErbB2 amplification or mutation can be identified using techniques that are known in the art. In one example, PTEN status of a patient or tissue sample is determined using IHC, and a histo score or HScore is assigned to the sample or patient. An e way of calculating HScore uses the formula: HScore = lls x 1)+(%2+cells x 2)+(%3+cells x 3) (See Shoman, N, et. al, Mod Path (2005) 18, 250-259). A mean PTEN HScore of noncancerous tissue from the same patient or a collection of patients can be used to determine whether patient or sample HScores are low or null. In one example, HScores of less than about 200 are considered low and pond to PTEN low, and HScores of about 0 are considered null.
One embodiment described is a method of tumor growth inhibition (TGI) in a patient suffering from a cancer comprising a PTEN mutation, AKT mutation (e.g., overexpression or amplification), PI3K mutation, or Her2/ErbB2 amplification or mutation, sing administering GDC-0068 or a pharmaceutically acceptable salt thereof and one of Folfox, a platinum agent, irinotecan, docetaxel, bicin, gemcitabine, SN-38, capecitabine, lomide, paclitaxel, bevacizumab, pertuzumab, tamoxifen, rapamycin and lapatinib or a pharmaceutically acceptable salt thereof to the patient. In n embodiments, the ation is synergistic. In certain embodiments, the TGI of the combination is greater than the TGI of either GDC-0068 or the chemotherapeutic agent alone. In certain ments, the TGI of the combination is about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70 or 75 percent greater than the TGI of either GDC-0068 or chemotherapeutic agent alone.
Methods of measuring TGI are known in the art. In one example method, average tumor volumes are determined and compared from the patient before and after treatment.
Tumor volumes can be measured in two dimensions (length and width) using any method in the art, for example UltraCal IV calipers (Fred V. Fowler Company) or by PET (positron emission tomography), or by some other method. The formula tumor volume (mm3) = h x width2) x 0.5 can be used. Measuring tumor volumes over multiple time periods can be done using a mixed-modeling Linear Mixed Effects (LME) approach (Pinheiro et al. 2009).
This ch can address both repeated measurements (and multiple patients). Cubic regression splines can be used to fit a non-linear e to the time courses of tumor volume at each dose level. These non-linear profiles can then be related to dose within the mixed model. Tumor growth tion as a percent of vehicle can be calculated as a percent area under the fitted curve (AUC) per day in relation to the vehicle, using the following formula: Using this formula, a TGI value of 100% indicates tumor stasis, r than about 1% but less than about 100% indicates tumor growth tion, and greater than about 100% indicates tumor regression.
In certain embodiments, the cancer comprises one or more of AKT, PI3k, PTEN and HER2 mutations or AKT, PI3k, PTEN or HER2 abberant signaling. In one example, the cancer is gastric cancer comprising high pAKT activity and PTEN low or null status.
Described is a method for treating a patient having a cancer that is associated with PTEN on or loss of expression, AKT on or amplification, PI3K mutation or amplification, or rbB2 amplification sing administering a combination of the invention to the patient. Also described is a method for identifying a patient having a cancer that that can be treated with a combination described comprising ining if the patient’s cancer is associated with PTEN mutation or loss of sion, AKT mutation or amplification, PI3K mutation or amplification, or Her2/ErbB2 amplification, wherein association of the patient’s cancer with PTEN mutation or loss of expression, AKT mutation or amplification, PI3K mutation or amplification, or Her2/ErbB2 ication is indicative of a cancer that can be treated with a combination (including those of the invention). Also described is a method further comprising treating the patient so identified with a combination described.
In another example, the cancer to be treated is associated with PTEN positive, low or null status in combination with HER2 positive or negative status. Examples include gastric cancer that is either (i) PTEN negative (HScore less than about 10, or 0) and Her2 negative, (ii) PTEN low (HScore less than about 200) and Her2 negative, (iii) PTEN negative and Her2 positive, or (iv) PTEN positive and Her2 ve. In this example, the cancer can be treated with a combination of a formula I compound, e.g., GDC-0068 or a salt thereor, and FOLFOX.
Example 16 Human dosing of GDC-0068 Patients with advanced or metastatic solid tumors were administered a hydrochloride salt of GDC-0068 orally and the safety, tolerability and response were assessed using, for e, PET scans and the incidence and nature of dose limiting toxicities (DLTs). Patients ed doses of 25 (n=3), 50 (n=3), 100 (n=3), 200 (n=3), 400 (n=3), 600 (n=8) and 800 (n=7) mg 68. No DLTs were observed at the 25, 50, 100, 200, 400 or 600 mg doses.
Grade 3 fatigue was observed at the 800 mg dose in one patient. All 3 patients at the 400 mg dose had greater than about 60% inhibition in the PRAS40 levels (downstream readout of AKT ing) as measured by IHC or RPPA assay.
Patients suffering from either tion resistant prostate cancer (n=10) or diagnostically positive metastatic breast cancer (having one or more of PTEN low or null status, PI3k mutation or AKT mutation or increased expression or ty, n=10) were administered GDC-0068 orally, once daily for first seven days of a 21 day cycle at doses of 600mg. Figure 21 shows the PET scan responses for the breast cancer patients. Figure 22 shows PET and tumor marker response in Patient 1 of Figure 21, who suffered from HER2-, PI3K mutant (H1047R) breast cancer. Patient 6 of Figure 24, who suffered from AKT mutant E17K (PI3K wild type and HScore of 240) breast cancer, had a complete response after cycle #1 on the PET scan, with all target and non-target lesions PET-negative, and no new lesions were seen. These responses trate that compounds of formula I, e.g., GDC-0068, treat patient’s roliferative diseases.
Surrogate PD ker assays o-GSK-3b in Platelet rich plasma (PRP) was used as a surrogate PD biomarker to measure Akt pathway inhibition in patients after treatment with GDC-0068 at different time points. Peripheral blood was collected in Vacutainer ning .38% of citrate as anti- ant. Blood was spun at 200 g for 15 min at room temperature. The PRP layer was carefully taken from the tube and then lysed in a buffer containing detergents, protease and phosphatase inhibitors. Phosphorylated and total GSK-3β levels in PRP lysates were measured using a phospho-GSK3 β /total-GSK3β multiplexed MSD assay. pGSk-3β levels were normalized to total GSk-3β levels and post-dose inhibition of pGSk-3β was expressed as a ratio of the pre-dose levels for each t. A dose- and time-dependent pharmacologic response was demonstrated, with a decrease in pGSK3β level of ≥ 75% at doses ≥ 200 mg.
Reverse Phase Protein Arrays (RPPA assay) Core-needle tumor biopsies from patients treated with GDCC0068 were fresh-frozen in OCT and sectioned into 8 um slices. Tissue was lysed in RPPA lysis buffer containing TPER, 300 mM NaCl and phosphatase inhibitors. oprotein signatures of the lysates were analysed using Reverse-Phase protein arrays: s were printed on nitrocellulose slides and stained with Sypro to determine total protein concentrations. Each slide was d with a different antibody at 4oC overnight. The data was then normalized to total n levels and spatial effects were removed using Quadrant median normalization.
Decreases of 60%–70% in pPRAS40 and ~50% decrease in Cyclin D1 (compared with baseline) ed in all 3 patients treated at 400 mg daily. For methods and overview of RPPA see: Reverse phase protein microarrays advance to use in al trials, Molecular gy. 2010 Dec;4(6):461-81, Mueller C et al.
Example 17 Human dosing of 68 in combination with Docetaxel A 21-day treatment period was run over multiple cycles. Patients with advanced or metastatic solid tumors were administered a hydrochloride salt of GDC-0068 orally, once daily, on Days 2 through 15 of all cycles, and docetaxel, 75 mg/m2 IV infusion through a vein was given over 1 hr on Day 1. Patients were separated into cohorts. Cohort 1 received 100 mg dose of GDC-0068. Cohort 2 received 200 mg, Cohort 3 received 400 mg and Cohort 4 received 600 mg of GDC-0068, respectively. Disease status was ed using Response Evaluation ia in Solid Tumors, Version 1.1 (RECIST v1.1). The safety and tolerability were assessed using the incidence and nature of dose limiting toxicities (DLTs) and the incidence, nature, and severity of adverse events and laboratory abnormalities (graded per NCI CTCAE v4.03). No DLTs were observed in Cohorts 1, 2 or 3.
Figure 23 shows results of one patient with l response in Akt1 E17K mutation breast cancer patient. The patient received three courses of prior chemotherapy but failed on all three. The t was given GDC-0068 on day 2 of the first cycle for about 15 days, once daily orally, after a treatment of docetaxel on day 1. No therapy was given for the next 28 days. Prior to the combination treatment (at screening), the patient’s tumor was 30.2 by 17.9 mm, and after the first cycle of the combination treatment, the patient’s tumor shrunk to 18.2 by 16.0 mm (a 39% decrease or PR). These responses demonstrate that compounds of formula I, e.g., 68, in combination with chemotherapeutic agents, e.g., docetaxel, treat patient’s hyperproliferative diseases, and can treat the diseases after prior treatments fail.
Example 18 Human dosing of GDC-0068 in combination with 5-FU, orin and oxaliplatin (FOLFOX) A 14-day treatment period was run over multiple cycles. Patients with advanced or metastatic solid tumors were administered escalating doses of a hydrochloride salt of GDC- 0068 orally, once daily, on Days 1 through 7 of all cycles, and mFOLFOX6 (Oxaliplatin 85 mg/m 2, orin 400 mg/m2 IV over 2 hr, and 5-fluorouracil 400 mg/m2 IV injection al bolus) and 5-fluorouracil 2400 mg/m2 IV over 46 hr) were administered as IV infusions through a vein on Day 1 of each 14-day cycle. Cohort 1 received 100 mg dose of GDC-0068. Cohort 2 received 200 mg and Cohort 3 received 400 mg of GDC-0068, respectively. Disease status will be assessed using Response Evaluation Criteria in Solid Tumors, Version 1.1 (RECIST v1.1). The safety and bility were assessed using the incidence and nature of dose limiting toxicities (DLTs) and the incidence, nature, and severity of adverse events and tory abnormalities (graded per NCI CTCAE v4.03). No DLTs were observed in Cohorts 1 or 2.
Figure 24 shows results of one patient with l response in PIK3CA mutant squamous carcinoma of . The patient received the above combination therapy. Prior to the combination treatment (at screening), the patient’s tumor was 22 mm, and after week 8 of the above ation treatment, the patient’s tumor shrunk to 13.1 mm (a 40% decrease or PR).
Figure 25 shows results of a treatment of GDC-0068 in ation with FOLFOX with partial response where patient suffered from oss (Hscore 40), KRAS-Wild-Type Colorectal Cancer, after failing prior treatments.
These responses demonstrate that compounds of formula I, e.g., GDC-0068, in combination with chemotherapeutic agents, e.g., docetaxel, treat patient’s hyperproliferative diseases, and can treat the diseases after prior treatments fail.
Further, since numerous modifications and changes will be readily apparent to those skilled in the art, it is not desired to limit the invention to the exact construction and process shown as bed above. Accordingly, all suitable modifications and equivalents may be considered to fall within the scope as d by the claims that follow.

Claims (59)

We claim:
1. The combination of a compound of a Ia 5 (Ia) or a pharmaceutically acceptable salt thereof and one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof.
2. The combination of claim 1, for use in the therapeutic treatment of a 10 hyperproliferative disorder.
3. The combination of claim 2, wherein the hyperproliferative disorder is cancer.
4. The combination of claim 3, wherein the cancer is associated with PTEN mutation.
5. The combination of claim 4, wherein the cancer is associated with AKT mutation, overexpression or amplification.
6. The combination of claim 4, wherein the cancer is ated with PI3K mutation.
7. The ation of claim 4, wherein the cancer is ated with Her2/ErbB2 mutation or amplification.
8. The combination of any one of claims 4-7, wherein the cancer is selected from breast, 25 lung, ovarian, prostate, melanoma, c, colon, renal, head and neck, and glioma cancers.
9. The combination of any one of claims 4-8, wherein the cancer is breast cancer.
10. The combination of any one of claims 4-9, wherein the combination further comprises oxaliplatin.
11. The combination of any one of claims 4-9, wherein the combination further comprises leucovorin.
12. The combination of claim 10 or claim 11, wherein the cancer is c, ovarian or 10 colon cancer.
13. The combination of any one of claims 1-12, wherein the combination of the compound of formula Ia and an agent selected from 5-FU, capecitabine or a pharmaceutically able salt thereof provides a synergistic effect in treating the hyperproliferative disorder.
14. The combination of claim 13, n Combination Index value of the synergistic effect is less than about 0.8.
15. The combination of claim 1, for improving the quality of life of a patient treated for a 20 hyperproliferative disorder.
16. The combination of claim 1, for use in the treatment of a hyperproliferative disorder in a mammal. 25
17. The combination of claim 1, for use in the treatment of elioma, endometrial, breast, lung, ovarian, prostate, pancreatic, melanoma, c, colon, glioma, or head and neck cancer in a mammal.
18. The combination of claim 17, wherein the te cancer is castration resistant 30 prostate cancer (CRPC).
19. The combination of claim 1, for use in the treatment or prevention of herapyresistant cancer.
20. A kit comprising a compound of formula Ia (Ia) or a ceutically acceptable salt thereof; and one or more of 5-FU and capecitabine or a 5 pharmaceutically acceptable salt thereof.
21. A kit of claim 20, further comprising a package insert or label indicating the administration of the compound of formula Ia, or a ceutically acceptable salt thereof, one or more of 5-FU and capecitabine or a pharmaceutically acceptable salt thereof.
22. A product comprising a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof; and one or more of 5-FU and capecitabine or a 15 pharmaceutically acceptable salt thereof, as a combined preparation for simultaneous, separate or sequential use in the therapeutic ent of a hyperproliferative disorder.
23. Use of a nd of formula Ia: (Ia) or a pharmaceutically acceptable salt thereof and one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a 5 medicament for the therapeutic treatment of a hyperproliferative disorder.
24. Use of claim 23, wherein the medicament is in a form for aneous, sequential or separate administration of the compound of formula Ia or a ceutically acceptable salt thereof, and the one or more agents.
25. Use of claim 23, wherein two medicaments are prepared, one comprising the compound of formula Ia or a pharmaceutically acceptable salt thereof, and the other comprising the one or more agents. 15
26. The use of a compound of formula Ia (Ia) or a pharmaceutically able salt thereof, in the cture of a ment for therapeutically treating a hyperproliferative disorder in a subject, in combination with one or 20 more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof.
27. The use of claim 26, wherein the medicament is in a form for simultaneous administration with the one or more agents.
28. The use of claim 26, wherein the medicament when administered, is administered simultaneously with the one or more agents.
29. The use of claim 26, wherein the medicament is in a form for sequential 10 administration with the one or more agents.
30. The use of claim 26, wherein the medicament when administered, is administered sequentially with the one or more agents. 15
31. The use of claim 26, wherein the ment is in a form for separate stration with the one or more .
32. The use of claim 26, wherein the medicament when administered, is administered separately with the one or more agents.
33. The use of any one of claims 23-32, wherein the hyperproliferative disorder is cancer.
34. The use of claim 33, wherein the cancer is ated with PTEN mutation. 25
35. The use of claim 33, wherein the cancer is associated with AKT mutation, overexpression or amplification.
36. The use of claim 33, wherein the cancer is associated with PI3K mutation. 30
37. The use of claim 33, wherein the cancer is associated with Her2/ErbB2 mutation or amplification.
38. The use of any one of claims 33-37, wherein the cancer is selected from, breast, lung, ovarian, prostate, melanoma, c, colon, renal, head and neck, and glioma cancers.
39. The use of any one of claims 33-38, wherein the cancer is breast cancer.
40. The use of any one of claims 33-39,wherein the medicament r comprises 5 oxaliplatin.
41. The use of any one of claims 33-39, further comprising the use of oxaliplatin.
42. The use of any one of claims 33-39, wherein the medicament is in a form for, is to be 10 administered, or when administered, is administered with oxaliplatin.
43. The use of any one of claims 33-39, wherein the medicament r comprises leucovorin. 15
44. The use of any one of claims 33-39, further comprising the use of leucovorin.
45. The use of any one of claims 33-39, wherein the medicament is in a form for, is to be administered, or when administered, is administered with leucovorin. 20
46. The use of any one of claims 40 to 45, wherein the cancer is c, ovarian, or colon
47. The use of any one of claims 23-46, wherein the combination of the compound of Formula Ia and an agent selected from 5-FU, capecitabine, or a pharmaceutically acceptable 25 salt thereof provides a istic effect in treating the hyperproliferative disorder.
48. The use of claim 47, wherein Combination Index value of the synergistic effect is less than about 0.8. 30
49. Use of a compound of formula Ia: (Ia) or a pharmaceutically acceptable salt thereof, and an agent selected from 5-FU, and 5 capecitabine, or a pharmaceutically able salt thereof, in the manufacture of a ment for the therapeutic use for improving the quality of life of a patient treated for a hyperproliferative disorder.
50. Use of a compound of formula Ia 10 HO (Ia) or a ceutically acceptable salt thereof, and one or more agents selected from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a 15 medicament for use in the therapeutic treatment of a hyperproliferative disorder in a mammal.
51. Use of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof, and one or more agents selected from 5-FU, 5 and tabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of mesothelioma, trial, breast, lung, ovarian, prostate, pancreatic, melanoma, gastric, colon, glioma, or head and neck cancer in a mammal.
52. The use of claim 51 wherein the prostate cancer is castration resistant prostate cancer 10 (CRPC).
53. Use of a compound of formula Ia (Ia) or a pharmaceutically acceptable salt thereof, and one or more agents ed from 5-FU, and capecitabine, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment or prevention of herapy-resistant cancer. 20
54. The use of any one of claims 50 to 53, wherein the medicament is in a form for simultaneous, sequential or separate administration of the nd of formula Ia, or a ceutically acceptable salt thereof, and the one or more agents.
55. The use of any one of claims 50 to 53, wherein two medicaments are prepared, one 5 comprising the compound of formula Ia or a pharmaceutically acceptable salt thereof, and the other comprising the one or more agents.
56. The combination as claimed in any one of claims 1 to 29, ntially as herein described with reference to any example thereof.
57. A kit as claimed in claim 20 or claim 21, substantially as herein described with reference to any example thereof.
58. A product as claimed in claim 23, substantially as herein described with reference to 15 any example thereof.
59. A use as claimed in any one of claims 23 to 55, substantially as herein described with reference to any e thereof. WO 35781
NZ617245A 2011-04-01 2012-03-30 Combinations of akt inhibitor compounds and chemotherapeutic agents, and methods of use NZ617245B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161470624P 2011-04-01 2011-04-01
US201161470803P 2011-04-01 2011-04-01
US61/470,803 2011-04-01
US61/470,624 2011-04-01
PCT/US2012/031720 WO2012135781A1 (en) 2011-04-01 2012-03-30 Combinations of akt inhibitor compounds and chemotherapeutic agents, and methods of use

Publications (2)

Publication Number Publication Date
NZ617245A NZ617245A (en) 2016-02-26
NZ617245B2 true NZ617245B2 (en) 2016-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
US10092567B2 (en) Combinations of AKT inhibitor compounds and chemotherapeutic agents, and methods of use
NZ617245B2 (en) Combinations of akt inhibitor compounds and chemotherapeutic agents, and methods of use
NZ617249B2 (en) Combinations of akt inhibitor compounds and vemurafenib, and methods of use
NZ617238B2 (en) Combinations of akt inhibitor compounds and abiraterone, and methods of use
NZ617246B2 (en) Combinations of akt inhibitor compounds and erlotinib, and methods of use