US20180339997A1 - Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production - Google Patents

Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production Download PDF

Info

Publication number
US20180339997A1
US20180339997A1 US15/963,876 US201815963876A US2018339997A1 US 20180339997 A1 US20180339997 A1 US 20180339997A1 US 201815963876 A US201815963876 A US 201815963876A US 2018339997 A1 US2018339997 A1 US 2018339997A1
Authority
US
United States
Prior art keywords
polymorph
crystalline
gdc
ray powder
theta
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/963,876
Other languages
English (en)
Inventor
Paroma Chakravarty
Chong Han
Sean M. Kelly
Karthik Nagapudi
Scott Savage
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
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 to US15/963,876 priority Critical patent/US20180339997A1/en
Publication of US20180339997A1 publication Critical patent/US20180339997A1/en
Priority to US16/875,545 priority patent/US11591345B2/en
Priority to US16/875,537 priority patent/US11028100B2/en
Priority to US18/101,951 priority patent/US20230167128A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the invention relates to polymorph forms of a PI3K inhibitor compound GDC-0077, named as (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide.
  • the invention also relates to processes to obtain polymorph forms of GDC-0077.
  • Phosphoinositide 3-kinases are lipid kinases that phosphorylate lipids at the 3-hydroxyl residue of an inositol ring (Whitman et al (1988) Nature, 332:664).
  • the 3-phosphorylated phospholipids (PIP3s) generated by PI3-kinases act as second messengers recruiting kinases with lipid binding domains (including plekstrin homology (PH) regions), such as Akt and phosphoinositide-dependent kinase-1 (PDK1). Binding of Akt to membrane PIP3s causes the translocation of Akt to the plasma membrane, bringing Akt into contact with PDK1, which is responsible for activating Akt.
  • the PI3-kinases Akt and PDK1 are important in the regulation of many cellular processes including cell cycle regulation, proliferation, survival, apoptosis and motility and are significant components of the molecular mechanisms of diseases such as cancer, diabetes and immune inflammation (Vivanco et al (2002) Nature Rev. Cancer 2:489; Phillips et al (1998) Cancer 83:41).
  • the main PI3-kinase isoform in cancer is the Class I PI3-kinase, p110 ⁇ (alpha) (U.S. Pat. No. 5,824,492; U.S. Pat. No. 5,846,824; U.S. Pat. No. 6,274,327).
  • GDC-0077 also known by the IUPAC name: (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, has potent PI3K activity (WO 2017/001645, US 2017/0015678, Edgar K. et al, #156, “Preclinical characterization of GDC-0077, a specific PI3K alpha inhibitor in early clinical development”, and Staben. S.
  • Powder X-ray Diffraction is a powerful tool in identifying different crystal phases by their unique diffraction patterns
  • Polymorphism is often characterized as the ability of a drug substance, i.e. Active Pharmaceutical Ingredient (API), to exist as two or more crystalline phases that have different arrangements and/or conformations of the molecules in the crystal lattices giving the crystals different physicochemical properties.
  • API Active Pharmaceutical Ingredient
  • Crystal structure determination at the atomic level and intermolecular interactions offer important information to establish absolute configuration (enantiomers), phase identification, quality control, and process development control and optimization.
  • X-ray Diffraction is widely recognized as a reliable tool for the crystal structure analysis of pharmaceutical solids and crystal form identification.
  • the crystal structure can be solved from X-ray powder diffraction data obtained by measurements at ambient conditions and/or at variable temperature or humidity.
  • the invention relates to polymorph forms of the PI3K inhibitor GDC-0077 (CAS Registry Number 2060571-02-8, Genentech, Inc.), named as (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, having the structure, Formula I:
  • An aspect of the invention is a pharmaceutical composition of a polymorph form of GDC-0077.
  • An aspect of the invention is a method of treating a hyperproliferative disorder in a mammal with a polymorph form of GDC-0077.
  • An aspect of the invention is a process for preparing a crystalline polymorph of GDC-0077.
  • An aspect of the invention is the crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form A polymorph that exhibits an X-ray powder diffraction pattern having a characteristic peak expressed in degrees 2-theta at approximately 5.7.
  • the Form A polymorph exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, and 19.0. In some embodiments, the Form A polymorph exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, 17.2, 19.0, 19.7, and 24.4.
  • An aspect of the invention is the crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form A polymorph that exhibits an X-ray powder diffraction pattern obtained using an incident beam of Cu K ⁇ radiation having a characteristic peak expressed in degrees 2-theta at approximately 5.7; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, and 19.0; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, 17.2, 19.0, 19.7, and 24.4.
  • An aspect of the invention is the crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form A polymorph that exhibits an X-ray powder diffraction pattern obtained using an incident beam of Cu K ⁇ (1.541904 ⁇ ) radiation generated using Cross Beam optics (40 kV ⁇ 44 mA) having a characteristic peak expressed in degrees 2-theta at approximately 5.7; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, and 19.0; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, 17.2, 19.0, 19.7, and 24.4.
  • Form A polymorph that exhibits an X-ray powder diffraction pattern obtained using an incident beam of Cu K ⁇ (1.54190
  • An aspect of the invention is the Form A polymorph as described herein characterized by the X-ray powder diffraction pattern substantially as shown in FIG. 4 .
  • An aspect of the invention is the Form A polymorph as described herein characterized by the X-ray powder diffraction peaks shown in Table 2.
  • An aspect of the invention is the Form A polymorph as described wherein a differential scanning calorimetry DSC shows a melting endotherm at approximately 212 to 215° C.
  • An aspect of the invention is the Form A polymorph as described wherein a differential scanning calorimetry DSC shows a melting endotherm at approximately 214° C.
  • An aspect of the invention is the Form A polymorph as described herein characterized by the 13 C SSNMR (solid-state nuclear magnetic resonance) spectra substantially as shown in FIG. 7A .
  • An aspect of the invention is the Form A polymorph as described herein characterized by the 19 F SSNMR (solid-state nuclear magnetic resonance) spectra substantially as shown in FIG. 7B .
  • An aspect of the invention is a crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form D polymorph that exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 10.8, 16.8, and 20.4.
  • the Form D polymorph exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 8.6, 10.8, 16.8, 19.2, and 20.4.
  • the Form D polymorph as described herein is characterized by the X-ray powder diffraction pattern shown in FIG. 15A . In some embodiments, the Form D polymorph as described herein is characterized by the X-ray powder diffraction peaks shown in Table 3.
  • An aspect of the invention is a crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form D polymorph that exhibits an X-ray powder diffraction pattern obtained using an incident beam of Cu K ⁇ radiation having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 10.8, 16.8, and 20.4; or having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 8.6, 10.8, 16.8, 19.2, and 20.4.
  • An aspect of the invention is a crystalline, anhydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form D polymorph that exhibits an X-ray powder diffraction pattern using an incident beam of Cu K ⁇ (1.541904 ⁇ ) radiation generated using Cross Beam optics (40 kV ⁇ 44 mA) having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 10.8, 16.8, and 20.4; or having characteristic peaks expressed in degrees 2-theta at approximately 7.5, 8.6, 10.8, 16.8, 19.2, and 20.4.
  • An aspect of the invention is a crystalline, trihydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form B polymorph that exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, and 25.2.
  • the Form B polymorph exhibits an X-ray powder diffraction pattern having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, 19.5, 20.1, 21.6, and 25.2.
  • the Form B polymorph as described herein is characterized by the X-ray powder diffraction pattern shown in FIG. 12C . In some embodiments, the Form B polymorph as described herein is characterized by the X-ray powder diffraction peaks shown in Table 2A.
  • An aspect of the invention is a crystalline, trihydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form B polymorph that exhibits an X-ray powder diffraction pattern using an incident beam of Cu K ⁇ radiation having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, and 25.2; or having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, 19.5, 20.1, 21.6, and 25.2.
  • An aspect of the invention is a crystalline, trihydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide designated the Form B polymorph that exhibits an X-ray powder diffraction pattern using an incident beam of Cu K ⁇ (1.541904 ⁇ ) radiation generated using Cross Beam optics (40 kV ⁇ 44 mA) having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, and 25.2; or having characteristic peaks expressed in degrees 2-theta at approximately 5.4, 10.5, 19.5, 20.1, 21.6, and 25.2.
  • An aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the crystalline, anhydrate polymorph of Form A as described above, and a pharmaceutically acceptable carrier, glidant, diluent, or excipient.
  • An aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the crystalline, anhydrate polymorph of Form D as described above, and a pharmaceutically acceptable carrier, glidant, diluent, or excipient.
  • An aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the crystalline, trihydrate polymorph of Form B as described above, and a pharmaceutically acceptable carrier, glidant, diluent, or excipient.
  • An aspect of the invention is the pharmaceutical composition as described above in the form of a tablet.
  • An aspect of the invention is the pharmaceutical composition as described above wherein the therapeutically effective amount is from about 1 to about 100 mg.
  • An aspect of the invention is the pharmaceutical composition as described above wherein the crystalline, anhydrate or trihydrate polymorph is milled.
  • An aspect of the invention is the process for preparing a crystalline polymorph comprising heating a slurry of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide in ethanol (with or without water) or n-propanol (with or without water), and then cooling the mixture whereby a Form A crystalline polymorph that exhibits an X-ray powder diffraction pattern having a characteristic peak expressed in degrees 2-theta at approximately 5.7; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, and 19.0; or having characteristic peaks expressed in degrees 2-theta at approximately 5.7, 11.4, 17.2, 19.0, 19.7, and 24.4, is formed.
  • the process comprises heating a slurry of GDC-0077 in ethanol in the presence of less than 40% (or less than 20%, or less than 10%) of water, and then cooling the mixture whereby forming the Form A polymorph.
  • the process further comprises seeding the mixture with crystalline GDC-0077 (e.g., a crystalline THF solvate).
  • An aspect of the invention is the process for preparing a crystalline trihydrate polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide comprising slurring (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide in water (e.g., DI water).
  • water e.g., DI water
  • the process comprises slurring a Form A polymorph of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide in DI water for 4 days at room temperature.
  • An aspect of the invention is a method for the treatment of cancer in a subject in need thereof comprising administering to the subject an effective amount of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), or a pharmaceutical composition comprising a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), and a pharmaceutically acceptable carrier, glidant, diluent, or excipient.
  • the cancer is a HR-positive and HER2-negative breast cancel expressing a PIK3CA mutation.
  • the method further comprises one or more additional therapeutic agents (e.g., fulvestrant, palbociclib and/or letrozole).
  • An aspect of the invention is a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), or a pharmaceutical composition comprising a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), for use in the treatment of cancer.
  • the cancer is a HR-positive and HER2-negative breast cancel expressing a PIK3CA mutation.
  • the polymorphs for use further comprise one or more additional therapeutic agents (e.g., fulvestrant, palbociclib and/or letrozole).
  • An aspect of the invention is the use of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), or a pharmaceutical composition comprising a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), in the manufacture of a medicament for use in the treatment of cancer.
  • the cancer is a HR-positive and HER2-negative breast cancel expressing a PIK3CA mutation.
  • the uses further comprise one or more additional therapeutic agents (e.g., fulvestrant, palbociclib and/or letrozole).
  • FIG. 1 shows XRPD pattern of starting material GDC-0077.
  • FIG. 2 shows DSC and TGA traces of starting material GDC-0077.
  • the desolvation/vaporization endotherm and the recrystallization endotherm occur at 64 and 141° C. respectively (onset), followed by the melting endotherm at 214° C.
  • TGA shows a weight loss of ⁇ 2.5% w/w before the melting event.
  • FIG. 3 shows overlay of the XRPD patterns of the different solid form hits, Forms 1-VI, obtained in the 96 well HTS polymorph screening for GDC-0077.
  • FIG. 4 shows XRPD of anhydrous Form I (Form A) GDC-0077.
  • FIG. 5A shows SEM image at 1000 ⁇ magnification (benchtop Phenom SEM (Nanoscience Instruments, Inc., AZ) of anhydrous Form I (Form A) GDC-0077.
  • FIG. 5B shows PLM image (Leica DM 4000B microscope equipped with a high resolution CCD camera and motorized stage (Clemex Technologies Inc., Longueuil, Quebec, Canada) at 200 ⁇ magnification) of anhydrous Form I (Form A) GDC-0077.
  • FIG. 6A shows Thermal analysis of anhydrous Form I (Form A) GDC-0077.
  • FIG. 6B shows TGA of Forms IV-VI.
  • FIG. 6C shows DSC of Forms IV-VI. Multiple transitions, attributable to desolvation, formation of metastable form and subsequent conversion to Form A and its melting, were observed in the DSC traces and have been indicated.
  • FIG. 6D shows XRPD of product phase obtained upon heating Form B (trihydrate) to 195° C., with Form A pattern included for comparison. As indicated, the trihydrate ultimately converts to anhydrous Form A at this temperature.
  • FIG. 7A shows 13C SSNMR (solid-state nuclear magnetic resonance) spectra of anhydrous Form I (Form A) GDC-0077.
  • FIG. 7B shows 19F SSNMR of anhydrous Form I (Form A) GDC-0077.
  • FIG. 8 shows Water sorption behavior of anhydrous Form I (Form A) GDC-0077.
  • FIG. 9 shows Isothermal TGA traces of trihydrate Form B GDC-0077 before and after equilibration at RT at 60° C.
  • FIG. 10A shows Water sorption behavior of trihydrate Form B GDC-0077 at 25° C.
  • FIG. 10B shows DSC and TGA of Form III.
  • FIG. 11 shows overlay of the XRPD patterns of Form A and Form III solid forms.
  • XRPD patterns of Form III at RT, and when heated to 165 and 195° C. are shown. At higher temperatures (>165 C), Form III/C converts to Form I/A.
  • FIG. 12A shows anhydrate (Form A) to hydrate (Form B) conversion upon slurring in DI water for 4 days. Conversion starts within 12 hrs as indicated by hydrate marker peak (*) appearing in the anhydrate XRPD pattern. Form conversion is complete by 96 hrs. XRPD pattern of Form B is included for reference.
  • FIG. 12B shows Slurry bridging experiment data for GDC-0077 hydrate-anhydrate system in ethanol-water mixtures at RT (room temperature).
  • the equilibrium RH (relative humidity) zone for the two forms was identified as 82-86%, equivalent to 65-83% w/w water content.
  • FIG. 12C shows XRPD of trihydrate Form B GDC-0077.
  • FIG. 13A shows XRPD of GDC-0077 THF solvate.
  • FIG. 13B shows Thermal analysis of GDC-0077 THF solvate
  • FIG. 13C shows overlay of the XRPD patterns of THF solvate (RT), heated to 175 and 210° C. and Form A. The solvate desolvates to intermediate anhydrous Form which ultimately converts to Form A
  • FIG. 14 shows Thermal analysis of Form D, a second anhydrous form obtained by desolvating THF solvate (polymorph of Form A). The phase transitions are marked against the respective endotherms.
  • FIG. 15A shows XRPD of anhydrous Form D GDC-0077.
  • FIG. 15B shows overlay of the XRPD patterns of Forms A, D and final solid form of GDC-0077 obtained after slurrying Forms A and D (1:1 mixture) overnight in n-propanol (RT).
  • Form D converts to Form A in the slurry.
  • FIG. 16A shows PLM of milled GDC-0077. Form A remains stable upon milling.
  • FIG. 16B shows DSC and MDSC traces of GDC-0077 milled lot. Milling induces disorder which is evidenced by the emergence of an exotherm (marked in inset at 113° C.) that indicated recrystallization of the disordered phase and subsequent melting of Form A (endotherm at 214° C.). MDSC does not reveal a Tg near the exotherm.
  • FIG. 17 shows the Solid form landscape for GDC-0077.
  • the term “about” when used in reference to x-ray powder diffraction pattern peak positions refers to the inherent variability of the peaks depending on, for example, the calibration of the equipment used, the process used to produce the polymorph, the age of the crystallized material and the like, depending on the instrumentation used. In this case the measure variability of the instrument was about + ⁇ 0.0.2 degrees 2-theta ( ⁇ ). A person skilled in the art, having the benefit of this disclosure, would understand the use of “about” in this context.
  • the term “about” in reference to other defined parameters, e.g., water content, C max , t max , AUC, intrinsic dissolution rates, temperature, and time indicates the inherent variability in, for example, measuring the parameter or achieving the parameter. A person skilled in the art, having the benefit of this disclosure, would understand the variability of a parameter as connoted by the use of the word about.
  • Polymorph refers to the occurrence of different crystalline forms of a compound differing in packing or conformation/configuration but with the same chemical composition. Crystalline forms have different arrangements and/or conformations of the molecule in the crystal lattice. Solvates are crystal forms containing either stoichiometric or nonstoichiometric amounts of a solvent. If the incorporated solvent is water, the solvate is commonly known as a hydrate. Hydrates/solvates may exist as polymorphs for compounds with the same solvent content but different lattice packing or conformation.
  • a single compound may give rise to a variety of polymorphic forms where each form has different and distinct physical properties, such as solubility profiles, melting point temperatures, hygroscopicity, particle shape, density, flowability, compactibility and/or x-ray diffraction peaks.
  • the solubility of each polymorph may vary, thus, identifying the existence of pharmaceutical polymorphs is essential for providing pharmaceuticals with predictable solubility profiles. It is desirable to investigate all solid state forms of a drug, including all polymorphic forms, and to determine the stability, dissolution and flow properties of each polymorphic form.
  • Polymorphic forms of a compound can be distinguished in a laboratory by X-ray diffractometry and by other methods such as, infrared or Raman or solid-state NMR spectrometry.
  • X-ray diffractometry and by other methods such as, infrared or Raman or solid-state NMR spectrometry.
  • polymorphs and the pharmaceutical applications of polymorphs see G. M. Wall, Pharm Manuf. 3:33 (1986); J. K. Haleblian and W. McCrone, J. Pharm. Sci., 58:911 (1969); “Polymorphism in Pharmaceutical Solids, Second Edition (Drugs and the Pharmaceutical Sciences)”, Harry G. Brittain, Ed. (2011) CRC Press (2009); and J. K. Haleblian, J. Pharm. Sci., 64, 1269 (1975), all of which are incorporated herein by reference.
  • XRPD means X-ray powder diffraction, an analytical technique which measures the diffraction of X-rays in the presence of a solid component.
  • Materials which are crystalline and have regular repeating arrays of atoms generate a distinctive powder pattern. Materials with similar unit cells will give powder patterns that are similar in position as measured in ° 2 ⁇ (theta). Solvates which exhibit this property are called isostructural or isomorphous solvates.
  • the intensity of the reflections varies according to the electron density causing diffraction as well as sample, sample preparation, and instrument parameters. Analysis of XRPD data is based upon the general appearance of the measured powder pattern(s) with respect to the known response of the X-ray diffraction system used to collect the data.
  • diffraction peaks that may be present in the powder pattern
  • their positions, shapes, widths and relative intensity distributions can be used to characterize the type of solid state order in the powder sample.
  • the position, shape and intensity of any broad diffuse scatter (halos) on top of the instrumental background can be used to characterize the level and type of solid state disorder.
  • the combined interpretation of the solid state order and disorder present in a powder sample provides a qualitative measure of the macro-structure of the sample.
  • 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 therapeutic products.
  • phrases “pharmaceutically acceptable salt” as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound of the invention.
  • Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, 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-
  • 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.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable 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.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric 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 pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic 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
  • an inorganic acid such as hydro
  • 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.
  • suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art.
  • an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid and the like
  • 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 pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic 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 discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1) 1 19; P. Gould, International J. of Pharmaceutics (1986) 33 201 217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; Remington's Pharmaceutical Sciences, 18 th ed., (1995) Mack Publishing 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.
  • phrases “pharmaceutically acceptable” 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 an association or complex of one or more solvent molecules and a compound of the invention.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • the compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • the present invention includes polymorphs of GDC-0077, and processes, methods, and reagents for the production of polymorphs of GDC-0077, shown as Formula I (CAS Registry Number 2060571-02-8):
  • GDC-0077 includes all stereoisomers, geometric isomers, tautomers, and pharmaceutically acceptable salts thereof.
  • GDC-0077 is the API (Active Pharmaceutical Ingredient) in formulations being developed for the clinical treatment of breast cancer and other disorders.
  • XRPD X-ray Powder Diffraction
  • All crystalline substances are distinguished by their crystallographic unit cells (and therefore peak positions). By comparing measured peak positions with those held in a database, the crystalline substance may be identified uniquely.
  • the positions of all peaks are generally a function of three parameters: a, b, c and three angles: alpha, beta, gamma ( ⁇ , ⁇ , ⁇ ) defining the elementary parallelepiped that constitutes the crystallographic unit cell.
  • FIG. 1 The XRPD pattern of GDC-0077 as starting material, prepared as in Example 1, is shown in FIG. 1 . As evident from the increased baseline counts and poorly resolved diffraction peaks, the starting material is poorly crystalline.
  • FIG. 2 shows DSC (Differential Scanning Calorimetry) and TGA (Thermogravimetry) traces of starting material GDC-0077. The desolvation/vaporization endotherm and the recrystallization endotherm occur at 64 and 141° C. respectively (onset), followed by the melting endotherm at 214° C. TGA shows a weight loss of ⁇ 2.5% w/w before the melting event. The TGA data ( FIG. 2 ) shows a weight loss of ⁇ 2.5% by 150° C.
  • the DSC thermogram shows a sharp melting endotherm with an extrapolated onset of ⁇ 214° C. preceded by a prominent shallow endotherm (possibly desolvation/vaporization) and an exotherm (crystallization/rearrangement/phase transformation) in the 50-175° C. range.
  • the desolvation/vaporization endotherm and the recrystallization endotherm occur at 64 and 141° C. respectively (onset), followed by the melting endotherm at 214° C.
  • TGA shows a weight loss of ⁇ 2.5% w/w before the melting event.
  • FIG. 3 shows overlay of the XRPD patterns of the different solid form hits, Forms 1-VI, obtained in the 96 well HTS polymorph screening for GDC-0077. The two most frequently obtained forms were Forms I and II, of which Form II matched the starting material ( FIG. 3 ).
  • Several other new polymorph hits were identified from the evaporation, precipitation and cooling plates which were scaled up 10 ⁇ fold, i.e. 150-200 mg each for further characterization. Table 1 summarizes the scale up condition for six different forms I-VI.
  • FIG. 4 shows XRPD of anhydrous Form I (Form A) GDC-0077.
  • Table 2 shows the XRPD Peak Search Report for GDC-0077 Form I/A.
  • FIG. 5A shows Scanning electron microscopy (SEM) at 1000 ⁇ magnification of anhydrous Form I (Form A) GDC-0077.
  • FIG. 5B shows Polarized light microscopy (PLM) at 200 ⁇ magnification of anhydrous Form I (Form A) GDC-0077.
  • Form I was found to be substantially crystalline with small, rod shaped crystals of 30-40 Lm (microns) in length.
  • Forms IV-VI appeared similar by XRPD ( FIG. 3 ) with differences in relative peak intensities in the 5-20° 20 range. These three forms showed a weight loss of ⁇ 13% w/w up to 150° C. by TGA ( FIG. 6B ), and multiple transitions up to the melting point at 214° C. by DSC ( FIG. 6C ). These transitions are speculated to be solvent loss (first endotherm, 88° C. for Forms IV and V) followed by crystallization of intermediate form and its subsequent melting/conversion to Form A, which melts at 214° C. Form VI shows a similar trend of desolvation followed by a recrystallization exotherm to Form A by DSC.
  • FIG. 9 shows the isothermal dehydration and the weight loss profile of the equilibrated solid.
  • FIG. 9 shows Isothermal TGA traces of trihydrate Form B GDC-0077 before and after equilibration at RT at 60° C. As evident from the data, the hydrate dehydrates readily, losing almost all of its lattice water (12%) at 60° C.
  • the water sorption desorption behavior of trihydrate Form B GDC-0077 at 25° C. is depicted in FIG. 10A .
  • the dynamic vapor sorption experiment provides information about the dehydration behavior of Form B. Dehydration of Form B commences rapidly below 40% RH (desorption curve) and is complete by the time the sample is exposed to 0% RH. Although there is a hysteresis showing equilibrium lag, the anhydrous product rehydrates beyond 40% RH, equally readily.
  • the XRPD of Form III was different from Forms IV-VI ( FIG. 3 ).
  • the thermal analysis DSC and TGA traces of Form III in FIG. 10B shows several transitions (desolvation/vaporization endotherm), similar to Form B in FIG. 6C , followed by recrystallization exotherm and melting of the final form, which was confirmed to be Form A by XRPD ( FIG. 11 ).
  • FIG. 11 shows overlay of the XRPD patterns of Form A and Form III solid forms. XRPD patterns of Form III at RT, and when heated to 165 and 195° C. are shown. Form III converts to Form A upon heating at ⁇ 165° C. ( FIG. 11 ).
  • Form III showed only a 3% w/w weight loss by TGA, its water content was found to be 11% by qNMR.
  • Form II was found to be the same as the starting material characterized in FIGS. 1 and 2 . Substantial similarity was observed upon comparison of the XRPD patterns of Form II, Form III and the product phase obtained upon isothermal dehydration of Form B at 60° C., indicating that Forms II and III are simply partially desolvated intermediates of Form B, and differing only in their degrees of desolvation.
  • Form is the partially dehydrated intermediate of Form B trihydrate. The possibility of existence of an intermediate hydrate form is also suggested from the step wise desorption profile of Form B ( FIG. 10A ).
  • Forms II/III partially dehydrated form
  • Form C At higher temperatures (>165 C), Form III/C converts to Form I/A.
  • FIG. 12B summarizes the results of the slurry bridging experiments of hydrate-anhydrate mixtures, where the water activity is plotted against the vehicle composition (% water, v/v).
  • FIG. 12B shows slurry bridging experiment data for GDC-0077 hydrate-anhydrate system in ethanol-water mixtures at RT (room temperature).
  • the equilibrium RH (relative humidity) zone for the two forms was identified as 82-86%, equivalent to 65-83% w/w water content.
  • the anhydrous form (Form A) was found to be stable up to water activity (a w ) of 0.82, whereas the hydrate (Form B) was found to be the stable form at a w >0.86.
  • the a w of anhydrate-hydrate equilibrium lies in the 0.82-0.86 range.
  • FIG. 12C shows XRPD of trihydrate Form B GDC-0077.
  • Table 2A shows the XRPD Peak Search Report for GDC-0077 Form B.
  • FIG. 13A shows the XRPD pattern of GDC-0077 THF solvate, which was found to contain 10% w/w THF as analyzed by LC-MS. A 14% w/w weight loss was observed by TGA. DSC showed multiple transitions which were further investigated by heating the sample to 175° C. and 201° C., prior to the occurrence of the endotherms ( FIG. 13B ). An overlay of the XRPD patterns of THF solvate, and the product phases obtained upon heating to 175 and 210° C. shows the solvate desolvates to intermediate anhydrous Form which ultimately converts to Form A ( FIG. 13C ). At 175° C.
  • THF solvate desolvates completely to form an intermediate anhydrous form (Form D) which subsequently melts and recrystallizes to Form A that melts at 215° C. (onset).
  • Form D intermediate anhydrous form
  • FIG. 14 shows Thermal analysis of Form D. The phase transitions are marked against the respective endotherms. Negligible weight loss ( ⁇ 1% w/w) prior to melting confirms that Form D is anhydrous.
  • FIG. 15A shows XRPD of anhydrous Form D GDC-0077. Table 3 shows the representative XRPD peaks of Form D.
  • Form A which melts at ⁇ 213-215° C. has a heat of fusion of ⁇ 100 J/g ( FIG. 6A ) whereas Form D shows a melting onset of 190° C. with a heat of fusion of ⁇ 82-48 J/g ( FIG. 14 ) by thermal analysis of the GDC-0077 THF solvate, depending on the sample history and purity.
  • FIG. 15B shows overlay of the XRPD patterns of Forms A, D and final solid form of GDC-0077 obtained after slurrying Forms A and D (1:1 mixture) overnight in n-propanol (RT).
  • Form D converts to Form A in the slurry.
  • Form A is the more stable form between RT to 214° C.
  • FIG. 17 shows the Solid form landscape for GDC-0077, and provides a comprehensive snapshot of the different solid forms identified via high throughput screening and during crystallization optimization.
  • the form landscape shows the phase transformation between the different forms and details the experimental conditions enabling these transformations as a guide for crystallization and scale up of the appropriate form.
  • XRPD FIG. 4
  • solid-state NMR FIGS. 7A and 7B confirmed Form A.
  • FIG. 7A shows 13C SSNMR (solid-state nuclear magnetic resonance) spectra of anhydrous Form I (Form A) GDC-0077.
  • FIG. 7B shows 19F SSNMR of anhydrous Form I (Form A) GDC-0077.
  • Thermal analysis DSC and TGA traces are included in FIG.
  • Form A is confirmed to be anhydrous.
  • Microscopy data SEM and PLM are shown in FIGS. 5A and 5B , where the GDC-0077 Form API particles appear to be plate like.
  • Table 4 shows the Particle Size Distribution (PSD) data for 5, 15 and 30 second sonication.
  • Water sorption data of anhydrous Form I (Form A) GDC-0077 is shown in FIG. 8 . The compound absorbs negligible moisture (0.25% w/w) up to 90% RH (25° C.).
  • Milling of Form A may optimize certain PK properties.
  • a scaled up Form A lot was milled and also placed on stability studies (40° C./75% RH, 25° C./60% RH, open vials). Three more lots were also milled which behaved similarly upon milling.
  • a milled representative lot was characterized and the physical form was determined. This lot was slurried in 100% ethanol to obtain Form A and milled using a jet mill at for 3.5 hrs with 60 psi pressure. The yield was found to be 91%.
  • GDC-0077 batch was placed on stability at 40° C./75% RH and 25° C./60% RH in open vials. Solid-state data was collected at 4 and 8 weeks to evaluate effect of temperature and humidity on the physical form.
  • the milled and stability samples were characterized by XRPD, PLM, DSC, TGA, Water sorption analysis and surface area analysis. DSC runs were performed in the modulated mode using non-hermetically crimped pans, a heating rate of 1° C./min from 0-175° C., modulation amplitude of ⁇ 1° C. and a period of 60 seconds.
  • FIG. 16A shows the crystallites to be of uniform size and of the order of about 5 ⁇ m, which corroborates the PSD data, and demonstrates the milled GDC-0077 Form A remains stable upon milling.
  • the indication of generation of disorder is more obvious in the DSC trace where a small exotherm is observed at 113° C. (inset of FIG. 16B ) followed by the melting endotherm of Form A at 214° C. ( FIG. 16B ).
  • This surface area determined by BET analysis was found to be twice that generated simply by taking into account the sauter mean diameter (D3,2) fluid dynamics method of defining particle size, which was 3.42 m2/g. This indicates that the marked increase in surface area is contributed by generation of surface disorder.
  • a 1% w/w increase in moisture uptake is observed in the water sorption profile of the milled lot when compared to the unmilled material (weight gain of 0.25% up to 90% RH, FIG. 8 ), which further confirms presence of disorder upon milling.
  • Crystalline forms of GDC-0077 detailed herein are useful for treating a human or animal patient suffering from a disease or disorder arising from abnormal cell growth, function or behavior associated with PI3K such as cancer, may thus be treated by a method comprising the administration thereto of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B).
  • a human or animal patient suffering from cancer may also be treated by a method comprising the administration thereto of a crystalline polymorph of GDC-0077 detailed herein. The condition of the patient may thereby be improved or ameliorated.
  • Methods of the invention also include treating cancer selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma (NSCLC), small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, pancreatic, myeloid disorders, lymphoma, hairy cells, buccal cavity, naso-pharyngeal, pharynx, lip, tongue, mouth, small intestine, colon-rectum, large intestine, rectum, brain and
  • Polymorphs of GDC-0077 may be employed alone or in combination with additional therapeutic agents for the treatment of a disease or disorder described herein, such as inflammation or a hyperproliferative disorder (e.g., cancer).
  • a crystalline polymorph of GDC-0077 detailed herein e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B
  • a pharmaceutical combination formulation, or dosing regimen as combination therapy, with an additional, second therapeutic compound that has anti-inflammatory or anti-hyperproliferative properties or that is useful for treating an inflammation, immune-response disorder, or hyperproliferative disorder (e.g., cancer).
  • the additional therapeutic may be a CDK4/6 inhibitor, Bcl-2 inhibitor, a JAK inhibitor, an anti-inflammatory agent, an immunomodulatory agent, chemotherapeutic agent, an apoptosis-enhancer, a neurotropic factor, an agent for treating cardiovascular disease, an agent for treating liver disease, an anti-viral agent, an agent for treating blood disorders, an agent for treating diabetes, and an agent for treating immunodeficiency disorders.
  • the second therapeutic agent may be an NSAID anti-inflammatory agent.
  • the second therapeutic agent may be a chemotherapeutic agent.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to GDC-0077 such that they do not adversely affect each other. Such compounds are suitably present in combination in amounts that are effective for the purpose intended.
  • a composition of this invention comprises a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), in combination with a therapeutic agent such as a CDK4/6 inhibitor.
  • the combination therapy may be administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • 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 therapeutic agents or treatments.
  • the combination therapy may provide “synergy” and prove “synergistic”, i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes, separate pills or capsules, or separate infusions.
  • an effective dosage of each active ingredient is administered sequentially, i.e., serially
  • effective dosages of two or more active ingredients are administered together.
  • a crystalline polymorph of GDC-0077 detailed herein may be combined with other therapeutic, hormonal or antibody agents such as those described herein, as well as combined with surgical therapy and radiotherapy.
  • Combination therapies according to the present invention thus comprise the administration of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), and the use of at least one other cancer treatment method.
  • the amounts of the crystalline polymorph of GDC-0077 and the other pharmaceutically active therapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • Additional therapeutic agents employed in combination with a crystalline polymorph of GDC-0077 detailed herein include 5-FU, docetaxel, eribulin, gemcitabine, cobimetinib, ipatasertib, paclitaxel, tamoxifen, fulvestrant, GDC-0810, dexamethasone, palbociclib, bevacizumab, pertuzumab, trastuzumab emtansine, trastuzumab and letrozole.
  • a method for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B).
  • the cancer is a breast cancer.
  • the cancer is a hormone receptor positive (HR+) breast cancer.
  • the cancer is an estrogen receptor positive (ER+) breast cancer.
  • the cancer is a HER2-negative breast cancer.
  • the cancer is a HR+ metastatic breast cancer.
  • the cancer is a HR-positive, HER2-negative advanced breast cancer.
  • the cancer is a HER2-negative, ER-negative and progesterone receptor (PR)-negative breast cancer.
  • the subject is human.
  • the subject is a postmenopausal woman.
  • the breast cancer subtype is Basal or Luminal.
  • the cancer has a PIK3CA mutation.
  • the cancer expresses a PIK3CA mutant selected from E542K, E545K, Q546R, H1047L and H1047R.
  • the cancer expresses a PTEN mutant.
  • the method of treating cancer further comprising administering to the subject one or more additional therapeutic agent(s).
  • the one or more additional therapeutic agent(s) is/are selected from a CDK4/6 inhibitor (e.g., palbociclib, ribociclib and abemaciclib), a selective estrogen receptor degrader (SERD) (e.g., fulvestrant), and an aromatase inhibitor (e.g., letrozole).
  • the additional therapeutic agent palbociclib.
  • the additional therapeutic agent is fulvestrant.
  • the one or more additional therapeutic agents are palbociclib and letrozole.
  • An aspect of the invention is a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), or a pharmaceutical composition comprising a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), for use in the treatment of cancer.
  • the cancer is a HR-positive and HER2-negative breast cancel expressing a PIK3CA mutation.
  • the polymorphs for use further comprise one or more additional therapeutic agents (e.g., fulvestrant, palbociclib and/or letrozole).
  • An aspect of the invention is the use of a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), or a pharmaceutical composition comprising a crystalline polymorph of GDC-0077 detailed herein (e.g., crystalline anhydrate Form A, crystalline anhydrate Form D, or crystalline trihydrate Form B), in the manufacture of a medicament for use in the treatment of cancer.
  • the cancer is a HR-positive and HER2-negative breast cancel expressing a PIK3CA mutation.
  • the uses further comprise one or more additional therapeutic agents (e.g., fulvestrant, palbociclib and/or letrozole).
  • a polymorph form of GDC-0077, Formula I may be formulated in accordance with standard pharmaceutical practice for use in a therapeutic combination for therapeutic treatment (including prophylactic treatment) of hyperproliferative disorders in mammals including humans.
  • the invention provides a pharmaceutical composition comprising GDC-0077 in association with one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient.
  • Suitable carriers, diluents, glidants, and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the compound of 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 composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers 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.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • compositions of a polymorph form of GDC-0077 may be prepared for various routes and types of administration with pharmaceutically acceptable diluents, carriers, excipients, glidants or stabilizers (Remington's Pharmaceutical Sciences (1995) 18th edition, Mack Publ. Co., Easton, 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.
  • 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.
  • 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 ordinarily can be stored as a solid composition, a tablet, a pill, a capsule, a lyophilized formulation or as an aqueous solution.
  • the pharmaceutical formulations of the invention 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 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 medical practitioners.
  • Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl, ethanol, or benzylalcohol; alkyl parabens such as methyl or propyl paraben; catechol; 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
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Other examples of drug formulations can be found in Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, Vol 3, 2 nd Ed., New York, N.Y.
  • Tablets may comprise one or more pharmaceutically acceptable carrier, glidant, diluent, or excipient selected from microcrystalline cellulose, lactose, sodium starch glycolate, and magnesium stearate.
  • glidants may be selected from silicon dioxide, powdered cellulose, microcrystalline cellulose, metallic stearates, sodium aluminosilicate, sodium benzoate, calcium carbonate, calcium silicate, corn starch, magnesium carbonate, asbestos free talc, stearowet C, starch, starch 1500, magnesium lauryl sulfate, magnesium oxide, and combinations thereof.
  • the pharmaceutical formulations include 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 generally are found in Remington's Pharmaceutical Sciences 18 th 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.
  • 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.
  • compositions may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation 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.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the
  • compositions of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, inhalation, intradermal, intrathecal, epidural, and infusion techniques), transdermal, rectal, nasal, topical (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 Pharmaceutical Sciences, 18 th Ed., (1995) Mack Publishing Co., Easton, Pa. Other examples of drug formulations can be found in Liberman, H. A.
  • the compounds may be administered by intralesional administration, including perfusing or otherwise contacting the graft with the inhibitor before transplantation. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound is administered 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 1 mg to about 100 mg of a polymorph form of GDC-0077, such as about 2 mg to about 50 mg, about 3 mg to about 20 mg, about 3 mg to about 15 mg, about 3 mg to about 20 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 9 mg, about 12 mg, about 15 mg, or about 20 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 pharmacodynamic (PD) properties, including absorption, distribution, metabolism, and excretion of the particular compound.
  • PK pharmacokinetic
  • PD pharmacodynamic
  • toxicity factors may influence the dosage and administration dosing regimen.
  • 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
  • GDC-0077 was prepared according to WO 2017/001645, US 2017/0015678, each of which are incorporated by reference.
  • GDC-0077 Form A (anhydrate) was obtained by slurring GDC-0077 in ethanol at 50° C. for 4 hours and then evaporating the solvent under nitrogen purge to give the highly crystalline Form A.
  • DSC thermogram showed one endothermic transition with an onset at ⁇ 212-214° C. with an associated heat of fusion of ⁇ 107 J/g.
  • Water solubility of Form A at room temperature is 30.8 ⁇ g/mL at a pH of 7.06.
  • the trihydrate form was obtained by slurrying Form A in DI water over 4 days (RT), centrifuging the slurry to remove supernatant and then drying the solid at RT for a few hours.
  • 96-Well plate automated HTS using the Symyx CM2 system (Freeslate Inc., CA) was conducted to identify potential polymorphic forms for GDC-0077. Approximately 20 mg of API was added to each well, to which 800 ⁇ l (microliters) of solvent (neat or mixture) was added and the slurry was stirred for 2 hours at 50° C.
  • Solvents included water, 1,2-dichloroethane, heptane, cyclohexane, ethanol, 1-propanol, acetonitrile, butylamine, nitromethane, 1,4-dioxane, benzene, perfluoroheptane, ethyl acetate, (trifluoromethyl)benzene, butan-2-one (MEK), 1,2-dimethoxyethane, 2-methyltetrahydrofuran, carbon tetrachloride, dimethylacetamide, tetrahydrofuran (THF), acetone, anisole, toluene, and 2-ethoxyethanol.
  • MEK butan-2-one
  • DI Deionized
  • XRPD patterns were collected with a Rigaku SmartLab® diffractometer (Rigaku Corp., Tokyo, Japan), using an incident beam of Cu K ⁇ (1.541904 ⁇ ) radiation generated using Cross Beam optics (40 kV ⁇ 44 mA).
  • GDC-0077 powder samples were packed using the top fill method onto zero-background holders and scans were acquired at a scan speed of 1 or 3.0°/min and step size of 0.02 or 0.04° 20 (2-theta) over 2-40° 20 range in the bragg-brentano or parallel beam configuration (reflection geometry). Data was analyzed using commercial software (JADE®, version 9, Materials Data Inc., Livermore, Calif.).
  • thermogravimetric analyzer (Discovery TGA, TA instruments) 3-4 mg of GDC-0077 samples were heated in an open aluminum pan from RT to 350° C. at a heating rate of 10° C./min and RT to 3500 under dry nitrogen purge. Temperature calibration was performed using Alumel® and Nickel. Standard weights of 100 mg and 1 gm were used for weight calibration.
  • thermogravimetric analyzer (Q500 TGA, TA instruments) 3-4 mg of GDC-0077 samples were heated in an open aluminum pan from RT to 60° C. at a heating rate of 10° C./min held isothermally at 60° C. overnight. Samples were then either analyzed by XRPD or left to equilibrate at RT for 4 hours in the sample pan and then rerun isothermally at 60° C. using the same experimental parameters as mentioned above.
  • Samples were dispersed in silicon oil and observed under cross polarizers of a video enhanced Leica DM 4000B microscope equipped with a high resolution CCD camera and motorized stage (Clemex Technologies Inc., Longueuil, Quebec, Canada) at 200 ⁇ magnification. Photomicrographs were acquired using the Clemex Vision PE software (Clemex Technologies Inc., Longueuil, Quebec, Canada).
  • Particle size analysis was performed using a Malvern Mastersizer 2000 instrument equipped with a Hydros 2000SM wet dispersion attachment (Malvem Instruments Ltd., Malvern, UK). ⁇ 30 mg of API was weighed into a vial and 1 mL of 0.1% Span 85 in heptane was added. The vial was sonicated for 5 seconds, about 0.3 mL was added to the sampler at a stir speed of 1500 rpm, and a PSD was performed at an obscuration of 10-20%. The sample was then sonicated for 10 more seconds (a total of 15 seconds), about 0.3 mL was added to the sampler, a PLM image was acquired, and PSD was performed.
  • the same sample was then sonicated for another 15 seconds (a total of 30 seconds), about 0.3 mL was added, a PLM photo was taken and PSD was performed. From the sonication study, an appropriate sonication period was chosen. The PLM images and PSDs were used to determine how much sonication the sample needed to disperse clumps but prevent or minimize crystal fracturing. Three more samples, of ⁇ 10 mg, were weighed into vials and 1 mL of 0.1% Span 85 in heptane was added. The samples were sonicated for the sonication period determined in the sonication study. The final PSD analysis was performed in triplicate using the predetermined sonication period. The instrument was rinsed twice with isopropyl alcohol (IPA) and once with heptane before being filled with 0.1% Span 85 in heptane for each sample. After the last sample had run, the instrument was rinsed with IPA once.
  • IPA isopropyl alcohol

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicinal Preparation (AREA)
US15/963,876 2017-04-28 2018-04-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production Abandoned US20180339997A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US15/963,876 US20180339997A1 (en) 2017-04-28 2018-04-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,545 US11591345B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,537 US11028100B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US18/101,951 US20230167128A1 (en) 2017-04-28 2023-01-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762491812P 2017-04-28 2017-04-28
US15/963,876 US20180339997A1 (en) 2017-04-28 2018-04-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/875,545 Division US11591345B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,537 Continuation US11028100B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production

Publications (1)

Publication Number Publication Date
US20180339997A1 true US20180339997A1 (en) 2018-11-29

Family

ID=62492573

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/963,876 Abandoned US20180339997A1 (en) 2017-04-28 2018-04-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,545 Active US11591345B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,537 Active US11028100B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US18/101,951 Pending US20230167128A1 (en) 2017-04-28 2023-01-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production

Family Applications After (3)

Application Number Title Priority Date Filing Date
US16/875,545 Active US11591345B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US16/875,537 Active US11028100B2 (en) 2017-04-28 2020-05-15 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US18/101,951 Pending US20230167128A1 (en) 2017-04-28 2023-01-26 Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production

Country Status (12)

Country Link
US (4) US20180339997A1 (ja)
EP (1) EP3615541A1 (ja)
JP (2) JP6998969B2 (ja)
KR (1) KR102342776B1 (ja)
CN (1) CN110650963B (ja)
AU (1) AU2018259089B2 (ja)
BR (1) BR112019013292A2 (ja)
CA (1) CA3048591A1 (ja)
IL (2) IL298518A (ja)
MX (1) MX2019012507A (ja)
TW (1) TWI826373B (ja)
WO (1) WO2018197653A1 (ja)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190263793A1 (en) * 2015-07-02 2019-08-29 Genentech, Inc. Benzoxazepin oxazolidinone compounds and methods of use
US10781219B2 (en) 2016-12-15 2020-09-22 Genentech, Inc. Process for the preparation of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[F]imidazo[1,2-D][1,4]oxazepin-9-yl)amino) propanamide
CN112830935A (zh) * 2019-11-25 2021-05-25 上海翰森生物医药科技有限公司 含三并环类衍生物自由碱的晶型及其药物组合物

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6998969B2 (ja) * 2017-04-28 2022-02-10 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト (s)-2-((2-((s)-4-(ジフルオロメチル)-2-オキソオキサゾリジン-3-イル)-5,6-ジヒドロベンゾ[f]イミダゾ[1,2-d][1,4]オキサゼピン-9-イル)アミノ)プロパンアミドの多形体及び固体形態と、生産方法
US20220242811A1 (en) 2019-05-21 2022-08-04 Jiangsu ForeFront Pharmaceutical Co., Ltd Crystal form of treprostinil sodium salt and preparation method therefor
EP3993785A4 (en) * 2019-07-07 2023-08-02 Olema Pharmaceuticals, Inc. ESTROGEN RECEPTOR ANTAGONIST SCHEMES
EP4067363A4 (en) * 2019-11-25 2023-12-27 Shanghai Hansoh Biomedical Co., Ltd. SALT OR CRYSTAL FORM HAVING THREE CONDENSED RINGS AND PHARMACEUTICAL COMPOSITION THEREOF
TWI801730B (zh) * 2020-04-07 2023-05-11 神雲科技股份有限公司 具系統設定資料同步功能的伺服器

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846824A (en) 1994-02-07 1998-12-08 Ludwig Institute For Cancer Research Polypeptides having kinase activity, their preparation and use
GB9208135D0 (en) 1992-04-13 1992-05-27 Ludwig Inst Cancer Res Polypeptides having kinase activity,their preparation and use
US6274327B1 (en) 1992-04-13 2001-08-14 Ludwig Institute For Cancer Research Polypeptides having kinase activity, their preparation and use
ES2444779T3 (es) 2009-09-28 2014-02-26 F. Hoffmann-La Roche Ag Compuestos de benzoxazepina inhibidores de la PI3K y su utilización en el tratamiento de cáncer
CN103476770B (zh) 2010-11-25 2017-02-15 拉蒂欧制药有限责任公司 阿法替尼盐和多晶型物
WO2015091305A1 (en) 2013-12-16 2015-06-25 F. Hoffmann-La Roche Ag Polymorphs of 2-(4-(2-(1-isopropyl-3-methyl-1h-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)-1h-pyrazol-1-yl)-2-methylpropanamide, methods of production, and pharmaceutical uses thereof
TW202108592A (zh) * 2015-07-02 2021-03-01 瑞士商赫孚孟拉羅股份公司 苯并氧氮呯噁唑啶酮化合物及其用途
SI3555107T1 (sl) * 2016-12-15 2021-07-30 F. Hoffmann-La Roche Ag Postopek za pripravo (S)-2-((2-((S)-4(difluorometil)-2-oksooksazolidin-3-il)-5,6-dihidrobenzo (f)imidazo(1,2-d)(1,4)oksazepin-9-il)amino) propanamida
JP6998969B2 (ja) * 2017-04-28 2022-02-10 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト (s)-2-((2-((s)-4-(ジフルオロメチル)-2-オキソオキサゾリジン-3-イル)-5,6-ジヒドロベンゾ[f]イミダゾ[1,2-d][1,4]オキサゼピン-9-イル)アミノ)プロパンアミドの多形体及び固体形態と、生産方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Brittain et al. "Polymorphism in Pharmaceutical Dosage Forms." Polymorphism in Pharmaceutical Solids XX (January 1999) pp.235-238). (Year: 1999) *
Vippagunta et al (Crystalline Solids: Advanced Drug Delivery Reviews Vol. 48 pages 3-26 published 2001) (Year: 2001) *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190263793A1 (en) * 2015-07-02 2019-08-29 Genentech, Inc. Benzoxazepin oxazolidinone compounds and methods of use
US10851091B2 (en) * 2015-07-02 2020-12-01 Genentech, Inc. Benzoxazepin oxazolidinone compounds and methods of use
US11760753B2 (en) 2015-07-02 2023-09-19 Genentech, Inc. Benzoxazepin oxazolidinone compounds and methods of use
US10781219B2 (en) 2016-12-15 2020-09-22 Genentech, Inc. Process for the preparation of (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[F]imidazo[1,2-D][1,4]oxazepin-9-yl)amino) propanamide
CN112830935A (zh) * 2019-11-25 2021-05-25 上海翰森生物医药科技有限公司 含三并环类衍生物自由碱的晶型及其药物组合物

Also Published As

Publication number Publication date
US11028100B2 (en) 2021-06-08
KR20190133724A (ko) 2019-12-03
IL267464B1 (en) 2023-01-01
US20200399286A1 (en) 2020-12-24
US11591345B2 (en) 2023-02-28
JP6998969B2 (ja) 2022-02-10
JP2020517713A (ja) 2020-06-18
IL267464B2 (en) 2023-05-01
CA3048591A1 (en) 2018-11-01
MX2019012507A (es) 2019-12-19
BR112019013292A2 (pt) 2019-12-10
AU2018259089A1 (en) 2019-07-04
US20230167128A1 (en) 2023-06-01
CN110650963B (zh) 2022-09-27
AU2018259089B2 (en) 2022-05-12
CN110650963A (zh) 2020-01-03
WO2018197653A1 (en) 2018-11-01
KR102342776B1 (ko) 2021-12-23
JP2021181446A (ja) 2021-11-25
IL298518A (en) 2023-01-01
IL267464A (en) 2019-08-29
TWI826373B (zh) 2023-12-21
AU2018259089A9 (en) 2019-10-10
US20210094970A1 (en) 2021-04-01
EP3615541A1 (en) 2020-03-04
TW201843160A (zh) 2018-12-16
WO2018197653A9 (en) 2019-09-19

Similar Documents

Publication Publication Date Title
US11028100B2 (en) Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
US10723730B2 (en) Solid forms of a selective CDK4/6 inhibitor
JP7320741B2 (ja) B-rafキナーゼのマレイン酸塩、結晶形、調整方法、及びその使用
AU2017203819B2 (en) Polymorphs of 2-(4-(2-(1-isopropyl-3-methyl-1H-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)-1H-pyrazol-1-yl)-2-methylpropanamide, methods of production, and pharmaceutical uses thereof
JP2019529465A (ja) キナゾリン誘導体の塩、その製造方法および使用

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE