WO2015196137A1 - Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide - Google Patents

Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide Download PDF

Info

Publication number
WO2015196137A1
WO2015196137A1 PCT/US2015/036784 US2015036784W WO2015196137A1 WO 2015196137 A1 WO2015196137 A1 WO 2015196137A1 US 2015036784 W US2015036784 W US 2015036784W WO 2015196137 A1 WO2015196137 A1 WO 2015196137A1
Authority
WO
WIPO (PCT)
Prior art keywords
dioxo
crystalline form
xrpd
polymorphic
pattern
Prior art date
Application number
PCT/US2015/036784
Other languages
French (fr)
Inventor
Ernest A. CARRA
Irene Chen
Katie Ann Keaton
Vahid Zia
Original Assignee
Gilead Sciences, 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 Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Priority to EP19167626.1A priority Critical patent/EP3564244B1/en
Priority to CA2950309A priority patent/CA2950309C/en
Priority to NZ726732A priority patent/NZ726732A/en
Priority to EP15734504.2A priority patent/EP3157931A1/en
Priority to AU2015276881A priority patent/AU2015276881B2/en
Priority to JP2016574182A priority patent/JP6386104B2/en
Priority to EP23195199.7A priority patent/EP4309736A3/en
Publication of WO2015196137A1 publication Critical patent/WO2015196137A1/en
Priority to AU2018203737A priority patent/AU2018203737A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring 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
    • A61K31/495Heterocyclic 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
    • A61K31/55Heterocyclic 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic 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 three hetero rings
    • C07D498/14Ortho-condensed systems
    • 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
    • C07D498/12Heterocyclic 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 three hetero rings
    • C07D498/18Bridged 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/537Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines spiro-condensed or forming part of bridged ring systems
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to novel crystalline forms and co-crystals of (2R,5SJ3aR)-8-hydroxy-7, ⁇
  • Human immunodeficiency vims infection and related diseases ate a major public health problem worldwide.
  • Human immunodeficiency virus type 1 (ffiV-1) encodes three enzymes which are required for viral replication: reverse transcriptase, protease, arid integrase.
  • reverse transcriptase a enzyme which is required for viral replication
  • protease a substrate for converting viral genome to a virus.
  • arid integrase integrase
  • drags targeting reverse transcriptase and protease are in wide use and have shown effectiveness, particularly when employed in combination, toxicity and development of resistant strains have limited their usefulness (Palclla, et al. N. Engl. J Med. (1998) 333:853-860; Richman, D, D. Nature (2001 ) 410:995-1001).
  • a goal of antiretrovira! therapy Is to achieve viral suppression in the HIV infected patient.
  • Treatment guidelines published by the United States Department of Health and Human Services provide that achievement of viral suppression requires the use of combination therapies, i.e., several drags from at least two or more drug classes.
  • decisions regarding the treatment of HIV infected patients are complicated when the patient requires treatment for other medical conditions. Because the standard of care requires the use of multiple different drugs to suppress HIV, as well as to treat other conditions the patient may be experiencing, the potential for drug interaction is a criterion for selection of a drug regimen. As such, there is a need for antirstroviral therapies having a decreased potential for drug interactions.
  • the present invention is directed to novel forms of (2R.5SJ 3a ⁇ -8- hydroxy-7,9 lioxG-N-(2,4 5 6-?TM ⁇
  • the present invention is directed to (2R,5S, 13aR)-S-hydroxy-
  • fee present invention is directed to i2 .SS, 13a V8- hydroxy-7,9 ⁇ ioxo-N ⁇
  • the present invention is directed to (2R,SS 5 13aR)-8-
  • the present invention is directed to (2R s 5S,13aR)- ⁇ hydroxy-7 5 9-dioxo ⁇ N ⁇ (2,4,0 rifiuQre ⁇
  • the present invention is directed to (2R,5S,1 aR) ⁇ 8 ⁇ hydroxy-7,9 ⁇ dioxo-N ⁇ (2,4,6-trifluorobenzyl)-2 s 3,4,5,7 s 9, 13,13 a ⁇ oeiahydre-2,5 ⁇
  • the present invention is directed to (2R,SS, 13aR)-8- hydroxy-7,9-dioxO"N-(2,4,6 rifluorobenzyl) ⁇ 2 ? 3 ! 4 s 5.7,9J.3 5 13a-ocfebydrO"2,5 »
  • the present invention Is directed to (2R,5S, 13 ⁇ -8- bydroxy ⁇ 7 s 9-dioxo-N-(2,4,6 rit1uorokinzyi ⁇ -2,3,4,5,7,9, ! 3, 13a-octahydro-2.5- methanopyrido[ I ⁇ 2':4,5]pyrazino[2,l -b] [ 1 s 3]oxazepine- i 0-catboxamide Form VII.
  • the present invention is directed to (2R 5 5S,13aR)-8- h dro ⁇ -dio o-N ⁇ -triflisorobeaxy ⁇ J ⁇ ,? ⁇ .13, ] 3a-octahydro ⁇ 2,5- ffiethanopyrido[172 ⁇ 4,S]pyrazmoP ⁇ Form VIII.
  • present Invention Is directed to a fnmarle acid co- cr ⁇ of (2R,5S 3aR) ⁇ 8-hydroxy-7 ⁇
  • the present invention is directed to a citric acid co-erystal of (2R,5S,13aR)-8 tydrox ⁇
  • the present invention is directed to an oxalic acid co- crystal of (2R,5S, 13aR)-8-hydroxy-7,9 ⁇ i-oxo-N ⁇
  • the present Invention is directed to crystalline forms and co-crystals of (2R s 5S 5 13aR)-8-hydroxy-7 ) 9"dioxo-N- ⁇ 2,4 5 6 ⁇ trii1.uorobenzyi) ⁇
  • the present invention is directed to novel forms of sodium
  • the present invention is directed to sodium.
  • the present invention is directed to novel forms of potassium (2R ! 5S, 13aR)-7 5 9 ⁇ dioxo-10-( ⁇ 2 J 4,6-trifiuorobenzyl)carbam.oyl)-2 s 3,4,5 s 7,9 5 I3, I 3a- octahydro ⁇ 2 > 5 ⁇ methaBopyrido[ l ⁇ 2 I :4 s 5]pyrazino[2J. ⁇ b][l s 3]oxaz €pin-S-oiaie, having the following structure (Formula ill):
  • the present invention is directed to potassium
  • the present invention is directed to potassium
  • the present invention is directed to hydraied potassium (2R,5S, .1 ;1 ⁇ 4R)"7,9-dioxo-10- ⁇ 2 3 4 5 6 fifluorobenzyi)ca banioyl) » 2 ; 3 5 4,S s 7,9, 13, 13a- octahydrO"2 > 5 ⁇ r eihanopyrido[ I ! ,2 ! A5]pyrazino[2, 1 -b] [ 1 ,3]oxa2;epm-8-olate.
  • the present invention is directed to methods of treating or prophyiactically preventing an HIV Infection by administering compound (e.g. Formulas (I), (II), and/or (III)) provided herein.
  • compound e.g. Formulas (I), (II), and/or (III) provided herein.
  • the present invention s directed to a compound (e.g. Formulas (I), (II), and/or (III)) provided herein for use in methods of treating or
  • the present invention is directed to the use of a compound (e.g. Formulas (I), ( ⁇ ), and/or (III)) provided herein in the manufacture of a medicament for treating or prophyiactically preventing HIV infection.
  • a compound e.g. Formulas (I), ( ⁇ ), and/or (III) provided herein in the manufacture of a medicament for treating or prophyiactically preventing HIV infection.
  • Figure 6 Actual and calculated XRPD pattern for (2R,5S,13aR)-8-hydroxy ⁇ 7 s 9» dio o-N ⁇ lfluoro iizylJ ⁇ ⁇ JS.Da-ociahydro- ⁇ S- methairopyTldo[r > 2 , :4 s 5]pyra2ino[2 -b][l,3]oxazepke ⁇ !0-carboxamide oxalic acid co- crystal Form I.
  • Fsg?ire 10 TGA for (2R.5S, 13aR>8-hydroxy-7.9-dioxo-N-(2,4,6-
  • Figure 14 DVS for (2R,5SJ3 ⁇ R) ⁇ S- ydrox --7 J 9-dloxo « N ⁇ (2,4,6-.
  • Figure 15 DVS for sodium (2R,5S 5 13aR)-7 s 9-dioxo ⁇ I0 ⁇ ((2 > 4,6- udil «orobenzj.'i)carbamoyl)-2 ! ,3 J 4 i 5 i 7,9,13,13a-octahydro ⁇ 2,5 ⁇
  • the invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of Formulas (I), ( ⁇ ), and (III) being isotonics! ly-Ja beled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 3 ⁇ 4 3 ⁇ 4 U C, i3 C, M C, 3 ⁇ 4, 15 N, i5 0, 37 0, 38 G, 33 P, 3 3 ⁇ 4 3S S, l % 3 ⁇ 4 n ⁇ and % respectively.
  • These radiolabeled compounds could he useful to help determine or measure the
  • Isotopically-labeled compounds of Formulas (f) (II) and (III), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i n 3 ⁇ 4 and carbon- 14. i.e. i4 C, are particularly useful for this purpose in view of their ease of i ncorporation and ready means of detection.
  • Substitution with heavier isotopes such as deuterium, i.e. 3 ⁇ 4 may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half- life may increase or dosage requirements may be reduced. Thus, heavier Isotopes may he preferred In some circumstances.
  • isoiopicaUy-labeled compounds of Formulas (I), (II), (III) can generally be prepared by conventional techniques known to those skilled in the art or by processes ' analogous to those described in the Examples as set out below using an appropriate
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an. efficacious therapeutic agent.
  • ' ptional or “optionally” means that the subsequently described event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • “optionally substituted aryi” means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • 'Thannaccutically acceptable carrier, diluent or excipient includes without limitation any adjuvant, carrier, excipient, glidant sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or ernuisifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • a "pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans.
  • a medium includes ail pharmaceutically acceptable carriers, diluents or excipients therefor.
  • Effective amount refers to an amount of a compound according to the invention, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician.
  • the amount of a compound according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological acti vity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidental! ⁇ ' with the compounds of the invention, and the age, body weight, general health, sex and diet of the patient.
  • a therapeutically effective amount can be determined routinely by one of ordinary skill in the art. having regard to their own knowledge, the state of the ait, and this disclosure,
  • treatment is intended to mean the
  • treatment also encompasses the administration of a compound or composition according to the present invention post-exposure of the individual to the virus but before the appearance of symptoms of the disease, and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching deteetible levels in the blood, and the admins stratioa of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life.
  • the term "treatment” as used herein is intended to mean the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load in a patient.
  • the term “'treatment” as used herein is further or alternatively intended to mean the administration of a compound or composition according to the present Invention to maintain a reduced viral load in a patient.
  • treatment also encompasses the administration of a compound or composition according to the present invention postexposure of the individual to the virus but before the appearance of symptoms of the disease; and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching deteetible levels in the blood, and the administration of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life, in certain embodiments, the term “treatment” as used herein is further or alternatively intended to mean the administration of a compound or composition according to the present invention post-exposure of the individual to the virus as a subsequent or additional therapy to a first-line therapy (e.g., for maintenance of low viral load), [0060] "Prevention” or "preventing *5 means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop.
  • prevention also encompasses the administration of a compound or composition according to the present inventio pre ⁇ exposure of the individual to the virus ⁇ e.g., pre ⁇ exposure prophylaxis), to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching detect! hie levels in the blood.
  • Subject refers to an animal, such as a mamma! (including a human), that has been or will be the object of treatment observation or experiment.
  • the methods described herein may be useful in human therapy and''or veterinary applications.
  • the subject is a mammal (or the patient).
  • the subject (or the patient) is human, domestic animals (e.g., dogs and cats), farm animals (e.g., cattle, horses, sheep, goats and pigs), and/or laboratory animals (e.g., mice, rats, hamsters, guinea pigs, pigs, rabbits, dogs, and monkeys).
  • the subject (or the patient) is a human.
  • “Human (or patient) in need thereof 5 refers to a human who may have or is suspect to have diseases or conditions that would benefit from certain treatment; for example, being treated with the compounds disclosed herein according to the present application.
  • antiviral agent as used herein is intended to mean an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a human being, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a human being.
  • inhibitor of HIV replication is intended to mean an agent capable of reducing or eliminating the ability of HIV to replicate in a host ceil, whether in vitro, ex vivo or in vivo.
  • a "tasjtomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • the present invention includes tantomers of any said compounds.
  • “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human
  • Unit dosage forms are physically discrete units suitable as unitary dosages for subjects (e.g.. human subjects and other mammals), each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical carrier.
  • earboxamide may be an intermediate to the synthesis of (2R,5S,I3aR)-S-hydroxy-7,9-dioso- " N-(2,4,6 rinuorobenzyl)-2,3,4,5,7,9, 13,1 a ⁇ oc ⁇ ahydro-2,5 ⁇
  • a polymorphic form or polymorph or eocrystal may have properties such as bioavaiiabiiity and stability at certain conditions that may be suitable for medical or pharmaceutical uses.
  • a crystalline form of (2R J 5S,13aR)-8 « hydroxy-7,9-dioxo-N-(2,4 > 6- trifluorobenzyl) ⁇ 2,3,4,5,7,9, 13, 13a-octahydro-2,5 ⁇ meihanopyrido[ 1 ',2';4,5]pyrazino[2, 1 - b][l i 3]oxazepine-10 ⁇ carboxamide may provide the advantage of bioavailability and stability, suitable for use as an active ingredient in a pharmaceutical composition.
  • Variations in the crystal structure of a pharmaceutical drug substaxs.ee or active ingredient may affect the dissolution rate (which may affect bioavailability, etc.), manufactiirabiilty (e.g., ease of handling, ability to consistently prepare doses of known strength) and stability' (e.g.. thermal stability, shelf life, etc.) of a pharmaceutical drug product or active ingredient.
  • dissolution rate which may affect bioavailability, etc.
  • manufactiirabiilty e.g., ease of handling, ability to consistently prepare doses of known strength
  • stability' e.g. thermal stability, shelf life, etc.
  • Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or delivery forms, such as solid oral dosage form including tablets and capsules.
  • crystalline forms may provide desired or suitable hygroscopicity, particle size controls, dissolution rate, solubility, purity, physical and chemical stability, ma ufacturabiiity, yield, and/or process control.
  • Carboxamide may provide advantages such as; improving the manufacturing process of an active agent or the stabiiiiy or storability of a drug product form of the compound or an acti ve ingredient and/or having suitable bioavailability and/or stability as an active agent,
  • the compound name provided above is named using ChemBioDraw Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols.
  • the compound may be named or identified with common names, systematic or non- systematic names.
  • the nomenclature systems and symbols that are commonly recognized in the art of chemistry including but not limited to Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (IUPAC).
  • the compound structure provided above may also be named or identified as (2R,5S,13aR)-8-hydJx>xy-7,9- dloxo ⁇ N ⁇ (2,4J-triilnorobenz)d)-2 ; 3,4,5,7,9, 13, i 3a-oetahydro ⁇ 2 5 5- methanopyrido[r,2 i :4,5]p>'razmo[2,l-b][1 ,3]oxazepine-10-carboxaniide under IUPAC and 2,S-Methanopyrido[ 1 ',2 ! :4,5] ⁇ > ⁇ 3 ⁇ [2, 1 ⁇ b] [ 1 ,3 Joxazepine- 10-carboxamide, 2 ⁇ 4,5, , , 13, 13a-oetahydro-8-hydro.xy ⁇ 7 5 ⁇
  • crystalline forms and co-crystals of (2 5 5S,13aR)-8 ⁇ hydiOxy ⁇ 7 s 9-di xo-N ⁇ (2 J 4,6-tri !uorobeiizyl)-2,3 s 4,5,7 s 9 ⁇ 13, 13a-octahydro-2,5- methanopyrido[r s 2 t :4 5]p) ' Tazino[2J -b][l ,3]oxazeplne 0-carboxamide are disclosed.
  • Polymorphic Form I exhibits an X-ray powder diffraction (XRPD) pattens substantially as shown in FIG. L
  • Polymorphic Form I may exhibit a differential scanning ealorimetry (DSC) thermogram substantially as shown in FIG. 7
  • Polymorphic Form I may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG. 10.
  • Polymorphic Form I may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 13.
  • terns "substantially as shown in” when referring, for example, to an XRPD pattern, a DSC thermogram, or a TGA graph includes a pattern, thermogram, or graph that i not necessarily identical to those depicted herein, but that falls within the limits of experimental error or deviations when considered by o e of ordinary skill in the art.
  • polymorphic Form I at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all of the following (a)-(k) apply: (a) polymorphic Form I has an XRPD pattern substantially as shown in FIG. 1 ; (b) polymorphic Form ⁇ has a DSC thermogram
  • polymorphic Form I has a TGA graph substantially as shown In FIG, 10;
  • polymorphic Form I has DVS graphs substantially as shown in FIG. 13;
  • polymorphic Form I has an endomermie event onset;
  • polymorphic Porta I has a moBociimc crystal system.;
  • polymorphic Form ⁇ has a P2(l) space group;
  • polymorphic Form I has a volume of 1855.44( 12) A 3 ;
  • polymorphic Fonn I has a Z value of 4; and
  • polymorphic Form ⁇ has a
  • polymorphic Form I has at. least one, at least two, at. least three, or all of the following properties:
  • polymorphic Form I has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 2e-reflections with the greatest intensity as the XRPD pattern substantially as shown in FIG. I ,
  • polymorphic Form I has an XRPD pattern comprising degree 20-refiections ( ⁇ / ⁇ 0.2 degrees 2 ⁇ ) at 27.4, 13.1, and 17.4. In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 28 ⁇ refleetions ( ⁇ /- 0.2 degrees 2 ⁇ ) at 27,4, 13.1, and 17.4 and one or more of the degree 28-reffeeiions (+/- 0.2 degrees 28) at 10.5, 20.6. and 25.0.
  • polymorphic Form ⁇ has an XRPD pattern comprising degree 29-re flections (+/- 0.2 degrees 28) at 27.4, 13,1, and 17.4 and one of the degree 28-ret3 ⁇ 4ctiorjs (+/- 0.2 degrees 20) at 10,5, 20.6, and 25.0.
  • polymorphic Form I has an XRPD pattern comprising degree 28 ⁇ ref1ections (+/- 0,2 degrees 20) at 27.4, 13,1, and 17.4 and two of the degree 28-reilectlons (+/- 0.2 degrees 2 ⁇ ) at 10.5, 20.6, and 25.0.
  • polymorphic Form I has an XRPD pattern comprising degree 28 -reflections ⁇ ⁇ ? ⁇ / ⁇ 0.2 degrees 20) at 27.4, 13.1, and 17,4 and three of the degree 2 ⁇ - reflections ⁇ - ⁇ -/- 0,2 degrees 28) at 10.5, 20.6, and 25.0, In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 20 -reflections (+/- 0,2 degrees 2 ⁇ ) at 27.4,
  • polymorphic Form I has an XRPD pattern comprising degree 28-refiections (+/- 0.2 degrees 28) at 27.4, 13.1, 17.4, 10.5, 20.6, 25.0 , 16.2, and 22.3. In one embodiment, polymorphic Form I has an XRPD pattern comprising any three degree 29-reflections (+/- 0,2 degrees 28) selected from the group consisting of 27.4, 13.1, 17.4, 10.5, 20.6, 25.0, 16.2, 22.3, 13.9, ⁇ 1.4, and 9.3.
  • polymorphic Form II at leas one, at least two, at least three, at least four, at least five, at least six, or all of the following (a)-(g) apply: (a) polymorphic Form II has an XRPD pattern substantially as shown in FIG.
  • polymorphic Form II has at least one. or all of the following properties:
  • polymorphic Form II has an X PD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 2e-refkctions with the greatest intensity as the XRPD pattern substantially as shown in FIG. 2,
  • polymorphic Form H has an XRPD pattern comprising degree 2 ⁇ - eflections (+/ ⁇ 0.2 degrees 2 ⁇ ) at 6,6, 21.4, and 10.6.
  • polymorphic Form II has an XRPD pattern comprising degree 20-reflections (+/- 0.2 degrees 2 ⁇ ) at 6.6, 21.4, and 10.6 and one or m.ore of the degree 28-refiectioris ⁇ +/- 0.2 degrees 2 ⁇ ) at 12.5, 16.2, and 14.3.
  • polymorphic Form II has an XRPD pattern comprising degree 20-reflections ⁇ /- 0.2 degrees 2 ⁇ ) at 6.6, 21.4, and 10.6 and one of the degree 29-reflections (+/- 0,2 degrees 2 ⁇ ) at 12.5, 16.2, and 14.3, In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 20 ⁇ reflections ( ⁇ * ⁇ /- 0.2 degrees 2 ⁇ ) at 6.6, 21.4, and 10.6 and two of the degree 20- reflections (+/- 0,2 degrees 20) at 12,5,
  • polymorphic Form II lias an XRPD pattern comprising degree 20-refiections ( ⁇ /- 0.2 degrees 2 ⁇ ) at 6.6, 21.4, and 10.6 and three of the degree 20- refiectlons (+/- 0,2 degrees 28) at 12.5, 16,2, and 14.3.
  • polymorphic Form II has an XRPD pattern comprising degree 28-reflections (+/- 0.2 degrees 28) at 6,6, 21,4, 10,6, 12.5, 16.2, and 14.3,
  • polymorphic Form II has an XRPD pattern comprising degree 2e ⁇ refleetions ( ⁇ /- 0.2 degrees 20) at 6.6, 21.4, 10,6, 12,5, 16.2,
  • polymorphic Form II has an XRPD pattern comprising any three degree 20 ⁇ f jections ⁇ +/- 0,2 degrees 2 ⁇ ) selected .from the group consisting of 6.6, 21.4, 10.6, 12.5, 16.2, 14.3, 25.6, and 23,5.
  • XRPD X-ray powder diffraction
  • Polymorphic Form III may exhibit a differential scanning calorimetry (DSC) thermogram substantially as shown in FIG, 8, Polymorphic Form.
  • may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG, 11.
  • Polymorphic- Form III may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 14.
  • polymorphic Form ⁇ at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all of the following (a)-(k) apply: (a) polymorphic Form III has an XRPD pattern substantially as shown in FIG. 3; (b) polymorphic Form III has a DSC thermogram substantially as shown in FIG. 8; (c) polymorphic Form ⁇ has a TGA graph substantially as shown in FIG. 11; (d) polymorphic Form has DVS graphs substantially as shown in FIG.
  • polymorphic Form I has an endothermic event onset
  • polymorphic Form III has a monoelinic crystal system
  • polymorphic Form III has a P2(l) space group
  • polymorphic Form ⁇ has a volume of 3884,0(8) A 3
  • polymorphic Form III has a 2 value of 8
  • polymorphic Form III has a density of 1 ,537 g/cra 3 .
  • polymorphic Form III has at least one, at least two, at least three, or ail of the following properties:
  • polymorphic Form III has an XRPD pattern displaying at least two, at least three, at least four, at least fi ve, or at least six of the degree 2G ⁇ refieetion,s with the greatest intensity as the XRPD pattern substantially as shown in FIG. 3.
  • polymorphic Form III has an XRPD pattern comprising degree 28-reilections (-f/ ⁇ 0.2 degrees 20) at 20.0, 18.5, and 9,6.
  • polymorphic Form III has an XRPD pattern comprising degree 28-reflectioras (+/ ⁇ 0.2 degrees 20) at 20.0, 18,5, and 9.6 and one or more of the degree 29-refkctions (+/- 0.2 degrees 20) at 22.5, 14.0, and 25,0.
  • polymorphic Form III has an XRPD pattern comprising degree 20-re flections (-17- 0.2 degrees 20) at 20.0, 18.5, and 9,6 and one of the degree 28-refiections (+/ ⁇ 0.2 degrees 28) at 22.5 S 14,0, and 25.0.
  • polymorphic Form III has an XRPD pattern comprising degree 2B-reflections (+/- 0,2 degrees 20) at 20.0, 18.5, and 9.6 and two of the degree 28 ⁇ refleciions (+/ ⁇ 0.2 degrees 2 ⁇ ) at 22.5, 14.0, and 25.0.
  • polymorphic Form III has an XRPD pattern comprising degree 28-refiections ( ⁇ -/- 0.2 degrees 20) at 20.0, 18.5, and 9.6 and three of the degree 28- reflections (+/- 0.2 degrees 28) at 22.5, 14.0, and 25.0
  • polymorphic Form HI has an XRPD pattern comprising degree 28 ⁇ refieolions (+/- 0.2 degrees 20) at 20,0, 18.5, 9.6, 22.5, 14.0, and 25.0
  • polymorphic Form III has an XRPD pattern comprising degree 20-reflections (+/- 0.2 degrees 28) at 20,0, 18.5, 9.6, 22,5, .14.0, 25.8, 12.1, and 27.0
  • polymorphic Form 111 has an XRPD pattern comprising any three degree 28 ⁇ ref lections (+/ ⁇ 0.2 degrees 20) selected from the group consisting of 20,0, 18.5, 9,6, 22.5, 1.4,0, 25.0, 12.1, 27.0, 16.2, and 29.0
  • at least one, at least two, at least three, at least four, at least five, at least sis, or all of the following (a)-(g) apply: (a) polymorphic Form IV has an XRPD pattern substantially as shown in FIG, 4; (b)
  • polymorphic Form IV has at least one, or all of the following properties:
  • polymorphic Form IV has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 29 ⁇ reiIectio «s with the greatest Intensity as the XRPD pattern substantially as shown in FIG, 4,
  • polymorphic Form IV has an XRPD pattern comprising degree 26 ⁇ reflections ( ⁇ * ⁇ /- ⁇ .2 degrees 20) at 16,3, 6.2, and 8,6,
  • polymorphic Form. IV has an XRPD pattern comprising degree 28 ⁇ reflections ⁇ +/- 0.2 degrees 20) at 16.3, 6.2, and 8,6 and one or more of the degree 20 ⁇ refiections ( ⁇ -/- 0.2 degrees 26) at 22.7, 22.3, and 25.8.
  • polymorphic Form IV has an. XRPD pattern, comprising degree 20-re flections ( ⁇ ⁇ -/- 0.2 degrees 20) at 16.3.
  • polymorphic Form IV has an XRPD pattern comprising degree 20-reflectIons (+/- 0,2 degrees 20) at 16,3, 6.2, and 8.6 and two of the degree 28-refleetions (+/- 0,2 degrees 20) at 22.7, 223, and 25.8.
  • polymorphic Form IV has an XRPD pattern comprising degree 20-reflections (+/- 0,2 degrees 28) at 16,3, 6.2, and 8,6 and three of the degree 20- reflections (+/- 0,2 degrees 2 ⁇ ) at 22.7, 22.3, and 25.8.
  • polymorphic Form IV has an XRFD pattern comprising degree 2G-re.0ecisons (+/- 0.2 degrees 20) at 16,3, 6.2, 8.6, 22.7, 22,3, and 25.8.
  • polymorphic Form ⁇ has an XRPD pattern comprising degree 2e ⁇ refleetions (+/- 0.2 degrees 20) at 16,3, 6.2, 8.6, 22.7, 22,3, 25.8, 20.0, arid 8.7
  • polymorphic Form IV has an XRFD pattern comprising any three degree 26-reflections ( ⁇ * ⁇ /- 0.2 degrees 28) selected from the group consisting of 16.3, 6.2, 8.6, 22.7, 22.3, 25.8 20,0, 18.7, 27.7, and 13.2,
  • polymorphic Form V at least one, at least two, at least three, at least four, at least five, or all of the following, (a)-(f) apply:
  • polymorphic Form V has a triclinic crystal system;
  • polymorphic Form V has a 1*2(1) space group; (d) polymorphic Form V has a volume of 970.18(14) A 3 ; (e) polymorphic Form V has a Z value of 2; a d (f) polymorphic Fonts V has a density of 1.573 Mg m 3 .
  • polymorphic Form V has the following properties:
  • polymorphic Form VI has the following properties:
  • polymorphic Form VII at least one. at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply;
  • polymorphic Form VII has a monoclinic crystal system;
  • polymorphic Form VII has a P2(l) space group;
  • polymorphic Form Vi has a votome of 12666(8) A ' ';
  • polymorphic Form VII has a Z value of 24; and
  • polymorphic Form VII has a density of 1.468 Mg.1 ⁇ 2 3 .
  • polymorphic Form VI has the following properties:
  • polymorphic Form VOL at least one, at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply;
  • polymorphic Form VIII has a monoclinic crystal system;
  • polymorphic Form VIII has a C2 space group;
  • polymorphic Form VIII has a volume of 1913.9(6) A 3 ;
  • polymorphic Form VIII has a Z value of 4; and
  • polymorphic Form VIII has a density of 1.560 Mg m 3 .
  • polymorphic Form VET has the following properties:
  • a form of sodium (2R,SS s I3aR)-7,9-dioxo-I0- ⁇ (2 5 4 5 6 rifliioroberizyl)caibamoyl)- 2,3,4,5,7,9,13,13a ⁇ aetahydro-2, 5 -methanopyrido [ 1 ⁇ 2':4,5]pyrazino 2., 1 -b] 1 ,3 ]oxazeph 8- oiate may be an intermediate to the synthesis of sodium (2R,5 S, 13aR) ⁇ 7,9-dioxo- 10-((2,4,6- trifl «oroben3 ⁇ 44)carbaffioyl)-2,3,4,5,7,9 s 13 s 13a ⁇ oetahydro-2,5 ⁇
  • 2 5 5 ⁇ msthanopyrido[1 ⁇ 2 ! :4,5]pyra2;ino[2,l-h][l,3]oxazepin-'8 ⁇ oIate may be the final product in the synthesis of sodium (2R,5 S, 13 aR)-7,9-dsoxo- 10-((2,4,6 ⁇ trifluorobenzyi)carbamoyl)- 2,3,4,5,7.9, 13,13a ⁇ octahydro ⁇ 2,5-me&anopyrido[ 1 s ,2 ! :4,5]pyrazino(2, 1 -b][ 1 ,3]oxazepin-8- olate.
  • a polymorphic form or polymorph or eocrystal may have properties such as bioavailability and stability at certain conditions that may be suitable for medical or pharmaceutical uses,
  • a crystalline form of sodium (2R s 5SJ 3a )-7,9-dioxo-10-((2,4,6- trifluoroben yl)carbamoyl)-2,3,4,5J J 9 9 13, 13a-oetahydro-2,5- methanopyrido[r i 2':4 ; ,5]pyraziuo[2J-bl[! ! 3]oxazepin-8-okte may provide the advantage of bioavailability and stability, suitable for use as an active Ingredient in a pharmaceutical composition.
  • a crystalline forro sodium (2R,5S, 13aR)-7,9-dioxo- 10- ( ⁇ 2 , ,6-trifluorobenxy i)carbamoyi) » 2,3,4 J 5,7,9, 13, i 3a-octahydro-2,5- inethanop>Tidoi l',2 , :4,5]pyrazirio[2, 1 -b] ⁇ l,3]oxazcpu 8-olate provides an advantage of improved bioavailability (Table 3) and/or stability (Table 4), Variations in die crystal structure of a pharmaceutical drug substance or active ingredient may affect the dissolution rate (which may affect bioavailability, etc.), anufaciurabi ⁇ ity (e.g., ease of handling, ability to consistently prepare doses of known strength) and stability (e.g., thermal stability, shelf life, etc) of a pharmaceutical drug product or active ingredient.
  • Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or deliver ⁇ ' forms, such as solid oral dosage form including tablets and capsules.
  • crystalline forms may provide desired or suitable h groseopidty. particle size controls, dissolution rate, solubility, purity; physical and chemical stability; manufact rabiiity, yield, and/or process control
  • advantages such as: improving the manufacturing process of an active agent or the stability or storability of a drug product form of the compound or an active ingredient, and/or having suitable bioavailability and/or stability as an active agent.
  • the compound name provided above is named using ChemBioDra Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols.
  • the compound may be named or identified with common names, systematic or non- systematic names, The nomenclature systems and symbols that are commonly recognized in the art of chemistry including but not limited to Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (iUPAC), Accordingly the compound structure provided above may be named or identified as sodium (2R,5S 5 13aR ⁇ -7,9 ⁇ dioxo-10- ( ⁇ 2,4,64rifluorobes3 ⁇ 43 ⁇ 4'l)carbasBoyl) ⁇ 2 s 3,4 s 5 3 7,9 J 13,13a-octahydro--2 5 S- meihanopyrido[f 5 2 ! :4,S)pyrazino[2,i-b][l,3joxazepin ⁇ 8-oiate under IUPAC.
  • meihanopyrido[r 5 2 , :4,5]pyrazmo[2 5 ' i ⁇ b][l 5 3]c*xa2ep ⁇ -3 ⁇ 4-olats 8 having the following structure (Formul II) are disclosed:
  • crystalline forms of sodium (2R,5S s 13aR)-7 s 9 ⁇ di !xo-10- ((2,4,6 ⁇ trifiuoroberi2yi)cafba£noyl)-2,3 s 4,5 s 7 5 9 s 13, 13a ⁇ octahydro-2 s 5- methanop rido[l V2':4,5]pyra;dri [2,l-b][l J]c>xazepin-S-olate are disclosed.
  • Polymorphic sodium Form I may exhibit a differential scanning ca!orimetry (DSC) thermogram substantially as shown in FIG, 9, Polymorphic sodium Form I may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG. 12, Polymorphic sodium Form I may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 15,
  • polymorphic sodium Form I at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or all of the following (a)-(j) apply: (a) polymorphic Form I has an XRPD pattern substantially as shown in FIG. 5 and/or FIG, 16; (b) polymorphic sodium Form I has a DSC thermogram substantially as shown in FIG, 9; (c) polymorphic sodium Form I has a TGA graph substantially as shown in FIG.
  • polymorphic sodium Form I has DVS graphs substantially as shown in FIG, IS;
  • polymorphic sodium Form ⁇ has an orthorhombic crystal system;
  • polymorphic sodium Form I has a P212I21 space group;
  • polymorphic sodium Form I has a volume of 3879.2 A 3 ;
  • polymorphic Form I has a Z value of 4; and
  • polymorphic Form I has a density of L61.4
  • polymorphic sodium Form I has at least one, at least two, at least three, at least four, or all of the following properties:
  • polymorphic sodium Form I has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of fee degree 20-refiections with the greatest intensity as the XRPD pattern substantially as shown in FIG. 1 and/or FIG, 8,
  • polymorphic sodium Form I has an XRPD pattern comprising degree 2CJ ⁇ refieetions (-! ⁇ /- 0.2 degrees 2 ⁇ ) at 5.5, 16.1, and 23,3.
  • polymorphic sodium Form I has an XRPD pattern comprising degree 2 ⁇ - reflections (+/- 0,2 degrees 2 ⁇ ) at 5.5, 16.1, and 23.3 and one or more of the degree 20- refleetions (+/- 0,2 degrees 20) at 22.1, 28.5, and 22.5.
  • polymorphic sodium Form I has an XRPD pattern comprising degree IB-reflections (+/- 0.2 degrees 28) at 5.5, 16.1, and 23.3 and one of the degree 29-reflections (+/- 0.2 degrees 20) at 22,1, 28.5, and 22,5,
  • polymorphic sodium Form I has an XRPD pattern comprising degree 28-refleetions (+/- 0.2 degrees 20) at 5,5, 16.1 , and 23,3 and two of the degree 2 ⁇ - reflectkms (+/- 0.2 degrees 20) at 22.1, 28,5, and 22.5.
  • polymorphic sodium Pons I has an XRPD pattern comprising degree 20-refleciions (+/- 0.2 degrees 2 ⁇ ) at 5.5, 16.1, and 23,3 and three of the degree 20-rsSections 0.2 degrees 2 ⁇ ) at 22.1, 28.5, and 22.5.
  • polymorphic sodium. Form I has an XRPD pattern comprising degree 20-reflections ⁇ +/- 0.2 degrees 20) at 5.5, 16,1, 23.3, 22.1, 28.5, and 22.5.
  • polymorphic sodium Form I has an XRPD pattern comprising degree 20- refleciions 0.2 degrees 20) at 5.5, 16.1, 23.3, 22.1, 28.5, 22.5, 19.5, and 26.6
  • polymorphic sodium Form I has an XRPD pattern comprising any three degree 20-reflections (+/- 0.2 degrees 2 ⁇ ) selected from the group consisting of 5.5, 16.1, 23.3, 22.1, 28,5, 22.5, 19.5, 26.6, and 17.9.
  • oiate A form of a potassium ⁇ 2R,5S,13aR)-7,9-dioxo-I0-((2,4,6 ⁇ trifluorobenzyl)earhamoyl)- 2,3,4,5,7,9,13, 13a ⁇ oetahydro-2,5 ⁇ methanopyrido[I ⁇ 2':4,5]pys-azino[24-b][1 .
  • a polymorphic form or polymorph or cocrystal may have properties such as bioavailability and stability at certain conditions that may be suitable for medical or pharmaceutical uses.
  • trifluorobeiizy 3)carbamoyl) ⁇ 2 5 3.4,5,7,9,13, 13 a-ociahy dro-2, 5 - metMnopyrido r,2':4,5]pyrazirio[2, l-b][l,3]oxazepiri-8-oiale may provide the advantage of bioavailability and stability; suitable for use as an active ingredient in a pharmaceutical composition.
  • Variations in the crystal structure of a pharmaceutical drug substance or active ingredient may affect the dissolution rate ⁇ which may affect bioavailability, etc.), manufaet rabllity ⁇ e.g., ease of handling, ability to consistently prepare doses of known strength) and stability ⁇ e.g., thermal stability, shelf life . , etc) of a pharmaceutical drug product or active ingredient
  • dissolution rate which may affect bioavailability, etc.
  • manufaet rabllity e.g., ease of handling, ability to consistently prepare doses of known strength
  • stability e.g., thermal stability, shelf life . , etc
  • Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or delivery forms, sack as solid oral dosage form including tablets and capsules.
  • crystalline forms may provide desired or suitable hygroscopieity, particle size controls, dissolution rate, solubility, purity, physical and chemical stability,
  • (2R,5S, 13a )-7,9-dioxo- 10-i(2 5 4,6 rifluorobeii2 4)carban ⁇ yl)-2,3,4,5,7 i 9 ; 13, 13a-octahydro ⁇ 2 , 5 ⁇ methanopy ki [ 1 ',2 * :4 , 5 ]pyrazi n [2, 1 -b J [ 1 , ]oxazepin ⁇ 8-oiaie may provide advantages such as: improving the manufacturing process of an active agent or the stability or storability of a drug product form of the compound or an active ingredient, and/or having suitable bioavailability and/or stability as an active agent.
  • polymorphic Forms I, II, and III which may exhibit one or more favorable characteristics described above.
  • the processes for the preparation of the polymorphs described herein, and characterization of these polymorphs are described in greater detail below.
  • the compound name provided above is named using ChemBioDraw Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols.
  • the compound may be named or Identified with common names, systematic or non- systematic names.
  • T he nomenclature systems and. symbols that are commonly recognized in the art of chemistry Including but not limited to Chemical Abstract Service (CAS) and Internationa! Union of Pure and Applied Chemistry (iUPAC). Accordingly, the compound structure provided above may be named or identified as potassium.
  • potassium (2R,5S,I 3a )-7,9-dioxo-10 ⁇ ((2,4 5 6- trifiuorobenz 4)carbarnoyl)-2 ; 3 s 4 3 5,7J 5 13,13a ⁇ oetaliydro-2 s 5- methanopyrido[r,2':4 5 5]pyrazlno[2,l-b][l,3]oxazepiri"8"Olate Form II is disclosed.
  • hydrated potassium (2R,5S, 1 3aR)-7,9-dioxo- 10- ((2 > 4,6 Tifluorobenzyl)carbamoyl)-2 s 3 5 4 s 5 s 7,9 3 13, 13a-octahydn 2,5- methaiiopyr Ido[ 1 ',2' :4,5]pyraz.ino[2 5 1 ⁇ b] [ 1 , 3 ] oxazepin-8 -olate Is disclosed.
  • potassium Form I has an orthorhombic crystal system;
  • polymorphic potassium Form I has a P 21 21 2 space group;
  • polymorphic potassium Form I has a volume of 5101.9(4) A 3 ;
  • polymorphic potassium Form I has a Z value of 8; and
  • potassium Form I has a density of 1.498 Mg m 3 ,
  • polymorphic potassium Form 1 has the following properties:
  • polymorphic potassiwm Form ⁇ has a P 21 21 2 space group;
  • polymorphic potassium Form II has a volume of 5126,8(6) A 3 ;
  • polymorphic potassiwm Form II has a Z value of 4;
  • polymorphic potassium Form ⁇ has a density of 1.336 Mg1 ⁇ 2 3 .
  • polymorphic potassium Form II has the following properties:
  • polymorphic potassium Form III at least one, at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply: (a) polymorphic potassium Form ⁇ has a unit cell, as determined by crystal X-ray
  • polymorphic potassium Form HI has a volume of 1230.86 (8) A 3 ;
  • polymorphic potassium Form III has a Z value of 2; and
  • potassium Form IU has a density of 1,483 Mg/m
  • polymorphic potassium Form III has the following properties:
  • metham3 ⁇ 4pyrido[i , ,2 f :4 s S]pyrazmo[2J-b][L3]oxazepine-I O-carbo3 ⁇ 4amide is disclosed.
  • Formula ⁇ citric acid co-crystal has all of the following properties:
  • Formula I fumaric acid co-cry tal s at least one, at least two, at least three, at least four, at least five, or ail of the following (a)-(f) apply;
  • Formula ⁇ fumaric acid co-crystal has a monoclmic crystal system;
  • (c) Formula I fumaric acid co-crystal has a C2 space group;
  • (d) Formula I fumaric acid co-crystal has a volume of 5237.9(16) A 3 ;
  • (e) Formula i fumaric acid co-crystal has a
  • Formula I fumark acid co-crystal has all of the following properties:
  • Formula I oxalic acid co-crystal of (2R,5S J 3aR)- S- ydroxy-T ⁇ -dio ⁇
  • Formula i oxalic acid eo-crystal at least one, at least two, at least three, at least four, at least five, or ail of the following (a)-(f) apply:
  • Formula ⁇ oxalic acid co-crystal has a rnonoclink crystal system;
  • (d) Formula ⁇ oxalic acid co-crystal has a volume of 2231.95(14) A 5 ;
  • Formula I oxalic acid co-crystal has the following properties:
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 29-reflections (+/- 0.2 degrees 29) at 19.1. 14,5, and 9.1.
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 20- refleetions (+/- 0.2 degrees 20) at 1.9.1 , 14.5, and 9,1 and one or more of the degree 29- reflections (+/- 0.2 degrees 2 ⁇ ) at 7.6, 26.5, and 17.1.
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28-reflectlons (+/- 0,2 degrees 2 ⁇ ) at 19.1, 14.5, and 9/1 and one of the degree 20-reflections ( ⁇ ⁇ ⁇ /- 0.2 degrees 2 ⁇ ) at 7.6, 26.5, and 17.1.
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28-reilectiorts (+/- 0.2 degrees 20) at 19.1, 14.5, and 9,1 and two of the degree 20- reflections (+/- 0,2 degrees 2 ⁇ ) at 7,6, 26.5, and 17.1,
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 29-reflections (+/ ⁇ 0.2 degrees 28) at 19.1, 14.5, and 9.1 and three of the degree 20-reflections (+/- 0.2 degrees 2 ⁇ ) at 7.6, 26.5, and 17,1.
  • Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28 ⁇ refleetions ( ⁇ /- 0,2 degrees 2 ⁇ ) at 19.1, 14.5 » 9.1, 7,6, 26.5, and 17,1.
  • Formula 1 oxalic acid co-crystal has an XRPD pattern comprising degree 29- reflections (+/- 0.2 degrees 2 ⁇ ) at 19.1, 14,5, 9.1, 7.6, 26,5, 17.1, 21.8, and 39.4.
  • polymorphic Formula 1 oxalic- acid co-crystal has an XRPD pattern comprising any three degree 20-refiections ⁇ +!- 0.2 degrees 2 ⁇ ) selected from the group consisting of .19.1., 1.4.5, 9.1, 7.6, 26.5, 17.1, 21.8, 39.4, 29.7, and 11.6.
  • compositions of the present Invention comprise a compound of Formulas (I). (I ) or (ill), including forms and co-crystals thereof, and a pharmaceutically acceptable earrier, diluent or excipient
  • the compound of Formulas ( ⁇ ), (II), or (III) is present in the composition in an amount which is effective to treat a particular disease or condition of interest.
  • the activity of compounds of Formulas (I), (II), and (III) can be determined by one skilled in the art, for example, as described in co-pending application. U.S.
  • PHARMACEUTICAL USE Appropriate concentrations and dosages can be readily- determined by one skilled in the art.
  • a compound of Form ulas (I) s (H), and/or (III) is present in the pharmaceutical composition in an amount from about 25 mg to about 500 mg.
  • a compound of Formulas (I), (II), and/or (III) is present in the pharmaceutical composition in an amount of about 100 mg to about 300 mg.
  • a compound of Formulas (I), (II), and/or ( ⁇ ) is present in the pharmaceutical composition in an amoun t of about 5 mg to about 00 mg.
  • a compound of Formulas (I), (IT), and/or (III) is present in the pharmaceutical composition in an amount of about 25 mg to about 100 mg. In certain embodiments, a compound of Formulas (I), (II), and/or (III) is present In the pharmaceutical composition in an amount of about 50 mg to about 1.00 mg. In certain embodiments, a compound of Formula (1), ( ⁇ ), and/or (III) Is present in the pharmaceutical composition in an amount of about 5 mg, 25 mg, 50 mg, 75, mg, 100 mg, 200 mg, 300 mg, 400 mg or about 500 mg.
  • compositions comprising at least one, at least two, at least three, at least four, at least five, at least six, at least seven, or all of polymorphs ⁇ e.g., any one or more of Formula ⁇ polymorphic Forms I, U, III, IV, V, VI, VII and VIII) as described herein.
  • a composition comprising one of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided.
  • a composition comprising two of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided.
  • a composition comprising three of Formula I is provided.
  • polymorphic Forms L II, III, IV, V, VI, VII, and VIII described herein is provided.
  • a composition comprising four of Formula 1 polymorphic Forms 1, XL III, IV, V» VI, VII, and VIII described herein is provided.
  • a composition comprising five of Formula I polymorphic Forms I, II, III. IV, V VI, VII, and VIII described herein is provided.
  • a composition comprising six of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided.
  • a composition comprising seven of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided.
  • compositions described herein comprising eight of Formula I polymorphic Forms I, II, HI, IV, V, VI, VII, and VIII described herein is provided.
  • the compositions described herein may comprise substantially pisre polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
  • the composition comprises a polymorphic form of (2 ,5S S i S ⁇ -g-hydroxy-T ⁇ -dlo o-N-CZ ⁇ -iTiil ioro sn ⁇ 'O ⁇ J ⁇ .SJ ⁇ , 13,13a ⁇ oe ⁇ ahydn 2,5 ⁇ methanopytido[ I ! ,2' :4 5 5
  • compositions comprising a polymorphic form as described herein, wherein the (2R,5S, 1 Sa ⁇ -S-hydrox -T ⁇ -di so-N-C ⁇ -t fluoroben yl) ⁇ ,;? ⁇ ,?, 1 ⁇ , 13,13a- octa ydro-2,5-methanopyrido[ 1 ' s 2 ! :4,S]pyr azino[2, 1 -b] [ 1 5 3]oxa epis e ⁇ 10-carboxamidc within the composition is substantially pure (i.e., substantially pure Form i. Form II, Form Hi, Form ⁇ ', Form V.
  • compositions comprising a polymorphic form of (2R,5S, 13aIt)-8-hydroxy-7,9-dioxo-N- (2,4 s 6-trIf!uorobenzyl)-2,3,4,5 5 7,9 5 i3,I 3a-octahydro-2,5- methanopyrido[1 ⁇ 2';4,5]pyrazlno[2,l-h][l ,3]oxa i;pirie-i0-carboxaniide, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%
  • the carboxamide present in the composition is one of the polymorphic forms disclosed herein.
  • the composition includes at least about 50%, at least about 60%, at least about 70%. at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of one of the polymorphic forms of (2R,5S, 13aR)-8 iydroxy-7,9-dioxo-N-(2,4,6-irif]uorobenzyl)- 2,3,4,5,7,9, .13, 13a-octahydro-2,5-methar!Op>'Tido[i , ,2 , :4,5]pyrazino[2, 1 -b][.l ,3]oxazepke-10 ⁇ carboxamide.
  • compositions comprising a polymorphic form disclosed herein, less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of (2R,5 S, 1 ;1 ⁇ 2R ⁇ -8-hydrox.y-7,9-dioxo ⁇ ⁇
  • impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present
  • im urities may, for example, include by-products from synthesizing (2R S 5 S, 13aR)-81 ⁇ 2droxy-7.9-dioxo ⁇ - ⁇ 2A ⁇
  • impurities include by-products from the process of synthesizing i2R,5S 5 13aR) ⁇ 8-hydroxy-?,9 ⁇ dio3 ⁇ 4o ⁇ N-(2,4 s 6 ⁇ trifto ⁇
  • Impurities include contaminants from the process of syHthesteing (2R,5S 3aR) ⁇ 8-hydrGx
  • impurities include degradation products of
  • impurities include other polymorphic forms of (2R,5S,13aR)-8-hydrox r y-7 s 9- dioxo-N"(2,4 ⁇ 6 rif!uorobenzyi) ⁇ 2 s 3 ; 4,5,7,9 5 13, 13a-octahydro--2,5- methanopyrido[ 1 ',2' :4, 5]pyrazino[2, I -b] [ I,3]oxazepine- !
  • impurities include water or solvent.
  • impurities are selected from the group consisting of by-products from synthesizing (2R,5S,13aR) ⁇ S ⁇ hydroxy ⁇ 7 s 9 ⁇ dioxo ⁇ M ⁇ (2 s 4,6- trifjuorobenzy!)-2,3 s 4,S,? ⁇
  • the composition comprising a polymorphic form disclosed herein has less than about 5%, less man about 4%, less than about 3%, less than about 2%. or less than about 1% by weight of amorphous or non -crystalline (2R,5S, 13aR)-8- d o ⁇ -dio o-N ⁇ .
  • compositions comprising at least one polymorph ⁇ e.g., any one or more of Formula II polymorphic Forms ⁇ ) as described herein.
  • a composition comprising Formula ⁇ polymorphic Form 1, described herein is provided, in other embodiments, the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities,
  • the composition comprises a polymorphic form of sodium (2R,5S 5 13aR) ⁇ 7,9-dioxo ⁇ 10-( ⁇ 2 3 4 : ,6-trifJuoroberiz 'l)carbanioyl) ⁇ 2 s 3,4 s 5,7 5 9, 13,13a ⁇ oetahydro- 2,5"meihanopyrido[1 ⁇ 2 , :4,5]pyrazmo[2,l-b3[l,31oxazepin-8 «oiate.
  • compositions comprising a polymorphic form as described herein, wherein the sodium (2R,SS 3aR)-7 s 9-dioxo-10-((2 5 4,0-tri ⁇
  • compositions comprising a polymorphic form, of sodium (2R s 5SJ3aR)-7,9 ⁇ dioxo-lG- ( ⁇ 2 A6 ⁇ tr3f1uorobenzy l)eatbamoyl)-2,3 AS.,7,9, 13, 13a ⁇ octahydro-2,5 ⁇
  • meihanopyrido[r s 2 , :4 ? 5]pyrazmo[2,l-b][.U3]oxazepin-8-olate at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% ot ⁇ sodium (2R,SS,13aR) ⁇ 7 s 9-d ⁇
  • the composition includes at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least, about 98%, or at least about 99% of Form I of sodium (2R,5S,!3aR)-7,9 ⁇ dioxO"10-((2,4,6- ⁇
  • compositions comprising a polymorphic form disclosed herein, less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of sodium (2R,5S, 13aJl)-7,9-dioxo ⁇ i 0-((2,4.6- trifluorohenzyi)cai-bamoyl)-2,3,4,5,7,9, 13,13a ⁇ octahydro-2,5- methaiiopysido[l ! ,2 ?
  • composition are other polymorphs of sodium (2R,5S 5 13aR)-7,9"dioxo ⁇ iO-((2,4,6-triOuorohenzyl)carbanioyl) ⁇ 2,3,4,5,7,9, 13, 13a ⁇ oetahydro-2,5-metbanopyrido[ l ! ,2 f :4,5]pyrazino[2, 1 -b][1 ,3]oxazepin ⁇ 8 ⁇ elate and/or impurities.
  • impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present.
  • Impurities may. for example, include by-products from synthesizing sodium (2 s 5SJ3a )-7,9-diaxo-10-((2,4,0 ⁇
  • impurities include by-products from the process of synthesizing sodium ⁇ 2R,5S i 13aR.)-7 5 9"dIoxo-10- ⁇ (2,4,6 rifluoroben£yi)carbamoyi)- 2.3,4 J,7,9 ! i343a-octahydro-2,5-Biethanopyrido 3 2':4,5]pyi-azino[2, l ⁇ b][!,3]oxazepm-8- olate.
  • impurities include contaminants from the process of synthesizing sodium (2R,5S, 33aR)-7,9-dioxo- 10-((2A6-triftuorobenzy ⁇ carbam ⁇
  • impurities include degradation products of sodium
  • impurities include other polymorphic forms of sodium (2R,5S, 13aR ⁇ 7,9-dk>xo-10 ⁇ ((2,4,6 - trifluorobefizyl)carbamoyl)-2,3,4,5,? 5 9, 13,13a-ociahydro-2,5- methan ridol ⁇ '. ⁇ jpyra irjo ⁇ j l-bjl l ⁇ joxaze isi-S-olaie,
  • impurities include water or solvent.
  • impurities are selected from the group consisting of by* products from synthesizing sodium (2 ,5S,13aR)-7,9 ⁇ dioxo-I0-((2,4,6- trifluoroben2;yl)carbamoyl)-2 3 3,4 s 5,7 s 9,13 s l3a-octahydro-2 5 5 ⁇
  • the composition comprising Formula II, Form I disclosed herein has less than about 5%, iess than about 4%. less than about 3%, less than about 2%, or less than about 1% by weight of amorphous or non-crystalline sodium
  • the term "substantially pure” or “substantially free” with respect to a particular polymorphic form of a compound means that the composition comprising the polymorphic form contains less than 95%, less than 90%, less than 80%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 40%, less than 30%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% by weight of other substances, including other polymorphic forms and/or impurities.
  • "substantially pure” or “substantially free of refers to a substance free of other substances, including other polymorphic forms and/or impurities. Impurities may, for example, include by-products or left over reagents from chemical reactions, contaminants, degradation products, other polymorphic forms, water, and sol vents.
  • compositions comprising at least one, or all of polymorphs ⁇ e.g. , any one or more of Formula III polymorphic Forms I, II, and III) as described herein.
  • a composition comprising one of Formula III polymorphic Forms I, II, and III described herein is provided.
  • a composition comprising two of Formula ⁇ polymorphic Forms I, II, and III described herein is provided,
  • the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
  • the composition comprises a polymorphic form of potassium (2R,5S, 13aR)-7,9 ⁇ dioxo-l O- ⁇ -rulluorobe ⁇ ca b moyl) ⁇ ,? ⁇ , 13,13a- oe-tahydro ⁇ 2,5-methanopyrido[! ⁇ 2':4,S]p ⁇
  • compositions comprising a polymorphic form as described herein, wherein the potassium (2R,5S, 13aR) ⁇ ?,9 ⁇ dioxo- 10- ⁇ (2,4,6-trifluorobenz l ⁇ c6.rba oyl)- 2 s 3A5,7,9 5 I3,13a ⁇ Q Ctahydr0-2,5 ⁇ meta
  • compositions comprising a polymorphic form of potassium (2R,5S, 13aR)-7,9-dioxo- 10-i(2 5 4,6-trifluoroben:3 ⁇ 44)eaA ⁇
  • octahydro ⁇ 2,5-metha3 ⁇ 4opyrido[r,2':4,5]pyraK?no[2, l-h ' ][l ,3]oxa ⁇ pin ⁇ 8 ⁇ olate at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of potassium (2 5 5S,13aR)-7,9-dioxo-10-( ⁇ 2,4 5 6- irifiuoroben l)carbamoyi)-2 5 3 :i 4,5,7,9,13,13a-octahydro-2 ;!
  • the composition includes at least, about 50%. at least about 60%, at least about 70%, at least about 80%, at least about 85%. at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of one of the polymorphic forms potassium (2 ,5S, 13aR)-7,9 ⁇ dioxG- 10-((2 ,5S, 13aR)-7,9 ⁇ dioxG- 10-((2
  • compositions comprising a polymorphic form disclosed herein, less than about 50%. less than about 40%, less than about 30% 5 less than about 20%, less than about 10%, less than about 5%. less than about 4%, less than about 3%, less than about 2% or less than about 1% of potassium (2R 5 5S,13aR)-7,9-dioxo-10-((2 ! 4,6- trifiuorobe3 ⁇ 1)carhaBioyi)-2,3,4 ?
  • impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present, impurities may, for example, include by-products from synthesizing potassium (2R.5SJ 3aR) ⁇ 7,9 ⁇ diGXQ-10-((2,4 s 6 riflu ⁇
  • impurities include by-products from the process of synthesizing potassium (2R 5 5 S, 13 aR)-7,9 ⁇ dioxo- 10-((2,4,6»trifluorobenzyl)carbamoyi)-2 5 3 ,4, 5 ,7,9, 13,13a- octah ⁇ 'dro-2 s 5-metha.nopyrido[r,2 i ;4 J 5]pyTazino[2,l-b][l s 3]oxazepk-8-oiate.
  • impurities include contaminants from the process of synthesizing potassium (2R,5S, 13aR)-7,9-dioxo- 10-((2,4 s 6-trifluofoben2yl)earbamoyl)-2 5 3,4,5 5 7,9, 13, 13a ⁇ octahydro ⁇ 2,5"S3 ⁇ 4ethanopyrido[r,2 , :4,5]pyrazino[2, i ⁇ b][ 1 ,3]oxazeph ⁇ -olate.
  • impurities include degradation products of potassium (2R.5S, 13aR) « ?,9 ⁇ dioxo - 10- ⁇ (2,4,6- U-ifluorobeRzy!carbamoy])-2,3,4,S,7,9 > 13, 13a ⁇ oeiahydro-2,5 ⁇
  • impurities include other polymorphic forms of potassium (2R «5S, i 3a ) ⁇ 7,9-dioxo ⁇ I0 ⁇ ( ⁇ 2,4,6- trifl uorobenzyl) arbamoyl) ⁇ 2, 3,4,5,7,9,13,13 a-octahydro-2,5 - methaiK)p rido ,2':4.S]p azmo[2,1 ⁇ b][l ,3]oxazepin-8-oiate.
  • Impurities include water or solve t
  • impurities are selected from, the group consisting of byproducts from synthesizing potassium (2 ,5S,13aR) ⁇ 7,9-"dioxo ⁇ 10 ⁇ 2,4,6 ⁇
  • the composition comprising a polymorphic form disclosed herein has less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% by weight of amorphous or non-crystalline potassium (2R,5S.13aR) ⁇ 7,9-dioxo- 10-((2,4,6-trifi orobeii3 ⁇ 4'l)carbamoyl)-2,3,4,5,7 s 9 ! 13,13a-octahydro- 2 J 5»methanopyrido[lV2 ! :4,5]py a7ino[2,l "b][l,3]oxazepin ⁇ 8--olaie.
  • compositions comprising at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or ail of polymorphs ⁇ e.g., any one or more of Formula I polymorphic Forms I, II, ill, IV, V, VI, VI , and VIII, Formula II polymorphic Form I, and/or Formula IH polymorphic Forms 1, 11, aud III) as described herein.
  • a composition comprising one of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form 1, and/or Formula III polymorphic Forms 1, 11, and III described herein is provided.
  • a composition comprising two of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form ⁇ , and/or Formula III polymorphic Forms I, II, and III described herein is provided,
  • a composition comprising three of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form 1, and/or Formula II polymorphic Forms I, II, and III described herein is provided,
  • a composition comprising four of Formal Formula 1 polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms L
  • a composition comprising five of Formula I polymorphic Forms I, II, IIL IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided.
  • a composition comprising six of Formula I polymorphic Forms I, ⁇ , III, IV, V, VI, VII, and VEIL Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided.
  • composition comprising seven of Formula I polymorphic Forms I, ⁇ , III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and HI described herein is provided.
  • compositions comprising eight of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic forms I, II, and III described herein is provided, In a particular embodiment a composition comprising nine of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I s and/or Formula 111 polymorphic Forms I, II, and III described herein is provided.
  • compositions comprising ten of Formula I polymorphic Forms L ⁇ , III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and HI described herein is provided.
  • a composition comprising eleven of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided.
  • the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
  • the tem "substantially pure” or “substantially free” with respect to a particular polymorphic form of a compound means that the composition comprising the polymorphic form contains less than 95%, less than 90%, less than 80%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 40%, less than 30%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% by weight of other substances, including other polymorphic forms and/or impurities.
  • "substantially pure” or “substantially free of 5 refers to a substance free of other substances, including other polymorphic forms and/or impurities. Impurities may, for example, include by-products or left over reagents from chemical reactions, contaminants, degradation products, other polymorphic forms, water, and solvents.
  • compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as solid dispersions and solid solutions.
  • Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal.
  • the pharmaceutical compositions is prepared for oral administration.
  • the pharmaceutical composition is a tablet.
  • Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the compositio to a patient.
  • compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units.
  • dosage forms are known, or will be apparen t, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000),
  • the composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention for treatment of a disease or condition of Interest in accordance with the teachings of this invention,
  • compositions of the invention may be prepared by methodology well known in the pharmaceutical art.
  • a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution, A surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalentiy interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous de lively system.
  • a solid pharmaceutical composition intended for oral administration can be prepared by mixing a compound of the invention with at least one suitable pharmaceutical excipient to form a solid eformulation composition, which then may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • a pharmaceutical composition is provided, which includes a compound of Formula ( ⁇ ), (II), or (III) and a pharmaceutical exe-ipient.
  • the compounds of the invention are administered in a therapeutically effecti ve amount which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy, in some embodiments, the compounds of the in ention can be administered alone or is combination with other antiviral agents once or twice daily for as long as the patient is infected, latently infected, or to prevent infection (e.g. for multiple years, months, weeks, or days).
  • a method for treating or preventing an HIV infection in a human having or at risk of having the infection comprising administering to the human a therapeutically effective amount of a compound disclosed herein in combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • a method for treating an HIV infection in a human having or at risk of having the infection comprising administering to the human a therapeutically effective amount of a compound disclosed herein in combination with a therapeutically effective amount of one or more (e.g., one. two, three, one or two, or one to three) additional therapeutic agents.
  • a method for treating an HIV infection in a human having or at risk of having the infection comprising administering to the human a therapeutically effective amount of a compound or composition disclosed herei In combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • a therapeutically effective amount of a compound or composition disclosed herei In combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • the present invention provides a method for treating an HI infection, comprising administering to a patient in need thereof a therapeutically effective amount of a compound or composition disclosed herein in combination with a therapeutically effective amount of one or more additional therapeutic agents which are suitable for treating an HIV infection.
  • One embodiment provides a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents for use in a method for treating or preventing an HIV infection in a human having or at risk of having the Infection,
  • One embodiment provides a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents for use in a method for treating an HIV Infection in a human having or at risk of having the infection.
  • One embodiment provides a compound disclosed herein for us in a method for treating or preventing a HIV infection in a human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • One embodiment provides a compound disclosed herein for use in a method for treating an HIV infection in a human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • the present invention provides a compound disclosed herein in combination with one or more additional therapeutic agents which are suitable for treating an HIV infection, for nse in a method for treating an HIV infection, in certain embodiments, the present invention provides a compound disclosed herein for use in a method for treating an HIV infection, wherein the compound is administered in combinatio with one or more additional therapeutic agents which are suitable for treating an HIV infection.
  • One embodiment provides the use of a compound disclosed herein thereof, in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents in the manufacture of a medicament for treating or preventing an HIV infection in a human having or at risk of having the Infection.
  • One embodiment provides the use of a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents in the manufacture of a medicament for treating an HIV infection in a human having or at risk of having the infection.
  • One embodiment provides the use of a compound disclosed herein in the manufacture of a medicament for treating or preventing an HIV infection in a human having or at risk of having the Infection, wherein the compound Is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
  • One embodiment provides the use of a compound disclosed herein thereof, in the manufacture of a medicament for treating an HIV infection in human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents, in certain embodiments, the present invention provides the use of a compound disclosed herein thereof, in
  • the present invention provides the use of a compound disclosed herein thereof for treating an HIV infection, wherein the compound is administered in combination with one or more additional therapeutic agents which are suitable for treating an HIV infection.
  • a compound as disclosed herein may be combined with one or more additional therapeutic agents in any dosage amount of the compound of Formulas ( ⁇ ), (II), and/or (III) (e.g., from 50 mg to 1000 mg of compound),
  • compositions comprising a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents, and a pharmaceutical ly acceptable carrier, diluent or exeipient are provided.
  • combination pharmaceutical agents comprising a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents are provided.
  • kits comprising a compound disclosed herein in combination with one or more (e.g., one. two, three, one or two, or one to three) additional therapeutic agents are provided.
  • the additional therapeutic agent may be an aod-HIV agent.
  • the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nndeoside inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV mtegrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gp!20 inhibitors, G6PD and NA.DH-oxida.se inhibitors, compounds that target the HIV capsid ("capsid inhibitors"; e.g., capsid polymerization inhibitors or capsid disrupting compounds such as those disclosed Irs WO 2013/006738 (Giiead Sciences),
  • the additional therapeutic agent may be an and- HIV agent.
  • the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nuc!eoside or non-nucleoiide inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or aliosteric) integrase inhibitors, HIV entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gp!20 inhibitors, G6PD and NADH-oxidase Inhibitors, HIV vaccines, HIV maturation inhibitors, latency reversing agents (e.g., histone deacety!ase inhibitors, proteasoroe Inhibitors, protein kinase C (PKC
  • capsid polymerization inhibitors or capsid disrupting compounds HIV nucleocapsid p7 (NCp?) inhibitors, HIV p24 capsid protein inhibitors), pharmacokinetic enhancers, immune-based therapies (e.g., Pd-1 modulators, Pd-Ll modulators, toil like receptors modulators,, IL-15 agonists, ), HIV antibodies, bispeeific antibodies and "antibody-like" therapeutic proteins (e.g., DARTs®, Duobodies®, Bites®, XmAbs®, TandAbs ®, Fab derivatives) including those targeting HIV gp!20 or gp4i, combination drags for HIV, HIV pi 7 matrix protein inhibitors, IL-13 antagonists, Peptidyl -prolyl cis-trans isoraerase A modulators.
  • Pd-1 modulators e.g., Pd-Ll modulators, toil like receptors modulators,, IL-15 agonists
  • HIV antibodies bispe
  • Protein disulfide isomera.se inhibitors, Complement C5a receptor antagonists, DNA methyitransferase inhibitor, HIV vif gene modulators, Vif dhiierization antagonists, HW-1 viral infcetivity factor inhibitors, TAT protein inhibitors, HIV-1 Nef modulators, Hck tyrosine kinase modulators, mixed lineage kinase-3 (MLK-3) inhibitors, HIV-l splicing inhibitors, Rev protein Inhibitors, Integrin antagonists, Nucleoprotein inhibitors, Splicing factor modulators, COMM domain containing protein 1 modulators, HIV ibonuclease H inhibitors, Retrocyelin modulators, CDK-9 inhibitors, Dendritic ICAM-3 grabbing nonintegrln 1 inhibitors, HIV GAG protein inhibitors, HIV POL protein inhibitors, Complement Factor H modulators, Ubiquitin ligase inhibitors, Deoxycytidme kinase inhibitors, Cy
  • the additional therapeutic is selected from tbe group consisting of HIV protease inhibitors, H1Y non-nucleoside or non-nucleotide inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or allosterie) integrase Inhibitors,
  • a compound of Formulas (I), (II), and/or (III) is formulated as a tablet, which may optionally contain one or more other compounds useful for treating HIV.
  • the tablet can contain another active ingredient for treating HIV, such as HIV protease inhibitors, HIV non-nucleoside or non-nucleotide Inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or allosterie) integrase inhibitors, pharmacokinetic enhancers, and combinations thereof.
  • the tablet can contain one or more active ingredients for treating HIV, such as HIV nucleoside or nucleotide Inhibitors of reverse transcriptase.
  • such tablets are suitable for once daily dosing,
  • the additional therapeutic agent is selected from one or more of:
  • HIV nucleoside or nucleotide inhibitors of reverse transcriptase selected from the group consisting of zidovudine, emfridtahine, didanosine, stavudins, xale-iiabine, lamivudine, abacavir, abacavir sulfate, aradoxovir, elvucit&bine, alovudine, MIV-210, ⁇ -FTC, D ⁇ d4FC, emtricitabine, phosphatide, fozivudine tidoxil, aprieitlbine (AVX754), KIM 461, GS-9131
  • HIV integrase inhibitors selected from the group consisting of ciircumin, derivatives ofcurcumin, chicoric acid, derivatives of chicoric acid, 3,5-dicafFeoylquinic acid, derivatives of 3,5-dicaffeoyiquInie acid, aurintriearboxyfie acid, derivatives of
  • aarintricarboxylic acid caffeic acid phenethyl ester, derivatives of caffeie acid phenethyi ester, iyrphostin, derivatives of iyrphostin, quercctin, derivatives of quercetin, S-1360, AR- 177, L-870812, and L-870810, raitegra , BMS-538158, GSK364735C, BMS-707035, MK- 2048, BA 01 1, elvitegravir, dolutegravir, dolutegravir sodium, and GSK-744;
  • NTNI HIV fion-catalyiic .site, or allosterk, integrase inhibitors
  • gp4i inhibitors selected from the group consisting of enfuvirtide, sifuvtrtide, a!buvirtlde, FB006M, and TRM 144;
  • CCRS Inhibitors selected from the group consisting of aplaviroc, vicriviroc, maraviroc, ccnicriviroc, PRG-I 40, INCB.I 5050, PF-232798 (Pfizer), and CCR5mAb004;
  • CD4 attachment inhibitors selected from the group consisting of ibalizumab (TMB-355) and BMS-068 (BMS-663068);
  • phamiacokmetic enhancers selected from the group consisting of cobicistai and SPI-452;
  • other drugs for treating HIV selected from the group consisting of BAS-I00, SPI- 452, REP 9, SP-Gi A, TNX-3SS, DES6, ODN-93, ODN-112, VGV-1, PA-457 (bevMmat), HRG214, VGX-410, D-247, AMZ 0026, CYT 99007A-221 HIV, DEBiO-025, BAY 50- 4798, MDX010 (ipilimumab), PBS 119, ALG 889, and PA-1G5QQ40 (PA-040), and combinations thereof.
  • drugs for treating HIV selected from the group consisting of BAS-I00, SPI- 452, REP 9, SP-Gi A, TNX-3SS, DES6, ODN-93, ODN-112, VGV-1, PA-457 (bevMmat), HRG214, VGX-410, D-247, AMZ 0026, CYT 99007A-221 HIV, DEBiO-025, BAY 50-
  • die additional therapeutic agent is selected from one or more of:
  • HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavir, fesamprenavir calcium, indinavir, indinavir sulfate, lopinavir, ritonavir, nelflnavir, nelfinavir mesylate, saquinavir, saquinavir mesylate, tipranavir, breeanavir, darunavir, DG-17, TMB-657 (FPL-100) and T C-31091 1 ;
  • HIV non-nuc!eoside or non-nncleotide inhibitors of reverse transcriptase selected from the group consisting of delavirdine, delavirdine mesylate, nevirapine, etravirine, dapivirine, doravirine, rilpivirine, efavirenz, M-G23, VM-1500, lentinan and AIC-292;
  • HIV nucleoside or nucleotide inhibitors of reverse transcriptase selected from the group consisting of VIDEX® and VIDEX® EC (didanasine. ddl) 5 zido vudine, emtricitabine, dldanosine, stavudine, zalcitabine, lamivudine, censavudine, abacavir, abacavir sulfate, amdoxovir, el ucitabine, alovudine, phosphazid, fozivudine tidoxil, aprkitabine, amdoxovir .
  • KP-1461 fosa!vudine tidoxil, ienofovir, tenofovir disoproxil, ienofovir disoproxil fumarate, tenofovir disoproxil hemifi.mia.rate, tenofovir alafenamide, tenofovir alafenamide
  • HIV integrase inhibitors selected from the group consisting of ewcwniin, derivatives of curcu in, chicoric acid, derivatives of chicoric acid, 3 ,5 -dicaffeoylquin ic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxyiic acid, derivatives of
  • aurintricarboxyiic acid caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of lyrphostin, qoercetin, derivatives of quercetin, raltegravir, dvitegravir, do!utegravir and eabotegravir;
  • NCINI HIV non-catalytic site, or allosterie, integrase inhibitors (NCINI) selected from the group consisting of CX-05168, CX-05045 and CX-14442;
  • HIV gp4.l inhibitors selected from the group consisting of enfovixtide, sifuvirtide and albuvirtide; (8) HIV entry inhibitors selected from the group consisting of cenicriviroc;
  • HIV g i O inhibitors selected from the group consisting of Radha-108 (Reeeptol) and BMS-663068;
  • CCR.5 inhibitors selected from the group consisting of apiaviroc, vicriviroc, rnaraviroe, cemcriviroc, PRO- 140, Adaptavir (RAP-J O 1 ), nifeviroc (TD-0232), TD-0680, and vMIP (Haimipu);
  • CD4 attachment inhibitors selected from the group consisting of ibalkuniab;
  • CXCR4 inhibitors selected from the group consisting of plerixafor, ALT- 1188, vMIP and Hairnipu;
  • Pharmacokinetic enhancers selected from the group consisting of cobicistat and ritonavir;
  • Immune-based therapies selected from the group consisting of dermaVir, interkukin-7, plaqnenll (hydroxychloroquine), proleukin (aldesleukin, if . -2), interferon alia, interferon alfa-2b, interferon alfa-n3, pegylated interferon alfa, interferon gamma, hydroxyurea, myeoplienoiate mofetil (MP A) and its ester derivative mycoptienolate mofetil (MMF), WF-10, ribavirin, l ' L-2, IL-12, polymer polyeffayleneimine (PEI), Gepon, VGV-1, MOR-22, BMS-936S59, toll-like receptors modulators (tkl, ilr2, tlr3, tir4, tlr5, tlr6, tlr7, tlrS, tir9, tlrlO, tir1 1 , tirl2 and t
  • HIV vaccines selected from the group consisting of peptide vaccines, recombinant subunit protein vaccines, live vector vaccines, DNA vaccines, virus-like particle vaccines (pseudovirion vaccine), CD4-derived peptide vaccines, vaccine combinations, rgpl2Q (AIDSVAX), ALVAC HIV (vCP! S2i)/AIBSVAX B/E (gp!20) (RV144), monomelic gp l 20 IHV-1 subtype C vaccine (Novartis), Remnne, ITV-l . Contre Vir, Ad5 ⁇ ENVA-48, DCVax- 001 (CDX-24G1), PEP-6409,Vacc-4x.
  • TVI-HIV-1, Ad-4 Ad4-env Clade C + Ad4-mGag
  • Ad-4 Ad4-env Clade C + Ad4-mGag
  • PrcVaxTat TL-01
  • SAV- 001 SAV- 001
  • AE-H SAV- 001
  • MYM-YIGl CombiHIVvac
  • ADVAX ADVAX
  • MYM-V201 MVA-CMDR
  • ETV-01 CDX-I4G1, rcAd26.MOS l .HiV-Env and DNA ⁇ Ad5 gag/pof/nef/nev (HYTN505);
  • HIV antibodies, bispecific antibodies and "antibody-ilke” therapeutic proteins such as DARTs®, Duobodies®, Bites®, XmAbs®, TandAbs ® strictly Fab derivatives
  • BMS-936559, TMB-360 and those targeting HIV gp!20 or gp41 selected from the group consisting of bavituximab, UB-42L C2F5, C2G12, C4E10, C2F5+C2G12+C4E10, 3-BNC- 1 17 , PGT145, PGT12L DX01 Q (ipilim ma ).
  • latency reversing agents selected from the group consisting of Histone deaeetylase inhibitor such as Romidepsin, vorinostai panobinostat; Proteasome inhibitors such as Yeleade; protein kinase C (PKC) activators such as indolaetam, Prostratin, Ingenol B and DAG-lactones, Ionomycin, GS -343, PMA, SAHA, BRD4 inhibitors, IL-15, JQI , disulfram, and amphotericin B;
  • Histone deaeetylase inhibitor such as Romidepsin, vorinostai panobinostat
  • Proteasome inhibitors such as Yeleade
  • protein kinase C (PKC) activators such as indolaetam, Prostratin, Ingenol B and DAG-lactones, Ionomycin, GS -343, PMA, SAHA, BRD4 inhibitors,
  • Np7 HIV nucleocapsid p7 (NCp7) inhibitors selected from the group cons sting of azod carbonamide
  • HIV maturation inhibitors selected from the group consisting of BMS-955176 and GSK-2838232;
  • PI3K inhibitors selected from the group consisting of sacredalisib, AZD-8186, bupariisib, CLR-457, pictilisib, neratinib, rigosertib, rigosertib sodium, EN-3342, TGR-1202, aipelisib, duvelisib, UCB-5857, tase!isib, XL-765, gedatoiisib, VS-5584, copanlisib, CA?
  • (22) other drugs for treating HIV selected from the group consisting of BanLee, MK ⁇ 8507, AG- 1105, TR-452, M -8591 , REP 9, CYT-107, alisporivir, NOV-205, IND-02, metenkefalin, PGN-007, Aeemannan, Gamimune, Prolastin, 1,5-dicaiFeoylquinic acid, ⁇ - 225, RPI-MN, VSSP.
  • a compound disclosed herein Is combined with two. three, four or more additional therapeutic agents in certain embodiments, a compound disclosed herein Is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein Is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein is combined with four additional therapeutic agents.
  • the two, three four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, or they can be selected from different classes of therapeutic agents, in a specific embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV non ⁇ nucleoside inhibitor of reverse transcriptase, in another specific
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound.
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucieoside inhibitor of reverse transcriptase, and an HIV protease inhibiting compound.
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HTV non-nucleoside inhibitor of reverse transcriptase, and a
  • a compound disclosed herein is combined with two HIV nucleoside or nucleotide inhibitors of reverse transcriptase.
  • a compound disclosed herein is combined with one, two, three, four or more additional therapeutic agents.
  • a compound dssciosed herein is combined with one additional therapeutic agent.
  • a compound disclosed herein is combined with two additional therapeutic agents.
  • a. compound disclosed herein is combined with three additional therapeutic agents.
  • a compound disclosed herein is combined with four additional therapeutic agents.
  • the one, two, three, four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, and/or they can be selected from d ifferent, classes of therapeutic agents.
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, ami an HIV protease inhibiting compound.
  • a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer.
  • a compound disclosed herein is combined with at least one HIV nucleoside inhibitor of reverse transcriptase, an integrase inhibitor, and a pharmacokinetic enhancer,
  • a compound disclosed herein is combined with two HIV nucleoside or nucleotide inhibitors of reverse transcriptase.
  • a compound disclosed herein is combined with at least one HIV nucleoside inhibitor of reverse transcriptase, an integrase inhibitor, and a pharmacokinetic enhancer.
  • a compound disclosed herein is combined with abacavir, abacavir sulfate, tenofovir, tenofovir disoproxlf &marate s tenofovir alafenamide, or tenofovir alafenamide hemifumarate,
  • a compound disclosed herein is combined with tenofovir, tenofovir disoproxil fimnarate, tenofovir alafenamide, or tenofovir alafenamide hemifumarate, [0211]
  • a compound disclosed herein is combined with a first additional therapeutic agent selected from the group consisting of: abacavir, abacavi sulfate, tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenoibvir alafenamide hemifumaraie and a second additiosal therapeutic agent selected from the group consisting of emtricitibirse and lamivudine.
  • a compound disclosed herein is combined with a first additional, therapeutic agent selected from the group consisting of: tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenofovir alafenamide hemlfumarate and a second additional therapeutic agent, wherein the second additional therapeutic agent is emtrichibine.
  • a compound disclosed herein is combined with one, two, three, four or more additional therapeutic agents selected from Ttiumeq®
  • a compound disclosed herein is combined with tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide, or tenofovir aiafenamide hemifumarate.
  • a compound, disclosed herein is combined with a first additional therapeutic agent selected from the group consisting of: abacavir sulfate, tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide. and tenofovir aiafenamide hemi fumaraie and a second additional therapeutic agent selected from the group consisting of erntrieitabine and laxnivudine.
  • a first additional therapeutic agent selected from the group consisting of: abacavir sulfate, tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide. and tenofovir aiafenamide hemi fumaraie
  • a second additional therapeutic agent selected from the group consisting of erntrieitabine and laxnivudine.
  • a compound disclosed herein Is combined with a -first additional therapeutic agent selected from the group consisting of: tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide, and tenofovir aiafenamide hemifnmarate and a second additional therapeutic agent, wherein the second additional therapeutic agent, is emtricitablne.
  • a compound disclosed herein is combined with 5-30 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemifumarate, or tenofovir aiafenamide and 200 mg emtrlcitablne, in certain embodiments, a compound disclosed herein is combined with 5-10; 5-15; 5-20; 5-25; 25-30; 20-30; 15-30; or 10-30 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemi fumaraie. or tenofovir akfenamide and 200 mg emtricitablne.
  • a compound disclosed herein is combined with 10 mg tenofovir aiafenamide fumaratc, tenofovir aiafenamide hemlfumaraie, or tenofovir aiafenamide and 200 mg emtricitablne. in certain embodiments, a compound disclosed herein is combined with 25 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemifumarate, or tenofovir aiafenamide and 200 mg emtricitablne.
  • a compound as disclosed herein may be combined with the agents provided herein in any dosage am ount of the compound (e.g., from 50 mg to 500 nig of compound) the same as if each combination of dosages were specifically and individually listed.
  • a compound disclosed herein is combined with 200-400 nig tenofovir disproxIL tenofovir disoproxil fumarate, or tenol vir disoproxii hemifurnarate and 200 mg emtrieitabine.
  • a compound disclosed herein is combined with 200-250; 200-300; 200-350; 250-350; 250-400; 350-400; 300-400; or 250» 400 mg tenofovir disoproxii, tenofovir disoproxii fumarate, or tenofovir disoproxil hemifurnarate and 200 nig emtrieitabine, In certain embodiments, a compound disclosed herein is combined with 300 mg tenofovir disoproxii fumarate, tenofovir disoproxii hemifumarate, or tenofovir disoproxii and 200 mg emtrlcitahlne.
  • a compound as disclosed herein may be combined with the agents provided herein m any dosage amount of the compound (e.g., from 50 mg to 500 mg of compound) the same as if each combination of dosages were specifically and individually listed.
  • the components of the composition are administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • a compound disclosed herein is combined with one or more additional therapeutic agents in a unitary dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration.
  • a compound disclosed herein is administered with one or more additional therapeutic agents.
  • Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of the compound disclosed herein and one or more additional therapeutic agents are both present in the body of the patient.
  • Co-administration includes administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents,
  • a unit dose of a compound disclosed herein is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents.
  • a unit, dose of one or more additional therapeutic agents is administered first, followed fay administration of a unit dose of a compound disclosed herein within seconds or minutes.
  • a unit dose of a compound disclosed herein is administered first, followed, after a period of hours ⁇ e.g..
  • a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound disclosed herein.
  • the crystalline forms are characterized by the interiattlce plane intervals determined by an X-ray powder diffraction pattern (XRPD).
  • XRPD X-ray powder diffraction pattern
  • diffractogram of XR PD is typically represented by a diagram plotting the intensity of the peaks versus the location of the peaks, i.e., diffraction angle 20 (two-theta) in degrees,
  • the intensities are often given in parenthesis with the following abbreviations; very strong - vst; strong - st; medium - m; weak ⁇ w; and very weak - vw.
  • the characteristic peaks of a given XRPD can be selected according to the peak locations and their relative intensity to conveniently distinguish this crystalline structure from others.
  • the measurements of the XRPD peak locations and/or intensity for a given crystalline form of the same compound will vary within a margin of error.
  • the values of degree 2 ⁇ allow appropriate error margins.
  • tha error margins are represented by
  • the degree 20 of about "8.7 ⁇ G.3" denotes a range from about 8,7- 0.3» i.e., about 9.0, to abont 8.7-0.3, i.e., about 8.4,
  • the appropriate error of margins for a XRPD can be ⁇ 0.5; ⁇ 0.4; ⁇ 0.3; ⁇ 0.2; ⁇ 0.1; ⁇ 0.05; or less.
  • the XRPD margin of error is ⁇ 0.2
  • a method of producing a composition comprising one or more polymorphs of ⁇ 2R,5S,13a )- ⁇ Hydroxy-7 > 9-dioxo- - ⁇ 2,4 ) 6 ⁇ trifi uof Plumbing3 ⁇ 4i)"2 A ⁇ hjP.SJoxazepinc ⁇ O-carboxamide, wherein the method comprises combining a compoimd of Formula (I) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the. compound of Formula (I).
  • composition comprising one or more polymorphs of (2R,5S, i :3 ⁇ 4R)-8-iiydroxy.7 s 9"dioxo ⁇ N 2,4 > 6 rifluorobenzyl).2.
  • Solvents suitable for polymorph formation may include, for example, methanol, ethanoi, water, isopropyl acetate, acetonitrile, tetrahydrofurao, methyl isobutyl ketone, and any mixtures thereof,
  • methanopyrido[l ⁇ 2';4,5]p ⁇ ajzino[2J-b][1 ]ox&zepine-10 ⁇ carboxamide produced according to any of the methods described herein.
  • the methods for preparing the polymorphs described herein may yield quantity and quality differences compared to the methods for preparing (2R,SS,13aR)-8-Hydroxy-7 5 9 ⁇ dioxo-N-(2 5 4 5 6"trifluoroben3 ⁇ 44)-23 s 4,5 : ,7 5 9 3 13,13a-octahYdrQ-2 ) 5- methanopyrido[ 1 ° s 2 ! :4 3 5]pyrazino[2, 1 -b] [ l 5 3] ixa/epine-' I G-earboxamide produced on laboratory scale.
  • a method of producing a composition comprising polymorphic Form I, Form ⁇ , or a mixture thereof, of (2R,5S, 13aR ⁇ -8- Iiydroxy-7,9 ⁇ dioxo ⁇ - (2A6 rifiuorabenzyl) ⁇ 2 ; 3,4,5,7,9, 13, ! 3a-octahydro ⁇ 2,5 ⁇
  • composition comprising polymorphic Form III of (2R J 5S ⁇ 3aR)-8 ⁇ Hydroxy ⁇ 7 5 9-dioxo-N-(2 > 4 5 6 ⁇ fluoroberizyl)-
  • carboxamidc wherein the method comprises combining (2R,5S,I3aR) ⁇ 8 ⁇ Hydroxy"7,9-dioxo- N-(2,4,6 ' rifdiorobeB S'l)-2,3 5 4,5,7,9 :i 13,I3a-octahydro--2,5- methanopyrido[1 ⁇ 2 !
  • composition comprising polymorphic Form IV, Form VII, and Form VIII, or a mixture thereof, of (2R 5 5S,!3aR)-8- Hydroxy-7.9 ⁇ ioxo-N-(2A6- ⁇
  • composition comprising polymorphic Form V of (2R,5S S 13aR)-8-Hydroxy-7,9 ⁇ toxo-N 2A ⁇
  • composition comprising polymorphic Form VI of(2R,5S, 13aR 8 : iydroxy-7,9Hiioxo-Ni:2 ; ⁇
  • the solvent is selected from the group consisting of methanol, water, and any mixtures thereof, In an embodiment, the solvent is a mixture of water and methanol.
  • oetahydro «2,5-metfaanopyrido[i ',2 ! :4,,5 jpyrazino[2, 1 ⁇ h][ 1 ,3 joxazepine- 10-carboxamide with a solvent, wherein the solvent is selected from the group consisting of methanol, water, aad any mixtures thereof, Irs an embodiment, the solvent is a mixture of water and methanol
  • olate e.g. a compound of Formula (If) is described herein.
  • composition comprising one or more polymorphs of sodium (2R S 5S,13aR.) ⁇ 7,9 ⁇ dioxo-10-((2,4 s 6- xrit1uorobenzyi>carbainoyi)-2,3,4,5,7 ! 9 J 13,13a-octaiiydfo-2 5 ⁇
  • the method comprises combining a compound of Formula (II) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the compound of Formula (II),
  • a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the compound of Formula (II)
  • Solvents suitable for polymorph formation may include, for example, methanol s etharsoL water, isopropyl acetate, acetonitrile, tetrahydrofuran, methyl isohutyl ketone, and any mixtures thereof.
  • the methods for preparing the polymorphs described herein may yield quantity and quality differences compared to the methods for preparing sodium (2R,5S 5 13aR)-7,9 ⁇ dioxo-IO-((2,4.6- trifIuoFobenzyl)carhamoyl)-2,3,4,5,7 s 9, 13J 3a-0 €iahydfo-2,5- produced on laboratory seals.
  • a method of producing a composition comprising polymorphic Form ! of sodium (2R,5S, 13aR ⁇ -7 5 9-dio3 ⁇ 4>i0- ⁇ (2 ; 4 5 6- trifluorobenzyl)carbamoyl)-2,3 ,4,5,7,9, 13, 13a-octa ydro-2 s 5- metbanopyrido r,2 !
  • a composition comprising one or more polymorphs of potassium (2R.5S, 13aR) ⁇ 7,9 ⁇ dioxo ⁇ l 0 ⁇ ((2,4,6 ⁇ criz1uoa>benzyi)carbaTnoyi)-2,3.4 5.7,9, 13, 13a-octa ydro-2 5 5- metha o ido[1 2 ! :4,5]p ziu [2J -b][l !
  • the method comprises combining a compound of Formula (III) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the compound of Formula (III).
  • composition comprising one or more polymorphs of potassium (2R,5S,13aR)-7,9-dioxo-10-((2,4,6- tri fluoroberszy S)carbamoyi)-2, 3,4,5,7,9,13, 13 a ⁇ octahydro-2,S - m.ethanopyrido[r,2 , :4,5]p ⁇ i razino[2,l-b][l ⁇ 3]oxazepin ⁇ 8-olate J wherein the method comprises combining potassium (2R ( 5S,13aR)-7,9-dioxo-10-((2 s 4,6-h"jfiyorobenzyl)carbamoy1) ⁇
  • Solvents suitable for polymorph formation may include, for example, methanol, etlianol, water, isopropyl acetate, aceionitrile, tetrahydrofuran, methyl isobutyl ketone, and any mixtures thereof.
  • the methods for preparing the polymorphs described herein may yield quantity and quality differences compared to the methods for preparing potassium (2R,5S,13aR)-7,9-d oxo ⁇ l 0- ⁇ (2 s 4,6 rifIuorobenzyl)carbamoyi) ⁇ 2 s 3,4 5 5 J 7,9 J I 3J 3a--octahydro-2,5- meihanop rido[r 5 2 ! :4 J S]pyraziao[2,l-b][l,3]oxazspin ⁇ 8 ⁇ olate produced on laboratory scale.
  • composition comprising polymorphic Form ⁇ of potassium (2R s 5SJ 3a )-7,9-dioxO"10- ⁇ 2,4,6 ⁇
  • a potassium base e.g. potassium acetate
  • a potassium base is a solvent to produce a composition comprising polymorphic Form II of potassium (2R,5S 5 13aR) ⁇ 7,9-dioxo ⁇ 10 ⁇ ((2 5 4,6- trifiuorobenzyl)carbamoy!)-2 s 3,4 s 5 5 7,9 s 13,13a--octahydrO'-2,5- methanopyrido[r s 2 , :4,5]p>TazIsio[2 s l--bJ[l i 3]oxa3 ⁇ 4epk-8 ⁇ o!ate )
  • the solvent is selected from the group consisting of acetonitriie, water, and any mixtures thereof. la an embodiment, the solvent is a mixture of acetonitriie and water.
  • composition comprising polymorphic Form III of potassium (2R ; 5S, 1 aR)-7, -dioxo- 10- ⁇ (2,4,6- trifiuorobenzyI)carbamoyi)-2J s 4,5,7 s 9J 3,13a-oetahyd!O-2,5 ⁇
  • potassium phosphate in a solvent to produce a composition comprising polymorphic Form II of potassium (2R, 5 S, 1 aRV7,9-dioxo- 1 £>- ⁇ ( 2,4,6- tritluoro benzy l)carbamoyi) ⁇ 2 5 3 ,4, 5, 7,9, 13,13 a-ociahydro-2,5 - metham-)pyrido[i ! ,2 f :4,5]pyfazko[2 J l4 ⁇ ][i ,3]oxazepin"8-olate J wherein the solvent is methanol,
  • pro ided is & method of producing a composition comprising polymorphic ciiric acid co-crystal, fbmaric acid co-crystal oxalic acid co-crystal, or a mixture thereof, of (2R3S, 1 Sal ⁇ -g-H dro y-T ⁇ -dioso-N-C ⁇ -triflu r benEy [)- 2,3,4,5 ,7,9,13 3a ⁇ oetahydro ⁇ 2,5 ⁇ rnet3 ⁇ 4 ⁇
  • methanop ⁇ Tido iy2 ⁇ 4,5]pyrazino with an acid e.g. citric acid, fumaric acid, or oxalic acid
  • an acid e.g. citric acid, fumaric acid, or oxalic acid
  • a solvent e.g. a solvent to produce a composition comprising polymorphic citric acid co-crystal, farrsarie acid co-crystal, oxalic acid co-crystal, or a mixture thereof, of the C2R.58J 3aR) ⁇ 8 ⁇ Hydroxy-7,9-diox0 ⁇
  • polymorphs described herein in the manufacture of a drag product
  • the one or more of the polymorphic forms described herein ⁇ e.g., one or more of polym orphic Forms I, IL III, IV, V s VI, VII and VIIJ) may be used as an intermediate in the manufacturing process to produce the drug product.
  • methanop iido[r,2 f :4,5]pyrazino[2J -fo][l,3]oxazepme ⁇ 10-carboxamide are used in the manufacture of an active pharmaceutical ingredient, fa certain embodiments.
  • 2,5-methanopyrido[ 1 ⁇ 2 ! :4,5]p> ⁇ a ⁇ is used in the manufacture of an active pharmaceutical ingredient.
  • Form II of (2R.5S, 1 :1 ⁇ 2R)-8 Jydroxy-7,9-dioxo-N--(2 5 4 J 6-triflirorobenz)'-! ⁇ 2 ! 3 > 4 s 5 5 7 5 9 5 13,13 ⁇ octahyd.ro- is used in the manufacture of an active pharmaceutical ingredient.
  • polymorphs described herein in the manufacture of a drug product.
  • the one or more of the polymorphic forms described herein may be used as an intermediate in fee manufacturing process to produce the drug product.
  • polymorphs described herein in the manufacture of a drug product.
  • the one or more of the polymorphic forms described herein e.g., one or more of polymorphic Forms I, II, and HI
  • 2,5-methanopyrido[i ⁇ 2 ,5]pyrazino[2J-b][i s 3]oxazepin-8 ilatc is used in the manufacture of an active pharmaceutical ingredient.
  • compositions comprising one or more of fee polymorphic forms described herein ⁇ e.g., one or more of polymorphic Forms I to VIII of ⁇ 2R,5SJ3aR) ⁇ 8 ⁇ Hydroxy ⁇ 7 s 9 ⁇ dioxo-N ⁇ (2 J 4 ) 6 ⁇ p-ifiuorobenzyi)-2 s 3,4 5 5 J 7 > 9, 13, 13a-octahydro-2,5- methanopyrido[ I !
  • an article of manufacture such as a container comprising a dosage form of one or more of the polymorphic forms described herein (e.g., one or more of polymorphic Forms I to VIE of ⁇ 2 3 5S > 13aR) » 8-Hydrox5'-7,9-dioxo-N- ⁇ 2,4 J 6.
  • the article of manufacture is a container comprising a dosage form, of one or more of the polymorphic forms described herein (e.g., one or more of polymorphic Forms I to VIII of (2R..5S, 13aR)-8-Hydroxy-7,9-dioxo-N-(2 A6- trIfluorohenzyl)-2,3 ,4,5,7,9, !
  • the dosage form is a tablet
  • kits also are contemplated.
  • a kit can comprise a dosage form of a pharmaceutical composition and a package insert, containing instructions for use of the composition in treatment of a medical condition.
  • the instructions for use in the kit may be for treating HIV.
  • the instructions for use in the kit may be for treating HIV,
  • the polymorphic, salt, co-crystal and solvate forms described herein may potentially exhibit Improved properties.
  • the polymorphic, salt, co-crystal and solvate forms described herein may potentially exhibit improved stability.
  • improved stability could have a potentially beneficial impact on the manufacture of the Compound of Formulas L II, and/or ill, such as for example offering the ability to store process intermediate for extended periods of time.
  • Improved stability could also potentially benefit a composition or pharmaceutical composition of the Compound of Formulas I, II, and/or ill.
  • solvate forms described herein may also potentially result in improved yield of the Compound of Formulas I, II, and/or III, or potentially result in an improvement of the quality of the Compound of Formulas 1, 11, and/or HI.
  • the polymorphic, salt and solvate forms described herein may also exhibit improved
  • Citric Acid Co-crystal Form I [0290] C2R,5S,i3aR 8-hydroxy-? 5 9-diox ⁇
  • octahydro"2,5-metliaiTopyrkk)[1 ⁇ 2 : :4.5 lpyrazino[2 5 1 -b][ tJJoxazepirse- 10-carboxamide (131 mg) aod fumaric acid (103 mg) were added to a glass vial.
  • Te rahydrofura (1 mL) was added, the vial capped, and the suspension stirred at about 21 °C for two days.
  • An additional 1 mL of tetrahydrofuran was added, and the mixture was heated to 45 °C. After about 12 hours at 45 ft C the mixture was fou d to be fully dissolved wd was removed from the heat bath.
  • octahydro-2 > 5-me&anop ⁇ Tido[l ⁇ (147 mg) arid oxalic acid (134 mg) were added to a glass vial, Tetrahydrofuran (1. mL) was added, the vial capped, and the resulting solution was stirred at about 21 °C for about two days. The vessel was vented, and the solvent was allowed to evaporate at room temperature unassisted. After ne day solids were observed m " the vial and it was re-capped. Two days later large crystals were found in the vial and were identified as a 1 :1 Formula I oxalic acid co-crystal
  • octahydro ⁇ 2 5 5 ⁇ methanopyTido[r,2 t :4 ⁇ (1.96 g) was charged to a reaction flask and stirred, followed by etbanol (20 mL), The mixture was stirred at room temperature resulting in a suspension.
  • potassium hydroxide (253 mg) was dissolved in deionized water (5 ml,). The potassium hydroxide solution was transferred by syringe pump to the stirring suspension over about 2.5 hours followed by a rinse into the reactor with water (2 mL) after the base solution transfer was completed.
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • DFS dynamic vapor sorption
  • Figure 16 compares the calculated XRPD pattern of sodium ⁇ 2R,5S,13aR ⁇ 7 ! 9- io o 0H(2A6-t f iflu roben3 ⁇ 41)c ⁇
  • Differential scanning calorimetry Thermal properties were evaluated using a Differential Scanning Calorimetry (DSC) instrument (TA QIGGO, TA instruments, New Castle, DE, USA). Approximately 1 to 10 mg of solid sample was placed in a standard aluminum pan vented with a pinhole for each experiment and heated at a rate of 5 to 10 °C/min under a 50 mL/miii nitrogen purge. Data analysis was conducted using Universal Analysis 2000 Version 4.7A. (TA Instruments, New Castle, DE, USA), Heat of fusion analysis was conducted by sigmoidal integration of the endother ic melting peak.
  • DSC Differential Scanning Calorimetry
  • Thermogr itmtric analysis was performed on a TGA instrument (TA Q5G0, TA Instruments, New Castle, DE, USA). Approximately 1 to 10 mg of solid sample was placed in an open aluminum pan for each experiment and heated at a rate of 5 to 10 °C/min under a 60 mL/min nitrogen purge using. Data analysis was conducted using Universal Analysis 2000 Version 4.7A (TA Instruments, New Castle, DE, USA),
  • Dynamic vapor sorption The hygroscopicity was evaluated at room temperature using a dynamic vapor sorption (DVS) instrument (TGA Q5000 TA Instruments, New Castle, DE), Water adsorption and desorption were studied as a function of relative humidity (RH) over the range of 0 to 90% at 25 8 C. The relative humidity in the chamber was increased by 10% RH and held until the so lid and atmosphere reached equilibration. The equilibrium test was continued until passed or expired after 5 or 10 hours. At this point, RH was raised 10% higher and the process was repeated until 90% RH was reached and equilibrated. During this period, the water sorption was monitored.
  • DVS dynamic vapor sorption
  • the single crystal X-ray crystallography data for Formula I Forms I -IV are summ rized in Table 2A below.
  • the single crystal X-ray crystallography data for Formula I Forms V-VIII are summarized in Table 2 ⁇ - ⁇ below.
  • the indexing data for Formula II Form ! is summarized in Table 2C below.
  • the single crystal X-ray crystallography data for the co- crystals of the present, in vention summarized in Table 2C below.
  • the single crystal X-ray crystallography data for Formula III Forms I ⁇ is summarized in Table 2D-I below. Data from furt her characterization of the crystals ate summarized in Tables 3 A and 38 below. Data from further characterization of the crystals are also summarized in Tables 3A and 3C ⁇ i below.
  • Table 3A Crystal Data and Structure Refinement for Formula I Forms ⁇
  • Table 3A-I Crystal Data and Structure Refinement for Formula ⁇ Forms V - Vill
  • Table 3B Crystal Data and Structure Refinement for Formula I Co-Crystals
  • Table 3C-h Crystal Data and Structure Refinement for Formula ⁇ Form I, Formula HI Form II (Dimer), arid Formula III, Form III
  • Formula III is bydrated. In certain embodiments, Formula III is hydrated with five to six water molecules.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • AIDS & HIV (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to crystalline forms and co-crystals of (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanpyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide, the pharmaceutical formulations, and the therapeutic uses thereof. The present invention also relates to novel crystalline forms of sodium (2 R,5S,13aR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepin-8-olate Form I.

Description

CRYSTALLINE FORMS OF (2R,5S,13AR)-8-HYD OXY-7,9-DiOXO-N-(2>4,6-
TRIFLUOROBE ZYL)-2,3,4,5)759J3, 1 3A-OCTAHYD O~2;5" METHANOP YRIDO[ I ',2' :4,5]PYRAZINO[2, LB] [ l,3]OXAZEPI E- 10-
C A RB OX AMIDE
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to and the benefit, under 35 U.S.C. 1 19(e) of U.S. Provisional Patent Application Serial Nos. 62/015,238, filed June 20, 2014 and 62/017,183, filed June 25, 2014, the disclosures of which are hereby incorporated by reference in their entirety.
FIELD
[0002] The present invention relates to novel crystalline forms and co-crystals of (2R,5SJ3aR)-8-hydroxy-7,^^
2,5~raetham}pyrido[r;2!:4s5]pyrazino[2s I-bl[1 ]oxazepme-10-cm¾oxamide, the pharmaceutical formulations, and the therapeutic uses thereof,
BACKGROUND
[0003] Human immunodeficiency vims infection and related diseases ate a major public health problem worldwide. Human immunodeficiency virus type 1 (ffiV-1) encodes three enzymes which are required for viral replication: reverse transcriptase, protease, arid integrase. Although drags targeting reverse transcriptase and protease are in wide use and have shown effectiveness, particularly when employed in combination, toxicity and development of resistant strains have limited their usefulness (Palclla, et al. N. Engl. J Med. (1998) 333:853-860; Richman, D, D. Nature (2001 ) 410:995-1001).
[0004] A goal of antiretrovira! therapy Is to achieve viral suppression in the HIV infected patient. Treatment guidelines published by the United States Department of Health and Human Services provide that achievement of viral suppression requires the use of combination therapies, i.e., several drags from at least two or more drug classes. In addition, decisions regarding the treatment of HIV infected patients are complicated when the patient requires treatment for other medical conditions. Because the standard of care requires the use of multiple different drugs to suppress HIV, as well as to treat other conditions the patient may be experiencing, the potential for drug interaction is a criterion for selection of a drug regimen. As such, there is a need for antirstroviral therapies having a decreased potential for drug interactions.
[0005] As discussed in co-pending application United States Serial No, 14/133,855, filed December 1 , 2013 entitled "POLYCYCLIC-CARBAMOYL-PYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE", (2R,5SJ :¼R)~8~hydroxy~?,^
trifluorobenzyI)-2,3,4,5 s7,9513, 13a~octahydro-2>5-niethanopyrido[r,2,:4,5]pyrazino[25l- b] [ 1 J joxazepine- 10-carboxa de demonstrates anti-viral activity. As discussed in copending application PCX Serial No, US2013/076367, filed December 19, 2013 entitled "POLYCYCL1C-CARBAMOYI.PYRIDONE COMPOUNDS AND THEIR
PHARMACEUTICAL USE", (2R55S,13aR)-8-hydroxy-7i9-dioxo-N-(2,4,6 riflMorobenKyl)~ 2,3,4,5,7,9, 13,13a~oemhydro~2,S-methanopyrido[ i :4,5]p mzm.o[2, 1 ~b][l JJoxazepme-lO- earboxamide demonstrates anti-viral activity.
[0006] (2R,5S 3aR)-8-hydroxy~7s9-dioxo-N-(2s4,6-trifiuorobenzyI}.233,435,7,9, 13,13a- octahydro-2}5-meihanopwido[ 1 \2::4 5]pysaxioo[2, 1 -b] [ 1 ,3]oxazepine-l 0-carboxamides (Formula I), has the following structure:
Figure imgf000004_0001
[0007] It is desired to have physically stable forms of the compound that are suitable for the therapeutic use and the manufacturing process,
SUMMARY
[0008] In one aspect the present invention is directed to novel forms of (2R.5SJ 3a }-8- hydroxy-7,9 lioxG-N-(2,456-?TM^
r«ethanopyrido[ 1 \2' :4, 5 j pyraxino [2 s 1-b] [1 ,3]oxazepine- .10-carboxamide, [0009] In one embodiment, the present invention is directed to (2R,5S, 13aR)-S-hydroxy-
7,9"dioxo-N 2,4,6-trifluorobsEz>¾-2 ;4,5 ,9J 3JJa )c ah dro~255-- methanopyrido 1 i,21:4,5]pyrazino[2, 1 -b [ 1 Jjoxazepme- 10~carboxamide Form L
[0010] In a further embodiment, fee present invention is directed to i2 .SS, 13a V8- hydroxy-7,9^ioxo-N^
meihasopyrido[r,2!:4J5]pyrazino[2,l~b][I ,3]oxazepine~10~carbGxaniide Form 11
[0011] In a still further embodiment the present invention is directed to (2R,SS513aR)-8-
Figure imgf000005_0001
niethanopyrid [ V, 2 ' ;4, S]pyrazmo[2, 1 -b] [1,3] oxazepine- 10~carboxam ide Form III .
[0012] In a yet further embodiment, the present invention is directed to (2Rs5S,13aR)-§~ hydroxy-759-dioxo~N~(2,4,0 rifiuQre^^^
methano yn oj ^^Sjp razk^^ Form IV.
[0013] In a yet further embodiment, the present invention is directed to (2R,5S,1 aR)~8~ hydroxy-7,9~dioxo-N~(2,4,6-trifluorobenzyl)-2s3,4,5,7s9, 13,13 a~oeiahydre-2,5 ~
methaiiopyrido[r!2!:4,5]pyrazino[2,l-b][l,3]oxa^epine'-10-earboxarrdde Form V.
[0014] In a yet further embodiment, the present invention is directed to (2R,SS, 13aR)-8- hydroxy-7,9-dioxO"N-(2,4,6 rifluorobenzyl)~2?3!4s5.7,9J.3513a-ocfebydrO"2,5»
methancpyrido[ 1 >,2':4!5]pyrazino[2! l-b][ 153]oxa2epine- 1 O-earboxamide Form VI.
[0015] In a yet further embodiment, the present invention Is directed to (2R,5S, 13 }-8- bydroxy~7s9-dioxo-N-(2,4,6 rit1uorokinzyi}-2,3,4,5,7,9, ! 3, 13a-octahydro-2.5- methanopyrido[ I \2':4,5]pyrazino[2,l -b] [ 1 s3]oxazepine- i 0-catboxamide Form VII.
[0016] in a yet further embodiment, the present invention is directed to (2R55S,13aR)-8- h dro ^^-dio o-N^^^-triflisorobeaxy^^J^ ,?^.13, ] 3a-octahydro~2,5- ffiethanopyrido[172^4,S]pyrazmoP^ Form VIII.
[0017] In a certain embodiment fee present Invention Is directed to a fnmarle acid co- cr ^ of (2R,5S 3aR)~8-hydroxy-7^
octahydro-2,5-methanopyrido[ 1 ',2' :4,5]pyra7ino[2s 1. -b] [ 1 Jjoxazepine- 10-earboxamide.
[00 IS] In another embodiment, the present invention is directed to a citric acid co-erystal of (2R,5S,13aR)-8 tydrox^
octahydro-2,5-methaBopyrido[r,2!:4J5]p}?razIi o[2J-b|[1 ,3]ox.azepine 0-carboxamide.
[001 ] In yet another embodiment the present invention is directed to an oxalic acid co- crystal of (2R,5S, 13aR)-8-hydroxy-7,9→i-oxo-N^^
oct hydro~25Sn ethaiTO } ido[1^2^
[0020] In particular embodiments, the present Invention is directed to crystalline forms and co-crystals of (2Rs5S513aR)-8-hydroxy-7)9"dioxo-N-{2,456~trii1.uorobenzyi)~
2,3^5/7?9J3J3a-oe†ahydro~255~mei?¾atto^^
carboxamkie.
[0021] In a fbrther aspect, the present invention is directed to novel forms of sodium
Figure imgf000006_0001
2s5-me&anopyrido[1^2';455]p 'Ta ino[25l-b][L3]oxazepm-8-oiate, having the following structure (Formula II):
Figure imgf000006_0002
[0022] in a still further embodiment, the present invention is directed to sodium.
(2R,5S 13aR)-7, Uo cHl 0<{2A^
2j5~methanopyrido[ 1 2' :4sS]pyrazino[2, 1 -b] [ 1 ,3]oxazepin-8-olate Form I
[0023] In yet a further aspect, the present invention is directed to novel forms of potassium (2R!5S, 13aR)-759~dioxo-10-({2J4,6-trifiuorobenzyl)carbam.oyl)-2s3,4,5s7,95I3, I 3a- octahydro~2>5~methaBopyrido[ l\2I:4s5]pyrazino[2J.~b][ls3]oxaz€pin-S-oiaie, having the following structure (Formula ill):
(III)
Figure imgf000006_0003
[0024] In yet another embodiment, the present invention Is directed to potassium
(2R,5S 3aR)-7,9^ioxo-?.0~((2A^^
2,5~methanopyrido[l ',2' :4,5]pyrazino[2„ 1 -b] [ 1 Jjoxazepin- 8 -elate Form I.
[0025 ] In yet another embodiment, the present invention is directed to potassium
(2R,5S, I 3aR)-7,9-dioxo- 10-((2^,6'-triiluorc¾enz>'])carbamoyi)-2,3,4i5,7,9> 13,13a-oetah dn -
2,5-metIiaBop Tido[1^2i;4,5]pyrazino[25!-b][l,3]oxazepIn~8 ilate Form II,
[0026] In yet another embodiment, the present invention is directed to potassium
(2R,5S43aR)~7f9~dioxo~1 -((2s4,6^
2,S-methanopyrido[ 1 ',2' 5]pyra:?:ino[2, 1 -b][ 1 ,3]oxazepin«8-oiate Form III.
[0027] in a still other embodiment, the present invention is directed to hydraied potassium (2R,5S, .1 ;¼R)"7,9-dioxo-10-{{23456 fifluorobenzyi)ca banioyl)»2;354,Ss7,9, 13, 13a- octahydrO"2>5~r eihanopyrido[ I !,2! A5]pyrazino[2, 1 -b] [ 1 ,3]oxa2;epm-8-olate.
[0028] in still another embodiment, the present invention is directed to methods of treating or prophyiactically preventing an HIV Infection by administering compound (e.g. Formulas (I), (II), and/or (III)) provided herein.
[0029] In still another embodiment, the present invention s directed to a compound (e.g. Formulas (I), (II), and/or (III)) provided herein for use in methods of treating or
prophyiactically preventing an HIV infection.
[0030] In still another embodiment, the present invention is directed to the use of a compound (e.g. Formulas (I), (Π), and/or (III)) provided herein in the manufacture of a medicament for treating or prophyiactically preventing HIV infection.
DESCRIPTION OF THE FIGURES
[0031] Figure It XRPD pattern for (2R(5SJ3aR)-8-hydro.^
triflnQro e ¾d)-2,3ASA9J3
b][L3]oxazepine~iO~earboxamide Form I.
[0032] Figure li XRPD pattern for (2 S,i:½R)-8mydroxy-7;9-dioxo-N-(2i4,6- rrifluorobenzv 1)~2 A4 ? A 13, 13a-octahydro-2,5-methanopyrido[ 1 \2':4,5 jpyrazino[2, 1~ bj[l,3]o¾azepine~10~carboxamide Form IL [0033] Figure 3; XR.PD pattern for (2R,5SJ13aR)-8¾droxy-7f9-dioxo- -(2>4!6- trifluorobenz l)-!^
b][l 53]oxazepine-i0-carbox.amfde- Form ill.
[0034] Fi u e 4: XRPD palter?- for (2R.58, 13aR}-8-hydroxy-?J9-dioxo-N-{2,4,6- trIfl orobe.rsJiyl)-2 ,435 59;I343a-octahydro-2s5"methanopyri^
b] [ 1 , 3]oxazepine- 10-carfaoxanii de Form IV,
[0035] Figaws 5; XRPD pattern for sodium (2R5SS,13aR)-7,9-dioxa-10-ii2,4,6- iri fluorobesiay l)car bamoyl)-2, 3 ,4, 5 , 7,9, 13, 13a-octabydK>~255~
metha.KOpyrido[1^21:4J5]pyrazino[2sl-b][1 1oxazepk-8-olate Form L
[0036] Figure 6: Actual and calculated XRPD pattern for (2R,5S,13aR)-8-hydroxy~7s9» dio o-N^^^^ lfluoro iizylJ^^^^ ^JS.Da-ociahydro-^S- methairopyTldo[r>2,:4s5]pyra2ino[2 -b][l,3]oxazepke~!0-carboxamide oxalic acid co- crystal Form I.
[0037] Figure It DSC for (2R,5S, 13aR)-8-hydroxy~7,9^iox^
2,3,4,5s7,9 ,.13aH>c ahydro-2J5-meihanopyrido[l 2,:4,5]pyrazIno[2J-b][13
earboxamide Form I.
[003S] Figure 8 DSC for (2R,5S, 13aii)--8 iydroxy~7>9-d(oxo-N--(254,6 nfluoroben2yi)-
2,3,4,5,7,9, 13, 13a-ociabydTO~2?5-«iei ajiopwMoi F^'^SJ a inoP, 1 >][I ]oxa¾epifie 0- carboxamide Form 10.
[0039] Figure 9: DSC for sodium (2R.5S, ) 3aR.)-7,9-dioxo- 10 - ((2,4,6- triiluorobenzjd)cafbamoyl)~2s3,4,5,7,9! 13, 13a-octahydro-2,5~
metjaaHopyrido[1^2':4!5]pyfazmo[2 -b][I,3]oxazepk-8-oiate Form L
[0040] Fsg?ire 10: TGA for (2R.5S, 13aR>8-hydroxy-7.9-dioxo-N-(2,4,6-
Figure imgf000008_0001
b][l,3] xazepiise-30-carboxamide Form L
[0041] Figure II; TGA for (2R55Si 13aR}"8-hydroxy~7>9-dioxo-N-(2.4)6-- trifiuoFoberizyI)~2,3A3 s9 3 3a^
b] [ 1 ,3 joxazepirie- 10-carboxam ide Form III. [0042] Figure 12% TGA for sodium (2IL5S, 13aR)~?59~dioxo- 1 -{(2,456- tritluorobenz l)earbamoyI)~253A5,7A 13, 13a-ociahyd.ro~2,5- nieihanopyridoi 1 2';4,5 ]ρντδχΐηο[2,1 -b]| 1 ,3]oxazepin-8-csjate Form L
[0043] Figure 13; DVS for (2R,SSJ3a >8- 5ydroxy~7,9-dioxo--N~(2s4,6~
trifluoroben2yl)-2,3A5>7,9, 13J3aK ;tehydro-2,5-meihanopyrido{, 1 , )2!:4>5]pyraziHo[2i I - b] [1,3 j oxazepine- 10-carboxam kk- Form I.
[0044] Figure 14: DVS for (2R,5SJ3< R)~S- ydrox --7J9-dloxo«N^(2,4,6-.
tritluorobenz 1)~ ,: ^^^^
b][l53]oxazepine"10-carboxamide Form ill.
[0045] Figure 15: DVS for sodium (2R,5S513aR)-7s9-dioxo~I0~((2>4,6- iriil«orobenzj.'i)carbamoyl)-2!,3J4i5i 7,9,13,13a-octahydro~2,5~
methanopyrido[i ',2':4.5]pyrazsno(251 -b] [ ) ,3]oxazepin-8-oIate Form L
[0046] Figure 16; Calculated and Experimental XRPD pattern for .sodium
Figure imgf000009_0001
DETAILED DESCRIPTION
[0047] In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiment?; of the invention. However, one skilled in the art will understand that the invention may be practiced without these details. The description below of several embodiments is made with the understanding that the present disclosure is to be considered as an exemplification of the claimed subject matter, md is not intended to limit the appended claims to the specific embodiments illustrated. The headings used throughout this disclosure are provided for convenience only and are not to be construed to limit the claims in any way, Embodiments illustrated under any heading may be combined with embodiments illustrated under any other heading.
Definitions
[0048] Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and variations thereof, such ass "comprises" and "comprising" are to be construed in an open, inclusive seme, thai is as "including, but not limited to".
[0049] Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment.
Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
[0050] Embodiments that reference throughout tills specification to "a compound" or "e.g. Formulas (I), (II), and/or (III)" includes the polymorphic, salt, co-crystal, and solvate forms of the formulas and/or compounds disclosed herein. Thus, the appearances or the phrases "a compound" or "e.g. Formulas (T). (II), and/or (111)" includes Forms I-VIII of Formul I, Form i of Formula If, Forms HI of Formula Hi, and/or the f maric acid, citric acid, and oxalic acid co-crystals as described herein.
[00511 The invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of Formulas (I), (ΪΪ), and (III) being isotonics! ly-Ja beled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as ¾ ¾ UC, i3C, MC, ¾, 15N, i50, 370, 38G, 33P, 3¾ 3SS, l% ¾ n\ and % respectively. These radiolabeled compounds could he useful to help determine or measure the
effectiveness of the compoimds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain Isotopically-labeled compounds of Formulas (f) (II) and (III), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i n ¾ and carbon- 14. i.e. i4C, are particularly useful for this purpose in view of their ease of i ncorporation and ready means of detection.
10052] Substitution with heavier isotopes such as deuterium, i.e. ¾ may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half- life may increase or dosage requirements may be reduced. Thus, heavier Isotopes may he preferred In some circumstances. [0053] Substitution with positron emitting isotopes, such as i :C, iSF, tsO and i3 s can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy, isoiopicaUy-labeled compounds of Formulas (I), (II), (III) can generally be prepared by conventional techniques known to those skilled in the art or by processes 'analogous to those described in the Examples as set out below using an appropriate
Isotopkally-iabeled reagent in place of the non-labeled reagent previously employed.
[0054] "Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an. efficacious therapeutic agent.
[0055] ' ptional" or "optionally" means that the subsequently described event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryi" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
[0056] 'Thannaccutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or ernuisifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
[ 0057] A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes ail pharmaceutically acceptable carriers, diluents or excipients therefor.
[0058] "Effective amount" or "therapeutically effective amount" refers to an amount of a compound according to the invention, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician. The amount of a compound according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological acti vity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidental!}' with the compounds of the invention, and the age, body weight, general health, sex and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art. having regard to their own knowledge, the state of the ait, and this disclosure,
[0059] lit certain embodiments, the term "treatment" is intended to mean the
administration of a compound or composition according to the present Invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load i a patient, The term "treatment" also encompasses the administration of a compound or composition according to the present invention post-exposure of the individual to the virus but before the appearance of symptoms of the disease, and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching deteetible levels in the blood, and the admins stratioa of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life. In certain embodiments, the term "treatment" as used herein is intended to mean the administration of a compound or composition according to the present invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load in a patient. In certain embodiments, the term "'treatment" as used herein is further or alternatively intended to mean the administration of a compound or composition according to the present Invention to maintain a reduced viral load in a patient. The term "treatment" also encompasses the administration of a compound or composition according to the present invention postexposure of the individual to the virus but before the appearance of symptoms of the disease; and/or prior to the detection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching deteetible levels in the blood, and the administration of a compound or composition according to the present invention to prevent perinatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life, in certain embodiments, the term "treatment" as used herein is further or alternatively intended to mean the administration of a compound or composition according to the present invention post-exposure of the individual to the virus as a subsequent or additional therapy to a first-line therapy (e.g., for maintenance of low viral load), [0060] "Prevention" or "preventing*5 means any treatment of a disease or condition that causes the clinical symptoms of the disease or condition not to develop. The term
"prevention" also encompasses the administration of a compound or composition according to the present inventio pre~exposure of the individual to the virus {e.g., pre~exposure prophylaxis), to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching detect! hie levels in the blood.
[0061] The terms "Subject" or "patient" refer to an animal, such as a mamma! (including a human), that has been or will be the object of treatment observation or experiment. The methods described herein may be useful in human therapy and''or veterinary applications. In some embodiments, the subject is a mammal (or the patient). In some embodiments the subject (or the patient) is human, domestic animals (e.g., dogs and cats), farm animals (e.g., cattle, horses, sheep, goats and pigs), and/or laboratory animals (e.g., mice, rats, hamsters, guinea pigs, pigs, rabbits, dogs, and monkeys). In one embodiment, the subject (or the patient) is a human. "Human (or patient) in need thereof 5 refers to a human who may have or is suspect to have diseases or conditions that would benefit from certain treatment; for example, being treated with the compounds disclosed herein according to the present application.
[0062] The term "antiviral agent" as used herein is intended to mean an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a human being, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a human being.
[0063] The term "inhibitor of HIV replication" as used herein is intended to mean an agent capable of reducing or eliminating the ability of HIV to replicate in a host ceil, whether in vitro, ex vivo or in vivo.
[0064] A "tasjtomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tantomers of any said compounds.
[0065] Reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to "about X" includes description of "X". Also, the singular forms "a" and "the" include plural references unless the context clearly dictates otherwise. Thus, e.g., reference to "the compound" includes a plurality of such compounds and reference to "the assay"
i t inclisdes reference to one or more assays and equivalents thereof known to those skilled in the art.
[0066] "Pharmaceutically acceptable" or "physiologically acceptable" refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human
pharmaceutical use.
[0067] "Unit dosage forms" are physically discrete units suitable as unitary dosages for subjects (e.g.. human subjects and other mammals), each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical carrier.
Crystalline Forms Formula I
[0068] It s desirable to develop a crystalline form of (2R,5S, ί 3aR)-S-hydroxy-7,9-dioxo-
N»(2>456-trifluorobenz>'l)-2J3,4?5,7s9;135 '13a-octahyd.ro~2J5- methanopyrido[ I!,2!:4;,5]pyrazino[25l-b][l!3]oxazepine-i0~carboxarn;de that may be useful m the synthesis of (2R,5S513aR}-S }ydroxy-7,9-dioxo~ ~^
2J,4s5,7s9,135i3a~oetahydro-2,5~methanopyrido[1^2!:4,5]pyra2iRo[2 -b][! 53]oxazepine-10- earhoxamide. A form of a (2R,5S 3aK)~8 wdroxy-759~dIoxo-N-(2,456 rife
2,3,4,5 ,943,13a-octahydro-2,S-meth^
earboxamide may be an intermediate to the synthesis of (2R,5S,I3aR)-S-hydroxy-7,9-dioso- "N-(2,4,6 rinuorobenzyl)-2,3,4,5,7,9, 13,1 a~oc†ahydro-2,5~
methanopyrido [ 1 2' ;4,5]pyf zino[2s 1 -bj [ 1 ,3]oxazepine- 1 O-carboxarmde. A polymorphic form or polymorph or eocrystal may have properties such as bioavaiiabiiity and stability at certain conditions that may be suitable for medical or pharmaceutical uses.
[0069] A crystalline form of (2RJ5S,13aR)-8«hydroxy-7,9-dioxo-N-(2,4>6- trifluorobenzyl)~2,3,4,5,7,9, 13, 13a-octahydro-2,5~meihanopyrido[ 1 ',2';4,5]pyrazino[2, 1 - b][li3]oxazepine-10~carboxamide may provide the advantage of bioavailability and stability, suitable for use as an active ingredient in a pharmaceutical composition. Variations In the crystal structure of a pharmaceutical drug substaxs.ee or active ingredient may affect the dissolution rate (which may affect bioavailability, etc.), manufactiirabiilty (e.g., ease of handling, ability to consistently prepare doses of known strength) and stability' (e.g.. thermal stability, shelf life, etc.) of a pharmaceutical drug product or active ingredient. Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or delivery forms, such as solid oral dosage form including tablets and capsules. Compared to other forms such as non-crystalline or amorphous forms, crystalline forms may provide desired or suitable hygroscopicity, particle size controls, dissolution rate, solubility, purity, physical and chemical stability, ma ufacturabiiity, yield, and/or process control. Thus, crystalline forms of (211,58, 13aR)-8-hyd.roxy~7,9-dioxo~ ~(2,4,6~
trmuorobenzy!)-2,3,455,7;9J 3J ^
b][l,3]oxazepine-10"Carboxamide may provide advantages such as; improving the manufacturing process of an active agent or the stabiiiiy or storability of a drug product form of the compound or an acti ve ingredient and/or having suitable bioavailability and/or stability as an active agent,
[0070] The use of certain solvents has bee s found to produce different polymorphic forms of (21 ,58, 13a )-8 wdroxy~7 dioxo-N~^ 1 , 13a- octahydro-2,5-m.ethanopyrido[ 1 ',2!:4,5]ρ>τ ζίηο[2, 1 -b] [ 1 ,3]oxazepine- 10-carboxamide, including any one or more of polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, which may exhibit one or mors favorable characteristics described above. The processes fbr the preparation of the polymorphs described herein, and characterization of these polymorphs are described in greater detail below.
[0071] The compound name provided above is named using ChemBioDraw Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols. By way of example, the compound may be named or identified with common names, systematic or non- systematic names. The nomenclature systems and symbols that are commonly recognized in the art of chemistry including but not limited to Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (IUPAC). Accordingly, the compound structure provided above may also be named or identified as (2R,5S,13aR)-8-hydJx>xy-7,9- dloxo~N~(2,4J-triilnorobenz)d)-2;3,4,5,7,9, 13, i 3a-oetahydro~255- methanopyrido[r,2i:4,5]p>'razmo[2,l-b][1 ,3]oxazepine-10-carboxaniide under IUPAC and 2,S-Methanopyrido[ 1 ',2!:4,5]ρ>ΤΕ3ΕΪηο[2, 1 ~b] [ 1 ,3 Joxazepine- 10-carboxamide, 2^4,5, , , 13, 13a-oetahydro-8-hydro.xy~75^^^
(2R,5S513aR)- under CAS; CAS Registry Number 1611493-60-7.
[0072] in particular embodiments, crystalline forms and co-crystals of (2 55S,13aR)-8~ hydiOxy~7s9-di xo-N~(2J4,6-tri !uorobeiizyl)-2,3s4,5,7s9} 13, 13a-octahydro-2,5- methanopyrido[rs2t:455]p)'Tazino[2J -b][l ,3]oxazeplne 0-carboxamide are disclosed.
Formula 1, Form I
[0073] In one embodiment, provided Is polymorphic Form I of {2 .SS, 13aR)-8-hydroxy - 7,9~dloxo -N 2A6-trU¼o^
Figure imgf000016_0001
polymorph exhibits an X-ray powder diffraction (XRPD) pattens substantially as shown in FIG. L Polymorphic Form I may exhibit a differential scanning ealorimetry (DSC) thermogram substantially as shown in FIG. 7, Polymorphic Form I may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG. 10. Polymorphic Form I may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 13.
[0074] The terns, "substantially as shown in" when referring, for example, to an XRPD pattern, a DSC thermogram, or a TGA graph includes a pattern, thermogram, or graph that i not necessarily identical to those depicted herein, but that falls within the limits of experimental error or deviations when considered by o e of ordinary skill in the art.
[0075] Polymorphic Form I may have a unit cell as determined by crystal X-ray crystallography of the following dimensions: a ~ 11.4498 (4) A; b = 8.4767 (3) A; c = 1 .9163 (8) A; a - 90 °; β - 106.286 (2) °; and γ - 90 °.
[0076] In some embodiments of polymorphic Form I, at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all of the following (a)-(k) apply: (a) polymorphic Form I has an XRPD pattern substantially as shown in FIG. 1 ; (b) polymorphic Form Ϊ has a DSC thermogram
substantially as shown in FIG, 7; (c) polymorphic Form I has a TGA graph substantially as shown In FIG, 10; (d) polymorphic Form I has DVS graphs substantially as shown in FIG. 13; (e) polymorphic Form Ϊ has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a = 11.4498 (4) A; b - 8.4767 (3) A; c - 19.9163 (8) A; a = 90 °; β = 106.286 (2) °; and γ = 90 °; (£) polymorphic Form I has an endomermie event onset; (g) polymorphic Porta I has a moBociimc crystal system.; (h) polymorphic Form ί has a P2(l) space group; (I) polymorphic Form I has a volume of 1855.44( 12) A3; (j) polymorphic Fonn I has a Z value of 4; and (k) polymorphic Form Ϊ has a density of 1.609 g/cm3.
[0077] In some embodiments, polymorphic Form I has at. least one, at least two, at. least three, or all of the following properties:
(a) an XRPD pattern substantially as shown in FIG. 1 ;
(b) & DSC thermogram substantially as shown in FIG. 7;
(c) DVS graphs substantially as shown in FIG. 13; and id) a unit ceil, as determined by crystal X-ray crystallography, of the following dimensions: a = 11.4498 (4) A; b = 8.4767 (3) A; c = 19.9163 (8) A; = 90 °; β = 106.286 (2) °; and -/ = 90 °.
[0078] In some embodiments, polymorphic Form I has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 2e-reflections with the greatest intensity as the XRPD pattern substantially as shown in FIG. I ,
[0079] In certain embodiments, polymorphic Form I has an XRPD pattern comprising degree 20-refiections (÷/~ 0.2 degrees 2Θ) at 27.4, 13.1, and 17.4. In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 28~refleetions (÷/- 0.2 degrees 2Θ) at 27,4, 13.1, and 17.4 and one or more of the degree 28-reffeeiions (+/- 0.2 degrees 28) at 10.5, 20.6. and 25.0. In one embodiment, polymorphic Form Ϊ has an XRPD pattern comprising degree 29-re flections (+/- 0.2 degrees 28) at 27.4, 13,1, and 17.4 and one of the degree 28-ret¾ctiorjs (+/- 0.2 degrees 20) at 10,5, 20.6, and 25.0. In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 28~ref1ections (+/- 0,2 degrees 20) at 27.4, 13,1, and 17.4 and two of the degree 28-reilectlons (+/- 0.2 degrees 2Θ) at 10.5, 20.6, and 25.0. In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 28 -reflections { ·?·/·· 0.2 degrees 20) at 27.4, 13.1, and 17,4 and three of the degree 2Θ- reflections {-÷-/- 0,2 degrees 28) at 10.5, 20.6, and 25.0, In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 20 -reflections (+/- 0,2 degrees 2Θ) at 27.4,
IS 13,1, 7.4, 10.5, 20,6, and 25.0. In one embodiment, polymorphic Form I has an XRPD pattern comprising degree 28-refiections (+/- 0.2 degrees 28) at 27.4, 13.1, 17.4, 10.5, 20.6, 25.0 , 16.2, and 22.3. In one embodiment, polymorphic Form I has an XRPD pattern comprising any three degree 29-reflections (+/- 0,2 degrees 28) selected from the group consisting of 27.4, 13.1, 17.4, 10.5, 20.6, 25.0, 16.2, 22.3, 13.9, ί 1.4, and 9.3.
Formula I, Form II
[0080] in one embodiment, provided is polymorphic Form II of (2R,5 S, i 3aR)-8-hydroxy-
7J9-dioxo~N 2s456 rifi∞robenzyl)-2,3,4s5,759,13,13a-octahydro-2)
methaiiopyrido[ 1 f,2° :4,5 jpyrazino[2, i -b] [ 1 ,3]oxazep «e- 10-ear boxamide, wherein the polymorph exhibits an X-ray powder diffraction (XRPD) pattern substantially as shown in FIG, 2.
[0081] Polymorphic Form H may have a unit ceil as determined by crystal X-ray crystallography of the following dimensions: a = 8.5226 (7) A; b = 26.934 (2) A; c = 8.6861 (8) A; a = 90 °; β - 101.862 (2) °; and γ - 90 °.
[0082] In some embodiments of polymorphic Form II, at leas one, at least two, at least three, at least four, at least five, at least six, or all of the following (a)-(g) apply: (a) polymorphic Form II has an XRPD pattern substantially as shown in FIG. 2; (b) polymorphic Form II has a unit ceil, as determined by crystal X-ray crystallography, of the following dimensions: a - 8.5226 (7) A; b - 26.934 (2) A; c - 8.6861 (8) A; a - 90 ¾: β = 101.862 (2) °; and γ = 90 °; (c) polymorphic Form II has a monodi ne crystal system; (d) polymorphic Form II has a P2(l.) space group; (e) polymorphic Form II has a volume of 1951.3(3) A3; (f) polymorphic Form II has a Z value of 4; and (g) polymorphic Form II has a desisity of 1,537 Mg/m3.
[0083] In some embodiments, polymorphic Form II has at least one. or all of the following properties:
(a) an XRPD pattern substantially as shown in FIG. 2; (b) a unit celt, as determined by crystal X-ray crystallography, of the following dimensions: a - 8.5226 (7) A; b = 26.934 (2) A; c = 8.686! (8) A; a = 90 °; β = 101.862 (2) and v = 90 °.
[0084] In some embodiments, polymorphic Form II has an X PD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 2e-refkctions with the greatest intensity as the XRPD pattern substantially as shown in FIG. 2,
[0085] fn certain embodiments, polymorphic Form H has an XRPD pattern comprising degree 2Θ- eflections (+/~ 0.2 degrees 2Θ) at 6,6, 21.4, and 10.6. In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 20-reflections (+/- 0.2 degrees 2Θ) at 6.6, 21.4, and 10.6 and one or m.ore of the degree 28-refiectioris {+/- 0.2 degrees 2Θ) at 12.5, 16.2, and 14.3. In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 20-reflections τ/- 0.2 degrees 2Θ) at 6.6, 21.4, and 10.6 and one of the degree 29-reflections (+/- 0,2 degrees 2Θ) at 12.5, 16.2, and 14.3, In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 20~reflections (■■*/- 0.2 degrees 2Θ) at 6.6, 21.4, and 10.6 and two of the degree 20- reflections (+/- 0,2 degrees 20) at 12,5,
1 .2, and 14.3. In one embodiment, polymorphic Form II lias an XRPD pattern comprising degree 20-refiections (·÷·/- 0.2 degrees 2Θ) at 6.6, 21.4, and 10.6 and three of the degree 20- refiectlons (+/- 0,2 degrees 28) at 12.5, 16,2, and 14.3. In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 28-reflections (+/- 0.2 degrees 28) at 6,6, 21,4, 10,6, 12.5, 16.2, and 14.3, In one embodiment, polymorphic Form II has an XRPD pattern comprising degree 2e~refleetions (÷/- 0.2 degrees 20) at 6.6, 21.4, 10,6, 12,5, 16.2,
14.3, 25,6j and 23.5. In one embodiment, polymorphic Form II has an XRPD pattern comprising any three degree 20~f jections {+/- 0,2 degrees 2Θ) selected .from the group consisting of 6.6, 21.4, 10.6, 12.5, 16.2, 14.3, 25.6, and 23,5.
Formula 1, Form IE
[0086] In one embodiment, provided is polymorphic Form Hi of (2R,5S, 13aR)-8- hyd)T;xy-?,9-dioxo-N- 2:4,6 rii :orobenzyi}"2 !4 5;? 9d 3, 1 a-ociah ydro~2,5- methanopyrido[ I '^'^^p razmoP, 1 ~b] [ I„ 3]oxa2£pine- 10-carboxamide, wherein the polymorph exhibits an X-ray powder diffraction (XRPD) pattern substantially as shown in FIG, 3. Polymorphic Form III may exhibit a differential scanning calorimetry (DSC) thermogram substantially as shown in FIG, 8, Polymorphic Form. ΪΠ may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG, 11. Polymorphic- Form III may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 14.
[0087] Polymorphic Form III may have a unit ceil as determined by crystal X-ray crystallography of the following dimensions: a = 18,002 (2) A; b = 10.9514 (12) A; c = 20.687 (2) A; = 90 °; β == 107.770 (4) °; and γ - 90 °.
[0088] In some embodiments of polymorphic Form ΪΪΪ, at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all of the following (a)-(k) apply: (a) polymorphic Form III has an XRPD pattern substantially as shown in FIG. 3; (b) polymorphic Form III has a DSC thermogram substantially as shown in FIG. 8; (c) polymorphic Form ΪΪΙ has a TGA graph substantially as shown in FIG. 11; (d) polymorphic Form has DVS graphs substantially as shown in FIG. 14; (e) polymorphic Form ΙΙΪ has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a = 18.002 (2) A; b - 10.9514 (12) A; c - 20.687 (2) A; a - 90 °; β = 107.770 (4) l>; and y ::: 90 °; (f) polymorphic Form I has an endothermic event onset (g) polymorphic Form III has a monoelinic crystal system; (h) polymorphic Form III has a P2(l) space group; (i) polymorphic Form ΙΠ has a volume of 3884,0(8) A3; ( ) polymorphic Form III has a 2 value of 8; and (k) polymorphic Form III has a density of 1 ,537 g/cra3.
[0089] In some embodiments, polymorphic Form III has at least one, at least two, at least three, or ail of the following properties:
(a) an XRPD pattern substantially as shown in FIG. 3;
(b) a DSC thermogram substantially as shown in FIG. 8;
(c) DVS graphs substantially as shown in FIG, 1.4; and
(d) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a = 18.002 (2) A; h = 10.9514 (12) A; c = 20.687 (2) A; a = 90 °; β = 107.770 (4) °; md = 90 °.
[0090] In some embodiments, polymorphic Form III has an XRPD pattern displaying at least two, at least three, at least four, at least fi ve, or at least six of the degree 2G~refieetion,s with the greatest intensity as the XRPD pattern substantially as shown in FIG. 3.
[00 1] In certain embodiments, polymorphic Form III has an XRPD pattern comprising degree 28-reilections (-f/~ 0.2 degrees 20) at 20.0, 18.5, and 9,6. In one embodiment, polymorphic Form III has an XRPD pattern comprising degree 28-reflectioras (+/~ 0.2 degrees 20) at 20.0, 18,5, and 9.6 and one or more of the degree 29-refkctions (+/- 0.2 degrees 20) at 22.5, 14.0, and 25,0. In one embodiment, polymorphic Form III has an XRPD pattern comprising degree 20-re flections (-17- 0.2 degrees 20) at 20.0, 18.5, and 9,6 and one of the degree 28-refiections (+/~ 0.2 degrees 28) at 22.5S 14,0, and 25.0. In one embodiment, polymorphic Form III has an XRPD pattern comprising degree 2B-reflections (+/- 0,2 degrees 20) at 20.0, 18.5, and 9.6 and two of the degree 28~refleciions (+/~ 0.2 degrees 2Θ) at 22.5, 14.0, and 25.0. Is one embodiment polymorphic Form III has an XRPD pattern comprising degree 28-refiections (·÷-/- 0.2 degrees 20) at 20.0, 18.5, and 9.6 and three of the degree 28- reflections (+/- 0.2 degrees 28) at 22.5, 14.0, and 25.0, In one embodiment, polymorphic Form HI has an XRPD pattern comprising degree 28~refieolions (+/- 0.2 degrees 20) at 20,0, 18.5, 9.6, 22.5, 14.0, and 25.0, In one embodiment, polymorphic Form III has an XRPD pattern comprising degree 20-reflections (+/- 0.2 degrees 28) at 20,0, 18.5, 9.6, 22,5, .14.0, 25.8, 12.1, and 27.0, In one embodiment, polymorphic Form 111 has an XRPD pattern comprising any three degree 28~ref lections (+/~ 0.2 degrees 20) selected from the group consisting of 20,0, 18.5, 9,6, 22.5, 1.4,0, 25.0, 12.1, 27.0, 16.2, and 29.0.
Formula 1, Form IV
[0092] in one embodiment, provided is polymorphic Form iV of (2 ,SS, 13aR)-8- hydroxy»7,9-dioxo»N-(2,4,6~trlflnorobenzyl)-2,3,4s5,7,9sI3,13a ctahydn>2,5»
methanopyrido[1^2':455]pyra ino[2,l-h][ lJ3]oxa2;epine-10-carboxamide, wherein the polymorph exhibits an X~ray powder diffraction (XRPD) pattern substantially as s ow in FIG. 4.
[0093] Polymorphic Form IV may have a unit cell as determined by crystal X-ray crystallography of the following dimensions: a = 29.948 (2) A; b = 16,5172 (9) A; c = 13.2051 (8) A; a = 98 °; β = 108.972 (4) °; and γ = 90 °. [0094] In some embodiments of polymorphic Form IV, at least one, at least two, at least three, at least four, at least five, at least sis, or all of the following (a)-(g) apply: (a) polymorphic Form IV has an XRPD pattern substantially as shown in FIG, 4; (b)
polymorphic Form IV has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a - 29.948 (2) A; b - 16.5172 (9) A; c - 13.2051 (8) A; a = 90 °; β = 108.972 (4) °; and γ = 90 °; (c) polymorphic Form IV has a nionocikic crystal system; (d) polymorphic Form IV has a C2 space group; (e) polymorphic Form IV has a volume of 6177.3(7) A'; (f) polymorphic Form IV has a Z value of 12; and (g) polymorphic Form IV has a density of 1 , 84 Mg/irf .
[0095] in some embodiments, polymorphic Form IV has at least one, or all of the following properties:
(a) an XRPD pattern substantially as shown in FIG. 4;
(b) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a = 29.948 (2) A; b = 16,5172 (9) A; c = 13.2051 (8) A; = 90 °; p = .108.972 (4) °; and γ = 90 °.
[0096] In some embodiments, polymorphic Form IV has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of the degree 29~reiIectio«s with the greatest Intensity as the XRPD pattern substantially as shown in FIG, 4,
[0097] In certain embodiments, polymorphic Form IV has an XRPD pattern comprising degree 26~reflections (*/- Θ.2 degrees 20) at 16,3, 6.2, and 8,6, In one embodiment, polymorphic Form. IV has an XRPD pattern comprising degree 28~reflections {+/- 0.2 degrees 20) at 16.3, 6.2, and 8,6 and one or more of the degree 20~refiections (·÷-/- 0.2 degrees 26) at 22.7, 22.3, and 25.8. In one embodiment polymorphic Form IV has an. XRPD pattern, comprising degree 20-re flections (÷-/- 0.2 degrees 20) at 16.3. 6,2, and 8.6 and one of the degree 28-refkcticms (+/- 0.2 degrees 20) at 22.7, 22,3, and 25.8. In one embodiment, polymorphic Form IV has an XRPD pattern comprising degree 20-reflectIons (+/- 0,2 degrees 20) at 16,3, 6.2, and 8.6 and two of the degree 28-refleetions (+/- 0,2 degrees 20) at 22.7, 223, and 25.8. In one embodiment, polymorphic Form IV has an XRPD pattern comprising degree 20-reflections (+/- 0,2 degrees 28) at 16,3, 6.2, and 8,6 and three of the degree 20- reflections (+/- 0,2 degrees 2Θ) at 22.7, 22.3, and 25.8. Irs one embodiment, polymorphic Form IV has an XRFD pattern comprising degree 2G-re.0ecisons (+/- 0.2 degrees 20) at 16,3, 6.2, 8.6, 22.7, 22,3, and 25.8. in one embodiment, polymorphic Form ΪΥ has an XRPD pattern comprising degree 2e~refleetions (+/- 0.2 degrees 20) at 16,3, 6.2, 8.6, 22.7, 22,3, 25.8, 20.0, arid 8.7, In one embodiment, polymorphic Form IV has an XRFD pattern comprising any three degree 26-reflections (·*/- 0.2 degrees 28) selected from the group consisting of 16.3, 6.2, 8.6, 22.7, 22.3, 25.8 20,0, 18.7, 27.7, and 13.2,
Formula I, Form V
[0098] In one embodiment, provided is polymorphic Form V of (2R,5S,13aR.)-8- hydroxy~7,9-dioxG-N~(2A6 rifluoro
metljanopyrido[ s2':4,5]pyra ino[2,l-h][l,3]oxazepine-10 arboxaiTiide.
[0099] Polymorphic Form V may have a unit cell as determined by crystal X-ray crystallography of the following dimensions: a - 8.4993 (6) A: b == 8.7290 (8) A; c = 13.8619 (13) A; a - 99.278 (5) °; β - 101 ,427 (4) c; and y - 100.494 (4) *.
[0100] In some embodiments of polymorphic. Form V, at least one, at least two, at least three, at least four, at least five, or all of the following, (a)-(f) apply: (a) polymorphic Form V has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a - 8.4993 (6) A; b = 8.7290 (S) A; c = 13.8619 (13) A; a - 99.278 (5) °; β - 101.427 (4) °; and γ = 100.494 (4) °; (b) polymorphic Form V has a triclinic crystal system; (c)
polymorphic Form V has a 1*2(1) space group; (d) polymorphic Form V has a volume of 970.18(14) A3; (e) polymorphic Form V has a Z value of 2; a d (f) polymorphic Fonts V has a density of 1.573 Mg m3.
[01.01] In some embodiments; polymorphic Form V has the following properties:
[01.02] a nnit cell, as determined by crystal X-ray crystallography, of the following dimensions: a - 8.4993 (6) A; b - 8.7290 (8) A; c = 13.8619 (13) A; a = 99.278 (5) °: β = 101.427 (4) °; and γ - 100.494 (4) °.
Formula 1. Form VI
[0103] In one aspect, provided is polymorphic Form VI of a (2R.5S, 1.3a )-S-hydroxy-
Figure imgf000024_0001
metha.nopyrido[ I ', :4,5]pyrazino 2, 1 ~b] [ 153]oxazepme~ 10~carboxarolde.
[0104] Polymorphic Form VI may have a unit ceil as determined by crystal X-ray crystallography of the following dimensions: a ^ 1 .51.63 (5) A; b = 6.4S93 (2) A; c = 16.6066 (5) A; = 90 e; β - 103.5680 (13) °; and γ - 90 °.
[0105] Jo some embodiments of polymorphic Form VI, at least one, at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply: (a) polymorphic Form VI has a unit cell, as determined by crystal X-ray crystallography of the following dimensions: a = 19.5163 (5) A; b - 6,4593 (2) A; c = 16.6066 (5) A; a - 90 °; β = 103.5680 (13) °; and γ = 90 °; (b) polymorphic Form VI has a monoclinie crystal system; (c) polymorphic Form V has a P2(l) space group; (d) polymorphic Form VI has a volume of 2035.03(10) A3; (e) polymorphic Form V has a Z value of 4: and (f) polymorphic Form V has a density of 1.545 Mg/rrA
[0106] In some embodiments, polymorphic Form VI has the following properties:
(a) a unit cell, as determined by crystal X-ray etystallography, of the following dimensions: a = 19.5163 (5) A; b - 6.4593 (2) A; c = 16.6066 (5) A; a = 90 °; β = 103,5680 (13) Λ; and γ = 90 °„
Formula i. Form VII
[0107] In one embodiment provided is polymorphic Form VII of (2R,5S.J3aR)~8- hydroxy-7?9-dioxo- ~(2,4!6 ri.fluorobenzyl)-253,41557!9,13, 13a-octahydro-2.5- methanopyrido[ rs2,:4,S]pyraziri.o[2; l-b] [ 1 ,3 joxazepine- 1 G~earboxaro ide.
[0108] Polymorphic Form VII may have a unit cell as determined by crystal X-ray crystallography of the following dimensions: a :::: 30.785 (12) A; b = 16.685 (6) A; = 25.956 (10) A; a - 90 °; β - 108.189 (10) and γ - 90 °.
[0109] In some embodiments; of polymorphic Form VII, at least one. at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply; (a) polymorphic Form Vli has unit cell, as determined by crystal X-ray crystallography of the following dimensions: a = 30.785 (12) A; b = 16.685 (6) A; c = 25.956 (10) A; = 90 °; β = 108.189 (10) °; and y = 90 °; (b) polymorphic Form VII has a monoclinic crystal system; (c) polymorphic Form VII has a P2(l) space group; (d) polymorphic Form Vi has a votome of 12666(8) A''; (e) polymorphic Form VII has a Z value of 24; and (f) polymorphic Form VII has a density of 1.468 Mg.½3.
[01 10] in some embodiments, polymorphic Form VI has the following properties:
(a) a unit ceil, as determined by crystal X-ray crystallography of the following dimensions: a = 30.785 (12) A; b = 16.685 (6) A; c = 25,956 (10) A; a = 90 °; β = 108,189 (10) °; and γ = 90 °.
Formula I, Form VIII
[0111] In one embodiment, provided is polymorphic Form VIII of (2R,SS513aR)-8- hydroxy-7> -dioxo-' ~(2,4,6-trifl orotenzy!)-2,?u
methar!opyrido r,2':4s5]pyrazirio[2J-b][i J]oxazcpine-10-carboxamide.
[01 12] Polymorphic Form VIII may have a unit cell as determined by crystal X~ray crystallography of the following dimensions: a = 10.3242 (18) A; b = 10.7826 (17) A; e = 17.848 (3) A; a - 90 °; β - 105,578 (8) °; and γ - 90 °.
[0113] In some embodiments of polymorphic Form VOL, at least one, at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply; (a) polymorphic Form. Vlil bas a unit cell, as determined by crystal X-ray crystallography; of the following dimensions: a = 10.3242 (18) A; b - 10.7826 (1 ) A; c - 17.848 (3) A; a = 90 °; β - 105.578 (8) °; and γ = 90 °; (b) polymorphic Form VIII has a monoclinic crystal system; (e) polymorphic Form VIII has a C2 space group; (d) polymorphic Form VIII has a volume of 1913.9(6) A3; (e) polymorphic Form VIII has a Z value of 4; and (f) polymorphic Form VIII has a density of 1.560 Mg m3.
[0114] In some embodiments, polymorphic Form VET has the following properties:
(a) a unit ceil, as determined by crystal X-ray crystallography, of the following dimensions: a = 10.3242 (1 ) A; b - 10.7826 (17) A; c = 17.848 (3) A; a = 90 °; β = 105.578 (8) °; and γ = 90 °. Formula 11
[01 15] It Is desirable to develop a crystalline form of sodium (2R,5S,13aR)-7,9-dioxo~10-
((2 4,6-rrifluo:robenzyi)carba?noyi)-2!3,4,5.7,9, 13,13a-octaliydro~2,S-
Figure imgf000026_0001
that may be useful in the synthesis of sodium (2R.58, 13aR -?,9"dioxo- 10-((2,4,6-t.ri tluoro x;rs yi}carbamoyi)-
23,« ,5J. J 3 sa-oc hs'd^-2!5-irieihano y
olate. A form of sodium (2R,SSs I3aR)-7,9-dioxo-I0-{(25456 rifliioroberizyl)caibamoyl)- 2,3,4,5,7,9,13,13a~aetahydro-2, 5 -methanopyrido [ 1 \2':4,5]pyrazino 2., 1 -b] 1 ,3 ]oxazeph 8- oiate may be an intermediate to the synthesis of sodium (2R,5 S, 13aR)~7,9-dioxo- 10-((2,4,6- trifl«oroben¾4)carbaffioyl)-2,3,4,5,7,9s13s13a~oetahydro-2,5~
meiha^iopyrido[r,2,:4,5]pyra^mo[2,.l -b][i ,3|oxazepin-8-olate< A form of sodium
(2R,5S,13aR)~ ^dioxo 0 {2A6~trifl ^
255~msthanopyrido[1^2!:4,5]pyra2;ino[2,l-h][l,3]oxazepin-'8~oIate may be the final product in the synthesis of sodium (2R,5 S, 13 aR)-7,9-dsoxo- 10-((2,4,6~trifluorobenzyi)carbamoyl)- 2,3,4,5,7.9, 13,13a~octahydro~2,5-me&anopyrido[ 1 s,2!:4,5]pyrazino(2, 1 -b][ 1 ,3]oxazepin-8- olate. A polymorphic form or polymorph or eocrystal may have properties such as bioavailability and stability at certain conditions that may be suitable for medical or pharmaceutical uses,
[0116] A crystalline form of sodium (2Rs5SJ 3a )-7,9-dioxo-10-((2,4,6- trifluoroben yl)carbamoyl)-2,3,4,5JJ9913, 13a-oetahydro-2,5- methanopyrido[ri2':4;,5]pyraziuo[2J-bl[!!3]oxazepin-8-okte may provide the advantage of bioavailability and stability, suitable for use as an active Ingredient in a pharmaceutical composition. In certain embodiments, a crystalline forro sodium (2R,5S, 13aR)-7,9-dioxo- 10- ({2 , ,6-trifluorobenxy i)carbamoyi)»2,3,4J5,7,9, 13, i 3a-octahydro-2,5- inethanop>Tidoi l',2,:4,5]pyrazirio[2, 1 -b]{ l,3]oxazcpu 8-olate provides an advantage of improved bioavailability (Table 3) and/or stability (Table 4), Variations in die crystal structure of a pharmaceutical drug substance or active ingredient may affect the dissolution rate (which may affect bioavailability, etc.), anufaciurabi ί ity (e.g., ease of handling, ability to consistently prepare doses of known strength) and stability (e.g., thermal stability, shelf life, etc) of a pharmaceutical drug product or active ingredient. Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or deliver}' forms, such as solid oral dosage form including tablets and capsules. Compared to other forms such as noncrystalline or amorphous forms, crystalline forms may provide desired or suitable h groseopidty. particle size controls, dissolution rate, solubility, purity; physical and chemical stability; manufact rabiiity, yield, and/or process control Thus, crystalline forms of sodium (2R,55, 13aR)-7,9-df ooto-10«((2.,4I6 rifluorohe?izyl}carbamoyl)-2}3;4,5s?59s 13 J.3a~ octahydro~2;5-methamopyrido[i^2 5]pyra^^
advantages such as: improving the manufacturing process of an active agent or the stability or storability of a drug product form of the compound or an active ingredient, and/or having suitable bioavailability and/or stability as an active agent.
[0117] The use of certain solvents has beers found to produce different polymorphic forms of sodium (2R.5S, 13aR) 759~dioxo- 10-((2,4>6-triiluofoberizyl)carbainoyl)- 2,3,4,5,7,9,13, 13a-octahydro-2s5~methanopyrido[rs2':435]pyrazmo[2 ,1 -b][l,3]oxazepm~8~ olate, including polymorphic Form Ϊ, which may exhibit one or more favorable characteristics described above, in certain embodiments, Form I of sodium (2R,5S513aR)-7,9-dioxo-10- ((2s4,6 rifluorobenzyl)carbamoyl)-2,3J4,S57,9,13,13a«oetahydfo--2>5- methaiiopyrido[r,2!:455]pyrazino[2,l~b][l,3]oxazepirj-8-okte provides an advantage of improved bioavailability (Table 3) and/or stability (Table 4). The processes for the preparation of die polymorphs described herein and characterization of these polymorphs are described in greater detail below,
[01 IS] The compound name provided above is named using ChemBioDra Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols. By way of example, the compound may be named or identified with common names, systematic or non- systematic names, The nomenclature systems and symbols that are commonly recognized in the art of chemistry including but not limited to Chemical Abstract Service (CAS) and International Union of Pure and Applied Chemistry (iUPAC), Accordingly the compound structure provided above may be named or identified as sodium (2R,5S513aR}-7,9~dioxo-10- ({2,4,64rifluorobes¾¾'l)carbasBoyl)~2s3,4s537,9J13,13a-octahydro--25S- meihanopyrido[f52!:4,S)pyrazino[2,i-b][l,3joxazepin~8-oiate under IUPAC.
[0119] in particular embodiments, crystalline forms of sodium (2R,5S, 13aR)~759"dioxo~ i0-{{2s4,6 riflnoroben¾>d)carbanioyl)-2,3!455,7,9513J3a»octaliydro-2,5»
metfeanopyrido[i,,2, 4,5]p a ino[2J~b][L3]oxazepin43-olate are disclosed. Formula II Form I
[0120] In a certain embodiment novel forms of sodium (2RSSSJ 3 R)~7,9-dioxo~j.0~ ({2s436 riiluorobeiizy!)carbarnoyl)-2,3,4,5,7.,9, 13, 13a-ociahydro-2,5~
meihanopyrido[r52,:4,5]pyrazmo[25 'i~b][l53]c*xa2ep∞-¾-olats8 having the following structure (Formul II) are disclosed:
Figure imgf000028_0001
[0121] in a further embodiment, crystalline forms of sodium (2R,5Ss13aR)-7s9~di !xo-10- ((2,4,6~trifiuoroberi2yi)cafba£noyl)-2,3s4,5s759s 13, 13a~octahydro-2s5- methanop rido[l V2':4,5]pyra;dri [2,l-b][l J]c>xazepin-S-olate are disclosed.
[0122] in a certain embodiment sodium (2R,5S513aR)-7,9-dioxo-10-((2J4s6- tritluorobenzyi)carbamoy!)-2.3,4.5, 7,9, 13,13a-octahydro-2,S- methanopyTido[1^2¾,5]pyrazmo[2J-b][l,3]oxa2epin~S~olaie Form Ϊ is disclosed,
[0123] In one embodiment, provided is polymorphic Form I of sodium (2R;5SJ3a }»7,9 - iaxo-10-i(2s4,6~irMuQrobenzY^
methanop}'Tido[r,2,:4}5]pyrazino[2sl-b][L3]oxazepin~8-olate, wherein the polymorph exhibits an X-ray powder diffraction (XRPD) pattern substantially as shown in FIG, 5 and/or FIG. 16. Polymorphic sodium Form I may exhibit a differential scanning ca!orimetry (DSC) thermogram substantially as shown in FIG, 9, Polymorphic sodium Form I may exhibit a thermographic analysis (TGA) graph substantially as shown in FIG. 12, Polymorphic sodium Form I may exhibit dynamic vapour sorption (DVS) graphs substantially as shown in FIG. 15,
[0124] The term "substantially as shown in" when referring, for example, to an XRPD pattern, a. DSC thermogram, or a TGA graph includes a pattern, thermogram or graph that is not necessarily identical to those depicted herein, but that falls within the limits of experimental error or deviations when considered by one of ordinary skill in the art. [0125] Polymorphic sodium Form Ϊ may have a unit cell as determined by crystal X-ray crystallography of the following dimensions: a = 8.9561 (10) A; b = 13.9202 (14) A; c = 31,115 (3) A; a = 90 °; β === 90 °; and γ === 90 °.
[0126] In some embodiments of polymorphic sodium Form I, at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or all of the following (a)-(j) apply: (a) polymorphic Form I has an XRPD pattern substantially as shown in FIG. 5 and/or FIG, 16; (b) polymorphic sodium Form I has a DSC thermogram substantially as shown in FIG, 9; (c) polymorphic sodium Form I has a TGA graph substantially as shown in FIG. 12; (d) polymorphic sodium Form I has DVS graphs substantially as shown in FIG, IS; (e) polymorphic sodium Form I has a unit cell as determined by crystal X-ray crystallography, of the following dimensions: a = 8.9561 (10) A; b = 13.9202 (14) A; c - 31.115 (3) A; a - 90 °; β - 90 °; and γ = 90 °; (f) polymorphic sodium Form ί has an orthorhombic crystal system; (g) polymorphic sodium Form I has a P212I21 space group; (h) polymorphic sodium Form I has a volume of 3879.2 A3; (i) polymorphic Form I has a Z value of 4; and (j) polymorphic Form I has a density of L61.4
[0127] In some embodiments, polymorphic sodium Form I has at least one, at least two, at least three, at least four, or all of the following properties:
(a) an XRPD pattern substantially as shown in FIG. 5 and/or FIG. 16;
(b) a DSC thermogram substantially as shown in FIG. 9; (e) TGA graphs substantially as shown in FIG. 12;
(d) DVS graphs substantially as shown in FIG, 15; and
(e) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a = 8.9561 (10) A; b - 13.9202 (14) A; c = 31.115 (3) A; = 90 Λ; β = 90 °; and y = 90 °;
[0128] In some embodiments, polymorphic sodium Form I has an XRPD pattern displaying at least two, at least three, at least four, at least five, or at least six of fee degree 20-refiections with the greatest intensity as the XRPD pattern substantially as shown in FIG. 1 and/or FIG, 8,
[0129] In certain embodiments, polymorphic sodium Form I has an XRPD pattern comprising degree 2CJ~refieetions (-!/- 0.2 degrees 2Θ) at 5.5, 16.1, and 23,3. In one embodiment polymorphic sodium Form I has an XRPD pattern comprising degree 2Θ- reflections (+/- 0,2 degrees 2Θ) at 5.5, 16.1, and 23.3 and one or more of the degree 20- refleetions (+/- 0,2 degrees 20) at 22.1, 28.5, and 22.5. n one embodiment, polymorphic sodium Form I has an XRPD pattern comprising degree IB-reflections (+/- 0.2 degrees 28) at 5.5, 16.1, and 23.3 and one of the degree 29-reflections (+/- 0.2 degrees 20) at 22,1, 28.5, and 22,5, In one embodiment, polymorphic sodium Form I has an XRPD pattern comprising degree 28-refleetions (+/- 0.2 degrees 20) at 5,5, 16.1 , and 23,3 and two of the degree 2Θ- reflectkms (+/- 0.2 degrees 20) at 22.1, 28,5, and 22.5. In one embodiment polymorphic sodium Pons I has an XRPD pattern comprising degree 20-refleciions (+/- 0.2 degrees 2Θ) at 5.5, 16.1, and 23,3 and three of the degree 20-rsSections 0.2 degrees 2Θ) at 22.1, 28.5, and 22.5. In one embodiment, polymorphic sodium. Form I has an XRPD pattern comprising degree 20-reflections {+/- 0.2 degrees 20) at 5.5, 16,1, 23.3, 22.1, 28.5, and 22.5. In one embodiment, polymorphic sodium Form I has an XRPD pattern comprising degree 20- refleciions 0.2 degrees 20) at 5.5, 16.1, 23.3, 22.1, 28.5, 22.5, 19.5, and 26.6, in one embodiment, polymorphic sodium Form I has an XRPD pattern comprising any three degree 20-reflections (+/- 0.2 degrees 2Θ) selected from the group consisting of 5.5, 16.1, 23.3, 22.1, 28,5, 22.5, 19.5, 26.6, and 17.9.
Formula III
[0130] It is desirable to develop a crystalline form of potassium (2R,5S,13aR)-7,9-dioxo- IO"{(2,4,6-ttifluorobenz 4)carbanioyI)~2s3,4}5,7,9 35Oa-octahydro--255"
metharsopyrMo[1^2i:455]pyra¾ino[25l-b][ls3]ox32;ep¾~S"Ofete that may be useful in the synthesis of potassium (2R,SS,i 3a )-7,9-dioxo- 10-{(2A6-m^^
2,3,4,5,7,9,13,I3a-octaliydrQ~2s5^iete
oiate, A form of a potassium {2R,5S,13aR)-7,9-dioxo-I0-((2,4,6~trifluorobenzyl)earhamoyl)- 2,3,4,5,7,9,13, 13a~oetahydro-2,5~methanopyrido[I^2':4,5]pys-azino[24-b][1.3]osazepin"8" olate may be an intermediate to the synthesis of potassium (2R.5 S, 13aR)-7,9 -dioxo- 10 - ^^^iflno ob Bz Oc rbamo O^^^J ^.B^ a-oet hydro^^- methaiiopyr o[!'52,;4,S]pyrazmo[2}l-b][is3]oxazepin'-8--olaie. A polymorphic form or polymorph or cocrystal may have properties such as bioavailability and stability at certain conditions that may be suitable for medical or pharmaceutical uses.
[0131] A crystalline form, of potassium (2R55SJ3aR)-7!9-dioxo 0-{{2,4s6»
trifluorobeiizy 3)carbamoyl)~253.4,5,7,9,13, 13 a-ociahy dro-2, 5 - metMnopyrido r,2':4,5]pyrazirio[2, l-b][l,3]oxazepiri-8-oiale may provide the advantage of bioavailability and stability; suitable for use as an active ingredient in a pharmaceutical composition. Variations in the crystal structure of a pharmaceutical drug substance or active ingredient may affect the dissolution rate {which may affect bioavailability, etc.), manufaet rabllity {e.g., ease of handling, ability to consistently prepare doses of known strength) and stability {e.g., thermal stability, shelf life., etc) of a pharmaceutical drug product or active ingredient Such variations may affect the preparation or formulation of pharmaceutical compositions in different dosage or delivery forms, sack as solid oral dosage form including tablets and capsules. Compared to other forms such as non-crystalline or amorphous forms, crystalline forms may provide desired or suitable hygroscopieity, particle size controls, dissolution rate, solubility, purity, physical and chemical stability,
manufacturahility, yield, and/or process control. Thus, crystalline forms of potassium
(2R,5S, 13a )-7,9-dioxo- 10-i(254,6 rifluorobeii2 4)carban∞yl)-2,3,4,5,7i9; 13, 13a-octahydro~ 2 , 5 · methanopy ki [ 1 ',2* :4 , 5 ]pyrazi n [2, 1 -b J [ 1 , ]oxazepin~8-oiaie may provide advantages such as: improving the manufacturing process of an active agent or the stability or storability of a drug product form of the compound or an active ingredient, and/or having suitable bioavailability and/or stability as an active agent.
[0132] The use of certain solvents has been found to produce different polymorphic forms of potassium (2R,5S,13aR)-?.9~dioxo~lQ~((2,456-tri^
2,3,4^7, 3, Ba )ctahydro-2 methano
olate, including any one or more of polymorphic Forms I, II, and III which may exhibit one or more favorable characteristics described above. The processes for the preparation of the polymorphs described herein, and characterization of these polymorphs are described in greater detail below.
[0133] The compound name provided above is named using ChemBioDraw Ultra and one skilled in the art understands that the compound structure may be named or identified using other commonly recognized nomenclature systems and symbols. By way of example, the compound may be named or Identified with common names, systematic or non- systematic names. T he nomenclature systems and. symbols that are commonly recognized in the art of chemistry Including but not limited to Chemical Abstract Service (CAS) and Internationa! Union of Pure and Applied Chemistry (iUPAC). Accordingly, the compound structure provided above may be named or identified as potassium. (2R.5S, 13aR)-7,9-dioxo- 1 Q-((2,4s6-trifluorobenzyl)carba!Boyl)-2>3s4,S,7J9, 13,1 a-octah dro-2,5~
methanop>Tido[ ;2!:4,5]pyrazko 2Jl -b][l ,3]oxazepin~8~olate under iUPAC.
[0134] in particular embodiments, crystalline forms and co-crystals of potassium (2R,SS.13aR)-7,9^oxo-10^
2,5-rnethanopyndoi i:,2':4»5 jpyraviino 2. i.~b]['i 3] xazepm~8-oiatc are disclosed.
[0135] in yet a further embodiment, novel forms of potassium (2R.,5S.13a }-7,9-dioxo~ 10-{(2,4,6~ fluorobenzyl)carba i oyl)-2J54s5,759J3,13a-octahydro~2s5- metlianopyrido[i^2^4,5]pyrazino[2 -b][ l,31oxa2epI¾-S-aIates having the ibiiowing structure (Formula IB) are disclosed.
Figure imgf000032_0001
[0136] In yet another embodiment, potassium (2R,5S, 1.3aR)-?,9~dioxo- 10-{(2,4,6~ trlfino obenzy^c rbamoyl)^^^^;.?^, 13, 13a-ociahydro-2,S~
methauopyrido[r,2!:455]pyra^ino[2,! -b][lJ3]oxazepin-8-olate Form I is disclosed,
[0137] In yet another embodiment, potassium (2R,5S,I 3a )-7,9-dioxo-10~((2,456- trifiuorobenz 4)carbarnoyl)-2;3s435,7J513,13a~oetaliydro-2s5- methanopyrido[r,2':455]pyrazlno[2,l-b][l,3]oxazepiri"8"Olate Form II is disclosed.
[0138] In yet another embodiment, potassium (21 , 5$, 13aR)-7.9-dioxo- 10 ·((2,4,6· tritlaorobenzyl)carbamoy!)~2,3,455.7s9 1 , 13a-Qctahydro-'2.5"
methanop>Tido[l!,2':455]p Ta mo[2,l~b][l,3]oxazepIn-8-olate Form III is disclosed,
[0139] In a still other embodiment, hydrated potassium (2R,5S, 1 3aR)-7,9-dioxo- 10- ((2>4,6 Tifluorobenzyl)carbamoyl)-2s354s5s7,9313, 13a-octahydn 2,5- methaiiopyr Ido[ 1 ',2' :4,5]pyraz.ino[251 ~b] [ 1 , 3 ] oxazepin-8 -olate Is disclosed.
Formula 111, Form I
[0140] In one aspect, provided is polymorphic Form I of potassium (2R,S S, 13aR)-7,9- dioxo~ i.0~((254,6-trifiuo^
methanopyrido[ i\2':4,5 jpyrazino[2 J -b]| 1 ,3joxazepin~8~olaie.
[0141] Polymorphic potassium Form I may have a unit cell as determined by crystal X- ray crystallography of the following dimensions: a = 32.0409 (11) A; b = 10,2935 (4) A; c = 15,469! (?) A; a - 90 β - 90 °; and γ - 90 °,
[0142] In some embodiments of polymorphic potassium Form I, at least one, at least two, at least three, at least four, at least five, or all of the followkg (a)-(f) apply: (a) polymorphic potassium Form I has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a == 32.0409 (11) A; b - 10,2935 (4) A; c - 15.46 1 (7) A; a = 90 °; β
90 °; and γ ::: 90 c; (h) potassium Form I has an orthorhombic crystal system; (c) polymorphic potassium Form I has a P 21 21 2 space group; (d) polymorphic potassium Form I has a volume of 5101.9(4) A3; (e) polymorphic potassium Form I has a Z value of 8; and (!) potassium Form I has a density of 1.498 Mg m3,
[0143] In some embodiments, polymorphic potassium Form 1 has the following properties:
(a) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a - 32.0409 (11) A; b - 10.2935 (4) A; c = 15.4691 (?) A; a = 90 °; β = 90 °; and γ = 90 °,
Formula III, Form II (Dimer)
[0144] In one aspect, provided is polymorphic Form II of potassium (2R55SJ 3a )-?,9- dioxo»i0-{(2!4,6-trifluoroben¾yl)carbaffioyl)-2,3s4,5,7,9,l 3, 13a~ocf.ahydro-2,5~
methanopyrido! 1 :4, 5jpyrazino[2s 1 ~b] [ 1 ,3joxazepm-8-o late.
[0145] Polymorphic potassium Form II may have a unit cell as determined by crystal X- ray crystallography of the following dimensions: a = 32.0285 (17) A; b = 10.3029 (7) A; c = 15,5363 (10) A; = 90 °; β = 90 °; and γ = 90 °.
[0146] In some embodiments of potassium Form II, at least one, at least two, at least three, at least four, at least five, or all of the following (a)~(f) apply: (a) polymorphic potassium Form II has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a - 32.0285 (17) A; b - 10.3029 (7) A; c = 15.5363 (10) A; a - 90 °; β = 90 °; and γ - 90 *; (b) polymorphic potassium Form II has an orthorhombic crystal system;
(c) polymorphic potassiwm Form ΪΙ has a P 21 21 2 space group; (d) polymorphic potassium Form II has a volume of 5126,8(6) A3; (e) polymorphic potassiwm Form II has a Z value of 4; and (f) polymorphic potassium Form Π has a density of 1.336 Mg½3.
[0147] In some embodiments, polymorphic potassium Form II has the following properties:
(a) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a - 32,0285 (17) A; b = 103029 (7) A; c = 15.5363 (10) A; a = 90 °; β = 90 °; and γ = 90 °.
Formula III, Form III
[0148] In one aspect, provided is polymorphic Form III of potassium (2R,5S,I3aR)-7,9~ dioxo-10-((2,4,6-trii!uorobenzyl)carbamoyl)-2>354,5 ,9s13513a-octahydro-2,5~
rnethanopyrido[rs2!:4,53p5'Tazirio[2,l-b][l,3]oxazspin-8-olate.
[01 9] Polymorphic potassium Form III may have a unit ceil as determined by crystal X- ra crystallography of the following dimensions: a ::: 8.8412 (3) A; b :::: 10.8837 (4) A; c = 13.9107 (5) A; a - 71.3620 (1) *; β - 76.343 (2) *; and y - 82.943 (2) °.
[0150] In some embodiments of polymorphic potassium Form III, at least one, at least two, at least three, at least four, at least five, or all of the following (a)-(f) apply: (a) polymorphic potassium Form ΙΠ has a unit cell, as determined by crystal X-ray
crystallography, of the following dimensions: a = 8.8412 (3) A; b :::: 10,8837 (4) A; c = 13.9107 (5) A; a = 71.3620 (1) °; β === 76,343 (2) °; and γ - 82.943 (2) °; (b) potassium Form ΠΪ has a triclmic crystal system; (c) polymorphic potassium Form III has a PI space group;
(d) polymorphic potassium Form HI has a volume of 1230.86 (8) A3; (e) polymorphic potassium Form III has a Z value of 2; and (f) potassium Form IU has a density of 1,483 Mg/m
[0151 ] In some embodiments, polymorphic potassium Form III has the following properties:
! . a unit ceil, as determined by crystal X-ray crystallography, of the following dimensions a = 8.8412 (3) A; b - 10.8837 (4) A; c = 13.9107 (5) A; a = 713620 (1) °; β = 76,343 (2) °; and γ = §2.943 (2) °
Co-Crystals
Formula 1 Citric Acid Co-Crystal
[0152] In another embodiment, a citric acid co-crystal of (2R,5SJ13aR)-8-hydroxy-7,9- dioxo- 254.6 rifluoroben yl)~2,3,4s5 ,9513J3a-octaI ydiO-2J5~
metham¾pyrido[i,,2f:4sS]pyrazmo[2J-b][L3]oxazepine-I O-carbo¾amide is disclosed.
[0153] In one embodiment, provided is Formula I citric acid co-crystal of (2R,5S513aR)- 8»hydroxy-7>9~di x -N-(2,4,6-trifluorobsi5zyl)~253!4555759i 13, 13a-o ahydro~2,5- methaiiopyr o[ 52 4,5]pyrazino[2,i-b][l,31oxazepme-10-carboxamide.
[0154] Formula Ϊ citric acid co-crystal may have a unit cell as determined by crystal X~ ray crystallography of the following dimensions: a = 7,4315 (6) A; b = 15.5755 (13) A; c = 15.6856 (13) A; a - 88.784 (2) °; β - 77.029 (2) °; and γ - 76.832 (2) °.
[0155] in som embodiments of Formula I citric acid co-crystal, at least one, at least two, at least three, at least tour, at least five, or all of the following (a)~(g) apply: (a) Formula I citric acid co-cry stal has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a = 7,43.15 (6) A; b - 15.5755 (13) A; c - 15.6856 (13) A; = 88.784 (2) °; β = 77.029 (2) °; and γ 76.832 (2) °; (b) Formula Ϊ citric acid co-crystal has a triciraic crystal system; (c) Formula I citric acid co-crystal has a PI space group; (d) Formula I citric acid co-crystal has a volume of 1721.9(2) A3; (e) Formula I citric acid co-crystal has a Z value of 2; and (f) Formula I citric acid co-crystal has a density of 1.608 g/nr.
[0156] In some embodiments, Formula ϊ citric acid co-crystal has all of the following properties:
(a) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a = 7.4315 (6) A; b = 15.5755 (13) A; c = 15.6856 (13) A; « = 88,784 (2) °; p = 77.029 (2) °; and γ = 76.832
(iy,
Formula 1 Fumaric Acid Co-Crystal
[0157] in a certain embodiment, a fumaric acid co-crystal of (2 s5S}I3aR)-8-hydroxy~ 7 ~dioxo~N-{2,4,6 riftuoroben yl)-2,354,5!7J9!13s13a'-octahydro»2i5- roetlMnopyrido[I^2':455]p ^a isio[2J-b][1 ]oxazepirie-10-carboxamide is disclosed.
[0158] In one aspect, provided is Formula I fumaric acid co-crystal of (2R,5S513aR)-8- hydrox -7.9-diox^^
methanopyrido[ 1 ^2!:4SS jpyrazko[2, 1 -bj 11 ,3]oxazepine- 10-carboxamlde,
[0159] Formula I fumade acid co-crystal may have a unit cell as determined by crystal X- ray crystallography of the following dimensions: :::: 26.767 (5) A; b = 8.2313 (14) A; c = 24.089 (4) A; = 90 °; β - 99,283 (4) and γ - 90 ¾.
[01 0] In some embodiments of Formula I fumaric acid co-cry tals at least one, at least two, at least three, at least four, at least five, or ail of the following (a)-(f) apply; (a) Formula I fumark acid co-crystal has a unit cell, as determsned by crystal X-ray crystallography, of the following dimensions: a = 26.767 (5) A; b - 8.2313 (14) A; c - 24.089 (4) A; - 90 °; β = 99.283 (4) °; and γ 90 °; (b) Formula Ϊ fumaric acid co-crystal has a monoclmic crystal system; (c) Formula I fumaric acid co-crystal has a C2 space group; (d) Formula I fumaric acid co-crystal has a volume of 5237.9(16) A3; (e) Formula i fumaric acid co-crystal has a Z value of 8; and (f) Formula Ϊ fumaric acid co-crystal has a density of 1.503 Mg m3,
[0161] In some embodiments, Formula I fumark acid co-crystal has all of the following properties:
(a) a unit cell, as determined by crystal X-ray crystallography, of the following dimensions a = 26.767 (5) A; b :::: 8.2313 (14) A; e = 24.089 (4) A; a = 90 β = 99.283 (4) °; and γ = 90 c. Formula 1 Oxalic Acid Co-Crystal
[0162] In yet another embodiment a oxalic acid co-crystal of (2R.5S, 13ail)-8-hydroxy-
Figure imgf000037_0001
met hanopyrido[ 1 \25:4.5 )pyrazmo(2, 1 ~b] [1 ,3]oxaxepme- 10-carboxamide is disclosed.
[0163] In one embodiment provided is Formula I oxalic acid co-crystal of (2R,5SJ 3aR)- S- ydroxy-T^-dio ^
methanopyrido[ 1 's2':4s5]pyra2ko[2, 1 -b] [ 1 ,3]oxazepine- 10-carboxamide ,
[0164] Formula I oxalic acid co-crystal may have a unit ceil as determined by crystal X- ray crystallography of the following dimensions: a ~ 7.8562 (3) A; b = 14.5031 (5) A; c = 19.9756 (7) A; a = 90 °; β = 101.291 (2) °; md y = 90 c,
[0165] In some embodiments of Formula i oxalic acid eo-crystal, at least one, at least two, at least three, at least four, at least five, or ail of the following (a)-(f) apply: (a) Formula Ϊ oxalic acid co-crystal has a unit cell, as determined by crystal X-ray crystallography, of the following dimensions: a - 7.8562 (3) A; b = 14.5031 (5) A; c = 19.9756 (7) A; a === 90 °; β = 101.291 (2) °; and y ::: 90 D; (b) Formula Ϊ oxalic acid co-crystal has a rnonoclink crystal system; (c) Formula I oxalic acid co-crystal bas a P2( l) space group: (d) Formula Ϊ oxalic acid co-crystal has a volume of 2231.95(14) A5; (e) formula I oxalic acid co-crystal has a Z value of 4; and (f) Formula Ϊ oxalic acid co-crystal has a density of 1.604 g cm3.
[0166] Irs some embodiments, Formula I oxalic acid co-crystal has the following properties:
(a) unit cell, as determined by crystal X-ray crystallography, of the following dimensions a - 7.8562 (3) A; b - .14.5031. (5) A; c = 19,9756 (7) A; a = 90 °; β = 101.291 (2) °; and γ = 90 °.
[01 7] In certain embodiments, Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 29-reflections (+/- 0.2 degrees 29) at 19.1. 14,5, and 9.1. In one embodiment, Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 20- refleetions (+/- 0.2 degrees 20) at 1.9.1 , 14.5, and 9,1 and one or more of the degree 29- reflections (+/- 0.2 degrees 2Θ) at 7.6, 26.5, and 17.1. In one embodiment, Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28-reflectlons (+/- 0,2 degrees 2Θ) at 19.1, 14.5, and 9/1 and one of the degree 20-reflections (÷/- 0.2 degrees 2Θ) at 7.6, 26.5, and 17.1. In one embodiments Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28-reilectiorts (+/- 0.2 degrees 20) at 19.1, 14.5, and 9,1 and two of the degree 20- reflections (+/- 0,2 degrees 2Θ) at 7,6, 26.5, and 17.1, In one embodiment Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 29-reflections (+/~ 0.2 degrees 28) at 19.1, 14.5, and 9.1 and three of the degree 20-reflections (+/- 0.2 degrees 2Θ) at 7.6, 26.5, and 17,1. In one embodiment, Formula I oxalic acid co-crystal has an XRPD pattern comprising degree 28~refleetions (÷/- 0,2 degrees 2Θ) at 19.1, 14.5» 9.1, 7,6, 26.5, and 17,1. In one embodiment, Formula 1 oxalic acid co-crystal has an XRPD pattern comprising degree 29- reflections (+/- 0.2 degrees 2Θ) at 19.1, 14,5, 9.1, 7.6, 26,5, 17.1, 21.8, and 39.4. In one embodiment, polymorphic Formula 1 oxalic- acid co-crystal has an XRPD pattern comprising any three degree 20-refiections {+!- 0.2 degrees 2Θ) selected from the group consisting of .19.1., 1.4.5, 9.1, 7.6, 26.5, 17.1, 21.8, 39.4, 29.7, and 11.6.
Pharmaceutical Compositions
[0168] For the purposes of administration, in certain embodiments, the compounds described herein are administered as a raw chemical or are formulated as pharmaceutical compositions. Pharmaceutical compositions of the present Invention comprise a compound of Formulas (I). (I ) or (ill), including forms and co-crystals thereof, and a pharmaceutically acceptable earrier, diluent or excipient The compound of Formulas (Γ), (II), or (III) is present in the composition in an amount which is effective to treat a particular disease or condition of interest. The activity of compounds of Formulas (I), (II), and (III) can be determined by one skilled in the art, for example, as described in co-pending application. U.S. Serial No, 14/133,855, filed December 19, 2013 entitled "POLYCYCLiC-CARBAMOYLPY JDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE". The activity of compounds of Formulas (I), (II), and (III) can also he determined by one skilled on the art, for example, as described, in co-pending PCX Serial No, US2G 13/076367, filed December 19, 2013 entitled, "POLYCYCLIC-CAPvBAMOYLPYRlDONE COMPOUNDS AND THEIR
PHARMACEUTICAL USE," Appropriate concentrations and dosages can be readily- determined by one skilled in the art. In certain embodiments, a compound of Form ulas (I)s (H), and/or (III) is present in the pharmaceutical composition in an amount from about 25 mg to about 500 mg. In certain embodiments, a compound of Formulas (I), (II), and/or (III) is present in the pharmaceutical composition in an amount of about 100 mg to about 300 mg. In certain embodiments, a compound of Formulas (I), (II), and/or (ΠΪ) is present in the pharmaceutical composition in an amoun t of about 5 mg to about 00 mg. In certain embodiments, a compound of Formulas (I), (IT), and/or (III) is present in the pharmaceutical composition in an amount of about 25 mg to about 100 mg. In certain embodiments, a compound of Formulas (I), (II), and/or (III) is present In the pharmaceutical composition in an amount of about 50 mg to about 1.00 mg. In certain embodiments, a compound of Formula (1), (ΪΪ), and/or (III) Is present in the pharmaceutical composition in an amount of about 5 mg, 25 mg, 50 mg, 75, mg, 100 mg, 200 mg, 300 mg, 400 mg or about 500 mg.
Formula I
[01 9] Provided are also compositions comprising at least one, at least two, at least three, at least four, at least five, at least six, at least seven, or all of polymorphs {e.g., any one or more of Formula Ϊ polymorphic Forms I, U, III, IV, V, VI, VII and VIII) as described herein. I a particular embodiment, a composition comprising one of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising two of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising three of Formula I. polymorphic Forms L II, III, IV, V, VI, VII, and VIII described herein is provided. In a particular embodiment a composition comprising four of Formula 1 polymorphic Forms 1, XL III, IV, V» VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising five of Formula I polymorphic Forms I, II, III. IV, V VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising six of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising seven of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII described herein is provided. In a particular embodiment, a composition comprising eight of Formula I polymorphic Forms I, II, HI, IV, V, VI, VII, and VIII described herein is provided. In other embodiments, the compositions described herein may comprise substantially pisre polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
[0170] In some embodiments, the composition comprises a polymorphic form of (2 ,5SS i S^-g-hydroxy-T^-dlo o-N-CZ^^-iTiil ioro sn ^'O^J^.SJ^, 13,13a~oe†ahydn 2,5~methanopytido[ I !,2' :455 |pyrazino( 2, l~b][l .SJoxaz-epine- 10- arboxamide. In certain embodiments are provided compositions comprising a polymorphic form as described herein, wherein the (2R,5S, 1 Sa^-S-hydrox -T^-di so-N-C ^^-t fluoroben yl)^,;?^^,?,1}, 13,13a- octa ydro-2,5-methanopyrido[ 1 's2! :4,S]pyr azino[2, 1 -b] [ 153]oxa epis e~ 10-carboxamidc within the composition is substantially pure (i.e., substantially pure Form i. Form II, Form Hi, Form Γν', Form V. Form VI or Form VII or Form VIII), I particular embodiments of compositions comprising a polymorphic form of (2R,5S, 13aIt)-8-hydroxy-7,9-dioxo-N- (2,4s6-trIf!uorobenzyl)-2,3,4,557,95i3,I 3a-octahydro-2,5- methanopyrido[1^2';4,5]pyrazlno[2,l-h][l ,3]oxa i;pirie-i0-carboxaniide, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%
Figure imgf000040_0001
2,3A5 ,9, 13J 3a-octahydro-2,5-m
carboxamide present in the composition is one of the polymorphic forms disclosed herein. In certain embodiments, the composition includes at least about 50%, at least about 60%, at least about 70%. at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of one of the polymorphic forms of (2R,5S, 13aR)-8 iydroxy-7,9-dioxo-N-(2,4,6-irif]uorobenzyl)- 2,3,4,5,7,9, .13, 13a-octahydro-2,5-methar!Op>'Tido[i,,2,:4,5]pyrazino[2, 1 -b][.l ,3]oxazepke-10~ carboxamide.
[0171] in other embodiments of compositions comprising a polymorphic form disclosed herein, less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of (2R,5 S, 1 ;½R}-8-hydrox.y-7,9-dioxo~ ^
trifl uor oben yl)-2,3 ,4,5 ,7, 9, 13 , 13 a-octahydro^
b] [ i ,3]oxazepine- 10-carboxamids present in the composition are other polymorphs of (2R,5SJ 3aR)^¼'droxy-7,9^
2s5~methanopyrsdo[ 1 ^2':4,S]pyrazino[2, 1 -b] [ 1 ,3 Joxazepine- 10 -carboxamide and/or impurities.
[0172] In yet other embodiments of compositions comprising the polymorphic forms disclosed herein, impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present, im urities may, for example, include by-products from synthesizing (2RS5 S, 13aR)-8½droxy-7.9-dioxo~ -{2A^
2,3,4,5,7,9, 13, ! 3a KtahydrO"2J5-methaEopyrido[ ,2,:4„5]pyrazmo[2 ,I >][l,3joxazepme-lG- carboxarosde, contaminants, degradation products, other polymorphic forms, amorphous form, water, and solvents, in certain embodiments, impurities include by-products from the process of synthesizing i2R,5S513aR)~8-hydroxy-?,9~dio¾o~N-(2,4s6~trifto^
2,3,4,5,7,9, 13, 13a~oct3hydro~2.S"m
earboxamide. In certain embodiments, Impurities include contaminants from the process of syHthesteing (2R,5S 3aR)~8-hydrGx
2,3,4,5,7,9,13513a-octahydro-2i5-methanop5¾do[ ,2!>4>5]pyrazino[2>i~b][l33]oxazepke-10- carboxamide, lu certain embodiments, impurities include degradation products of
(2R,5S 3aR)-8½droxy-7,^
235-me4aBopyrido[l\2!:4,5]p Tazino[2>i~b][1 ]oxazep3ne-10-carhQxamide, In certain embodiments, impurities include other polymorphic forms of (2R,5S,13aR)-8-hydroxry-7s9- dioxo-N"(2,4}6 rif!uorobenzyi)~2s3;4,5,7,9513, 13a-octahydro--2,5- methanopyrido[ 1 ',2' :4, 5]pyrazino[2, I -b] [ I,3]oxazepine- ! 0~earhoxamide. In certain embodiments, impurities include water or solvent. In certain embodiments of compositions comprising a polymorphic form disclosed herein, impurities are selected from the group consisting of by-products from synthesizing (2R,5S,13aR)~S~hydroxy~7s9~dioxo~M~(2s4,6- trifjuorobenzy!)-2,3s4,S,?^
b][l,3]oxa splne-10-earboxamides contaminants, degradation products, other polymorphic forms, water, solvents and combinations thereof.
[0173] In yet other embodiments, the composition comprising a polymorphic form disclosed herein has less than about 5%, less man about 4%, less than about 3%, less than about 2%. or less than about 1% by weight of amorphous or non -crystalline (2R,5S, 13aR)-8- d o ^^-dio o-N^^.^- fluo obe zyO^^^ J^jajSa-octahyd o^jS- methan yrido^^'^Sjpy a i op,!^
Formula 11
[0174] Provided are also compositions comprising at least one polymorph {e.g., any one or more of Formula II polymorphic Forms Ϊ) as described herein. In a particular embodiment, a composition comprising Formula ΙΪ polymorphic Form 1, described herein is provided, in other embodiments, the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities,
[0175] In some embodiments, the composition comprises a polymorphic form of sodium (2R,5S513aR)~7,9-dioxo~ 10-({234:,6-trifJuoroberiz 'l)carbanioyl)~2s3,4s5,759, 13,13a~oetahydro- 2,5"meihanopyrido[1^2,:4,5]pyrazmo[2,l-b3[l,31oxazepin-8«oiate. In certain embodiments are provided compositions comprising a polymorphic form as described herein, wherein the sodium (2R,SS 3aR)-7s9-dioxo-10-((254,0-tri^
octahydro-2,5-methanop> ido[r,2i:4,5]pyrazino[2,l'-b][l}3]oxazepin-§-olate within the composition is substantially pure (i.e., substantially pure Form I). In particular embodiments of compositions comprising a polymorphic form, of sodium (2Rs5SJ3aR)-7,9~dioxo-lG- ({2 A6~tr3f1uorobenzy l)eatbamoyl)-2,3 AS.,7,9, 13, 13a~octahydro-2,5~
meihanopyrido[rs2,:4?5]pyrazmo[2,l-b][.U3]oxazepin-8-olate, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% ot\sodium (2R,SS,13aR)~7s9-d^
2,3,4,5,7,9,13 J 3a~ociahydsO~2}5-meihanopyrido[l ',2':4,5]pyrazmoj;2 J -b][l,3]oxazepin~8- olate present in the composition is Formula II, Form. I, disclosed herein. In certain embodiments, the composition includes at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least, about 98%, or at least about 99% of Form I of sodium (2R,5S,!3aR)-7,9~dioxO"10-((2,4,6-^
2,5-methanopwido[r»2f;4,5]p> azino[2,l-b][l Jjosazepin-8-oiate.
[0176] In other embodiments of compositions comprising a polymorphic form disclosed herein, less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of sodium (2R,5S, 13aJl)-7,9-dioxo~ i 0-((2,4.6- trifluorohenzyi)cai-bamoyl)-2,3,4,5,7,9, 13,13a~octahydro-2,5- methaiiopysido[l!,2?:4>5]pyrazino[25l"b][l,3]oxazepin-8-olate present In the composition are other polymorphs of sodium (2R,5S513aR)-7,9"dioxo~iO-((2,4,6-triOuorohenzyl)carbanioyl)~ 2,3,4,5,7,9, 13, 13a~oetahydro-2,5-metbanopyrido[ l!,2f:4,5]pyrazino[2, 1 -b][1 ,3]oxazepin~8~ elate and/or impurities.
[0177] In yet other embodiments of compositions comprising the polymorphic forms disclosed herein, impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present. Impurities may. for example, include by-products from synthesizing sodium (2 s5SJ3a )-7,9-diaxo-10-((2,4,0^
2,3,4,5 ?9S 13, 13a 5Ctahydro~2s5-methanop} id
olate, contaminants, degradation products, other polymorphic forms, amorphous form, water, and solvents. In certain embodiments, impurities include by-products from the process of synthesizing sodium {2R,5Si13aR.)-759"dIoxo-10-{(2,4,6 rifluoroben£yi)carbamoyi)- 2.3,4 J,7,9!i343a-octahydro-2,5-Biethanopyrido 32':4,5]pyi-azino[2, l~b][!,3]oxazepm-8- olate. In certain embodiments, impurities include contaminants from the process of synthesizing sodium (2R,5S, 33aR)-7,9-dioxo- 10-((2A6-triftuorobenzy{}carbam^
23,4,5,7,9,13, 13a-octahydro-2^
olate. In certain embodiments, impurities include degradation products of sodium
(2R, 58 J.3 aR)-7,9~dlox^
255~methanopyrido[ F^'^SJp aanop, l-b][ 1 ,3]Qxazepm~g~olate, n certain embodiments, impurities include other polymorphic forms of sodium (2R,5S, 13aR}~7,9-dk>xo-10~((2,4,6 - trifluorobefizyl)carbamoyl)-2,3,4,5,?59, 13,13a-ociahydro-2,5- methan ridol ^'.^^jpyra irjo^jl-bjl l^joxaze isi-S-olaie, In certain embodiments, impurities include water or solvent. In certain embodiments of compositions comprising a polymorphic form disclosed herein, impurities are selected from the group consisting of by* products from synthesizing sodium (2 ,5S,13aR)-7,9~dioxo-I0-((2,4,6- trifluoroben2;yl)carbamoyl)-233,4s5,7s9,13sl3a-octahydro-255~
methanopyrido[l\2':4,5]p azino 2.,I--b][l,3]oxazepin-S--olate, contaminants, degradation products, other polymorphic forms, water, solvents and combinations thereof.
[0178] In yet other embodiments, the composition comprising Formula II, Form I disclosed herein has less than about 5%, iess than about 4%. less than about 3%, less than about 2%, or less than about 1% by weight of amorphous or non-crystalline sodium
(2R.5S, 13aR)-7,9-dioxo- 10~({2,4,6 rifluorobenzyl)carbainoyl)"2i334,5,7,9, 13, .13a-octahydro- 2,5-methanop ido[rs2!:4551pyrazin.o[2J -b][ls3]oxazepin-8"Oiate.
[0179] in some embodiments, the term "substantially pure" or "substantially free" with respect to a particular polymorphic form of a compound means that the composition comprising the polymorphic form contains less than 95%, less than 90%, less than 80%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 40%, less than 30%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% by weight of other substances, including other polymorphic forms and/or impurities. In certain embodiments, "substantially pure" or "substantially free of refers to a substance free of other substances, including other polymorphic forms and/or impurities. Impurities may, for example, include by-products or left over reagents from chemical reactions, contaminants, degradation products, other polymorphic forms, water, and sol vents.
Formula III
[0180] Provided are also compositions comprising at least one, or all of polymorphs {e.g. , any one or more of Formula III polymorphic Forms I, II, and III) as described herein. In a particular embodiment, a composition comprising one of Formula III polymorphic Forms I, II, and III described herein is provided. In a particular embodiment, a composition comprising two of Formula ΙΙΪΙ polymorphic Forms I, II, and III described herein is provided, In other embodiments, the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
[0181] In some embodiments, the composition comprises a polymorphic form of potassium (2R,5S, 13aR)-7,9~dioxo-l O- ^^-iriiluorobe ^ca b moyl)^^^^,?^, 13,13a- oe-tahydro~2,5-methanopyrido[!^2':4,S]p ^ In certain embodiments are provided compositions comprising a polymorphic form as described herein, wherein the potassium (2R,5S, 13aR)~?,9~dioxo- 10-{(2,4,6-trifluorobenz l}c6.rba oyl)- 2s3A5,7,95I3,13a~QCtahydr0-2,5~meta
o!ate within the composition is substantially pure (I.e., substantially pure Form I, ΪΪ, and/or III). In particular embodiments of compositions comprising a polymorphic form of potassium (2R,5S, 13aR)-7,9-dioxo- 10-i(254,6-trifluoroben:¾4)eaA^
octahydro~2,5-metha¾opyrido[r,2':4,5]pyraK?no[2, l-h'][l ,3]oxa∞pin~8~olate, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of potassium (2 55S,13aR)-7,9-dioxo-10-({2,456- irifiuoroben l)carbamoyi)-253:i4,5,7,9,13,13a-octahydro-2;!5- methaxϊo >τido['1 2ί:4,5]p>τazmo[2,1.- ][l,3]oxaze in-8-olate present in the composition is oils of the polymorphic forms di sclosed herein. I n certain embodiments, the composition includes at least, about 50%. at least about 60%, at least about 70%, at least about 80%, at least about 85%. at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% of one of the polymorphic forms potassium (2 ,5S, 13aR)-7,9~dioxG- 10-((2
2s5-snethanop ido[l',2':4,S]pyrazirio[2,l"b][l,3]oxazepin-8-olate,
[0182] In other e bodiments of compositions comprising a polymorphic form disclosed herein, less than about 50%. less than about 40%, less than about 30%5 less than about 20%, less than about 10%, less than about 5%. less than about 4%, less than about 3%, less than about 2% or less than about 1% of potassium (2R55S,13aR)-7,9-dioxo-10-((2!4,6- trifiuorobe3^1)carhaBioyi)-2,3,4?5,7s9,13,13a--octahydro-255- r ethanop fido[ l 2':4>5]p razino[2J 'b][l>3]oxazepin-8-o!ate present in the composition are other polymorphs of potassium (2R,5S,13aR)-7.9-dioxo-10~((2s4,6~
trifiuorobesr^l)carbamo l)~2,3,4s5,7,9si3,.13a~oetahydro-2,5- methanopyrido r.2!:4,5]pyrazhio[2,l~b][l!3]oxa2ep}rj~8-oSate and/or impurities.
[0183] in yet other embodiments of compositions comprising the polymorphic forms disclosed herein, impurities make up less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the total mass relative to the mass of the polymorphic forms present, impurities may, for example, include by-products from synthesizing potassium (2R.5SJ 3aR)~7,9~diGXQ-10-((2,4s6 riflu^
2,3A53759,13J3a"OCtahy ro~2s5-methanopyrido[^
olate, contaminants, degradation products, other polymorphic forms, water, and sol vents. In certain embodiments, impurities include by-products from the process of synthesizing potassium (2R55 S, 13 aR)-7,9~dioxo- 10-((2,4,6»trifluorobenzyl)carbamoyi)-253 ,4, 5 ,7,9, 13,13a- octah}'dro-2s5-metha.nopyrido[r,2i;4J5]pyTazino[2,l-b][l s3]oxazepk-8-oiate. In certain embodiments, impurities include contaminants from the process of synthesizing potassium (2R,5S, 13aR)-7,9-dioxo- 10-((2,4s6-trifluofoben2yl)earbamoyl)-253,4,557,9, 13, 13a~octahydro~ 2,5"S¾ethanopyrido[r,2,:4,5]pyrazino[2, i~b][ 1 ,3]oxazeph §-olate. In certain embodiments, impurities include degradation products of potassium (2R.5S, 13aR)«?,9~dioxo - 10-{(2,4,6- U-ifluorobeRzy!)carbamoy])-2,3,4,S,7,9> 13, 13a~oeiahydro-2,5~
et hanopyridoi 1 ',2':4,5]pyra?ino[2, 1 -h] [ 1 , )oxazepin-8~olate. In certain embodiments, impurities include other polymorphic forms of potassium (2R«5S, i 3a )~7,9-dioxo~ I0~({2,4,6- trifl uorobenzyl) arbamoyl)~2, 3,4,5,7,9,13,13 a-octahydro-2,5 - methaiK)p rido ,2':4.S]p azmo[2,1 ~b][l ,3]oxazepin-8-oiate. In certain embodiments, Impurities include water or solve t, In certain embodiments of compositions comprising a polymorphic form disclosed herein, impurities are selected from, the group consisting of byproducts from synthesizing potassium (2 ,5S,13aR)~7,9-"dioxo~10~{{2,4,6~
friflnoroberffiy i)carbamoyI)~2,3,4,5s7,9, 13, 13a~octahydro-2,S- methanopyrido[r,2!:4,5]pyraziiio[25i-b][l,3]oxazepin-8-o1ate, contaminants, degradation products, other polymorphic forms, water, solvents and combinations thereof.
[0184] In yet other embodiments, the composition comprising a polymorphic form disclosed herein has less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% by weight of amorphous or non-crystalline potassium (2R,5S.13aR)~7,9-dioxo- 10-((2,4,6-trifi orobeii¾'l)carbamoyl)-2,3,4,5,7s9! 13,13a-octahydro- 2J5»methanopyrido[lV2!:4,5]py a7ino[2,l "b][l,3]oxazepin~8--olaie.
[0185 ] Provided are also compositions comprising at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or ail of polymorphs {e.g., any one or more of Formula I polymorphic Forms I, II, ill, IV, V, VI, VI , and VIII, Formula II polymorphic Form I, and/or Formula IH polymorphic Forms 1, 11, aud III) as described herein. In a particular embodiment a composition comprising one of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form 1, and/or Formula III polymorphic Forms 1, 11, and III described herein is provided. In a particular embodiment, a composition comprising two of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form Ϊ, and/or Formula III polymorphic Forms I, II, and III described herein is provided, In a particular embodiment, a composition comprising three of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form 1, and/or Formula II polymorphic Forms I, II, and III described herein is provided, In a particular embodiment, a composition comprising four of Formal Formula 1 polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms L II, and ΠΪ described herein is provided. In a particular embodiment, a composition comprising five of Formula I polymorphic Forms I, II, IIL IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided. In a particular embodiment, a composition comprising six of Formula I polymorphic Forms I, ΪΪ, III, IV, V, VI, VII, and VEIL Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided. In a particular embodiment, a composition comprising seven of Formula I polymorphic Forms I, ϊϊ, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and HI described herein is provided. In a particular embodiment, a composition comprising eight of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic forms I, II, and III described herein is provided, In a particular embodiment a composition comprising nine of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form Is and/or Formula 111 polymorphic Forms I, II, and III described herein is provided. In a particular embodiment, a composition comprising ten of Formula I polymorphic Forms L Π, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and HI described herein is provided. In a particular embodiment, a composition comprising eleven of Formula I polymorphic Forms I, II, III, IV, V, VI, VII, and VIII, Formula II polymorphic Form I, and/or Formula III polymorphic Forms I, II, and III described herein is provided. In other embodiments, the compositions described herein may comprise substantially pure polymorphic forms, or may be substantially free of other polymorphs and/or impurities.
[01 S6] In some embodiments, the tem "substantially pure" or "substantially free" with respect to a particular polymorphic form of a compound means that the composition comprising the polymorphic form contains less than 95%, less than 90%, less than 80%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50%, less than 40%, less than 30%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% by weight of other substances, including other polymorphic forms and/or impurities. In certain embodiments, "substantially pure" or "substantially free of 5 refers to a substance free of other substances, including other polymorphic forms and/or impurities. Impurities may, for example, include by-products or left over reagents from chemical reactions, contaminants, degradation products, other polymorphic forms, water, and solvents.
[0187] Administration of the compounds of the invention in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as solid dispersions and solid solutions. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. In one embodiment, the pharmaceutical compositions is prepared for oral administration. In a specific embodiment, the pharmaceutical composition is a tablet. Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the compositio to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparen t, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000), The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention for treatment of a disease or condition of Interest in accordance with the teachings of this invention,
[0188] The pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution, A surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
Surfactants are compounds that non-covalentiy interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous de lively system.
[0189] For example, a solid pharmaceutical composition intended for oral administration can be prepared by mixing a compound of the invention with at least one suitable pharmaceutical excipient to form a solid eformulation composition, which then may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. Accordingly in one embodiment a pharmaceutical composition is provided, which includes a compound of Formula (Ϊ), (II), or (III) and a pharmaceutical exe-ipient.
[01 0] The compounds of the invention are administered in a therapeutically effecti ve amount which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy, in some embodiments, the compounds of the in ention can be administered alone or is combination with other antiviral agents once or twice daily for as long as the patient is infected, latently infected, or to prevent infection (e.g. for multiple years, months, weeks, or days).
Combination Therapy
[0191] in one embodiment, a method for treating or preventing an HIV infection in a human having or at risk of having the infection is provided, comprising administering to the human a therapeutically effective amount of a compound disclosed herein in combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents. In one embodiment, a method for treating an HIV infection in a human having or at risk of having the infection is provided, comprising administering to the human a therapeutically effective amount of a compound disclosed herein in combination with a therapeutically effective amount of one or more (e.g., one. two, three, one or two, or one to three) additional therapeutic agents.
[01 2] In one embodiment, a method for treating an HIV infection in a human having or at risk of having the infection is provided, comprising administering to the human a therapeutically effective amount of a compound or composition disclosed herei In combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents.
[0193] In certain embodiments, the present invention provides a method for treating an HI infection, comprising administering to a patient in need thereof a therapeutically effective amount of a compound or composition disclosed herein in combination with a therapeutically effective amount of one or more additional therapeutic agents which are suitable for treating an HIV infection.
[0194] One embodiment provides a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents for use in a method for treating or preventing an HIV infection in a human having or at risk of having the Infection, One embodiment provides a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents for use in a method for treating an HIV Infection in a human having or at risk of having the infection. One embodiment provides a compound disclosed herein for us in a method for treating or preventing a HIV infection in a human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents. One embodiment provides a compound disclosed herein for use in a method for treating an HIV infection in a human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents. In certain embodiments, the present invention provides a compound disclosed herein in combination with one or more additional therapeutic agents which are suitable for treating an HIV infection, for nse in a method for treating an HIV infection, in certain embodiments, the present invention provides a compound disclosed herein for use in a method for treating an HIV infection, wherein the compound is administered in combinatio with one or more additional therapeutic agents which are suitable for treating an HIV infection.
[0195] One embodiment provides the use of a compound disclosed herein thereof, in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents in the manufacture of a medicament for treating or preventing an HIV infection in a human having or at risk of having the Infection. One embodiment provides the use of a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents in the manufacture of a medicament for treating an HIV infection in a human having or at risk of having the infection. One embodiment provides the use of a compound disclosed herein in the manufacture of a medicament for treating or preventing an HIV infection in a human having or at risk of having the Infection, wherein the compound Is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents. One embodiment provides the use of a compound disclosed herein thereof, in the manufacture of a medicament for treating an HIV infection in human having or at risk of having the infection, wherein the compound is administered in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents, in certain embodiments, the present invention provides the use of a compound disclosed herein thereof, in
combination with one or more additional therapeutic agents which are suitable for treating an HiV infection, in treating an HIV infection, in certain embodiments, the present invention provides the use of a compound disclosed herein thereof for treating an HIV infection, wherein the compound is administered in combination with one or more additional therapeutic agents which are suitable for treating an HIV infection.
[0196] A compound as disclosed herein (e.g., my compound of Formulas (I), (II), and/or (ill)) may be combined with one or more additional therapeutic agents in any dosage amount of the compound of Formulas (ϊ), (II), and/or (III) (e.g., from 50 mg to 1000 mg of compound),
[01 7] In one embodiment, pharmaceutical compositions comprising a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents, and a pharmaceutical ly acceptable carrier, diluent or exeipient are provided.
[0198] In one embodiment, combination pharmaceutical agents comprising a compound disclosed herein in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents are provided.
[01 9] In one embodiment, kits comprising a compound disclosed herein in combination with one or more (e.g., one. two, three, one or two, or one to three) additional therapeutic agents are provided.
[0200] In the above embodiments, the additional therapeutic agent may be an aod-HIV agent. For example, in some embodiments, the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nndeoside inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV mtegrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gp!20 inhibitors, G6PD and NA.DH-oxida.se inhibitors, compounds that target the HIV capsid ("capsid inhibitors"; e.g., capsid polymerization inhibitors or capsid disrupting compounds such as those disclosed Irs WO 2013/006738 (Giiead Sciences), US 2013/0165489 (University of Pennsylvania), and WO 2013/006792 (Pharma Resources), pharmacokinetic enhancers, and other drags for treating HIV, and combinations thereof.
[0201] In other embodiments, the additional therapeutic agent may be an and- HIV agent. For example, In some embodiments, the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nuc!eoside or non-nucleoiide inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or aliosteric) integrase inhibitors, HIV entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gp!20 inhibitors, G6PD and NADH-oxidase Inhibitors, HIV vaccines, HIV maturation inhibitors, latency reversing agents (e.g., histone deacety!ase inhibitors, proteasoroe Inhibitors, protein kinase C (PKC) activators, and BRD4 inhibitors), compounds that target the HIV capsid ("capsid inhibitors"; e.g.. capsid polymerization inhibitors or capsid disrupting compounds, HIV nucleocapsid p7 (NCp?) inhibitors, HIV p24 capsid protein inhibitors), pharmacokinetic enhancers, immune-based therapies (e.g., Pd-1 modulators, Pd-Ll modulators, toil like receptors modulators,, IL-15 agonists, ), HIV antibodies, bispeeific antibodies and "antibody-like" therapeutic proteins (e.g., DARTs®, Duobodies®, Bites®, XmAbs®, TandAbs ®, Fab derivatives) including those targeting HIV gp!20 or gp4i, combination drags for HIV, HIV pi 7 matrix protein inhibitors, IL-13 antagonists, Peptidyl -prolyl cis-trans isoraerase A modulators. Protein, disulfide isomera.se inhibitors, Complement C5a receptor antagonists, DNA methyitransferase inhibitor, HIV vif gene modulators, Vif dhiierization antagonists, HW-1 viral infcetivity factor inhibitors, TAT protein inhibitors, HIV-1 Nef modulators, Hck tyrosine kinase modulators, mixed lineage kinase-3 (MLK-3) inhibitors, HIV-l splicing inhibitors, Rev protein Inhibitors, Integrin antagonists, Nucleoprotein inhibitors, Splicing factor modulators, COMM domain containing protein 1 modulators, HIV ibonuclease H inhibitors, Retrocyelin modulators, CDK-9 inhibitors, Dendritic ICAM-3 grabbing nonintegrln 1 inhibitors, HIV GAG protein inhibitors, HIV POL protein inhibitors, Complement Factor H modulators, Ubiquitin ligase inhibitors, Deoxycytidme kinase inhibitors, Cyelin dependent kinase inhibitors Proprotein corsvertase PC9 stimulators, ATP dependent RNA helicase DDX3X inhibitors, reverse transcriptase priming complex inhibitors, HIV gene therapy, PI3 inhibitors, compounds such as those disclosed In WO 201.3/006738 (Gilead Sciences)., US 2013/0165489 (University of
Pennsylvania), WO 2013/091096A1 (Boehringer Ingelheim), WO 2009/062285 (Boehringer Ingelheim), US20140221380 (Japan Tobacco), US2014022I378 (Japan Tobacco), WO 2010/130034 (Boehringer Ingdheim), WO 2013/159064 (Gilead Sciences), WO
2012/145728 (Gilead Sciences), WO2012/003497 (Gilead Sciences), WO2014/ 100323 (Gilead Sciences), WO2012/145728 (Gilead Sciences), WO2013/159064 (Gilead Sciences) and WO 2012/003498 (Gilead Sciences) and WO 2013/006792 (Pharma Resources), and other drugs for treating HIV, and combinations thereof.
[0202] In certain embodiments, the additional therapeutic is selected from tbe group consisting of HIV protease inhibitors, H1Y non-nucleoside or non-nucleotide inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or allosterie) integrase Inhibitors,
pharmacokinetic enhancers, and combinations thereof.
[0203] in certain embodiments a compound of Formulas (I), (II), and/or (III) is formulated as a tablet, which may optionally contain one or more other compounds useful for treating HIV. In certain embodiments, the tablet can contain another active ingredient for treating HIV, such as HIV protease inhibitors, HIV non-nucleoside or non-nucleotide Inhibitors of reverse transcriptase, HIV nucleoside or nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, HIV non-catalytic site (or allosterie) integrase inhibitors, pharmacokinetic enhancers, and combinations thereof. In certain embodiments, the tablet can contain one or more active ingredients for treating HIV, such as HIV nucleoside or nucleotide Inhibitors of reverse transcriptase. In certain embodiments, such tablets are suitable for once daily dosing,
[0204] In further embodiments, the additional therapeutic agent is selected from one or more of:
(1) HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavlr, indinavir, lopinavir, ritonavir, nelfmavir, saquinavir, tipranavir, brecanavir, darimavir, TMC-126, TMC-114, mo enavir (DMP~4S0)5 JE-2147 (AG1776), =,·· 756423, RO0334649, ΝΪ-272, DPC-681 , DPC-684, GW640385X, DG17, PPL-100, DG35, and AG 1859; (2) HIV ηοπ-nuckoside or non-nucleotide inhibitors of reverse transcriptase selected from the group consisting of capravirine, emivsrsne, defaviridine, efevirenz, nevirapine, (÷) ca!anoiide As etravirfee, GW5634, DPC-083, DPC-961, DPC-963, ΜΓΛ 50, TMC-12Q, rilpivirinc, BILR 355 BS, VRX 840773, lersivirme (UK-453061), RDEA8G6, KM023 and MK-1439;
(3) HIV nucleoside or nucleotide inhibitors of reverse transcriptase selected from the group consisting of zidovudine, emfridtahine, didanosine, stavudins, xale-iiabine, lamivudine, abacavir, abacavir sulfate, aradoxovir, elvucit&bine, alovudine, MIV-210, ±-FTC, D~d4FC, emtricitabine, phosphatide, fozivudine tidoxil, aprieitlbine (AVX754), KIM 461, GS-9131
(Gilead Sciences), fosalvudine tidoxil (formerly HDP 99,0003), tenofovir, tenofovir dlsoproxil fUmarate, tenofovir alafenamide, tenofovir alafenamide hemifumarate, tenofovir alafenamide fumarate (Gilead Sciences), GS-7340 (Gilead Sciences), GS- 148 (Gilead Sciences), adefovir, adefovir dlpivoxll, CMX-001 (C merix) and CMX- 157 (Chimerix);
(4) HIV integrase inhibitors selected from the group consisting of ciircumin, derivatives ofcurcumin, chicoric acid, derivatives of chicoric acid, 3,5-dicafFeoylquinic acid, derivatives of 3,5-dicaffeoyiquInie acid, aurintriearboxyfie acid, derivatives of
aarintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeie acid phenethyi ester, iyrphostin, derivatives of iyrphostin, quercctin, derivatives of quercetin, S-1360, AR- 177, L-870812, and L-870810, raitegra , BMS-538158, GSK364735C, BMS-707035, MK- 2048, BA 01 1, elvitegravir, dolutegravir, dolutegravir sodium, and GSK-744;
(6) HIV fion-catalyiic .site, or allosterk, integrase inhibitors (NCTNI) including, but. not limited to, BI-224436, CXG516, CX05045, CX14442, compounds disclosed in WO
2009/062285 (Boehringer Ingelheim), WO 2 10/130034 (Boehringer Ingelheim), WO
2013/159064 (Gilead Sciences), WO 2012/145728 (Gilead Sciences), WO 2012/003497 (Gilead Sciences), WO 2012/003498 (Gilead Sciences) each of which is incorporated by references in its entirety herein;
(7) gp4i inhibitors selected from the group consisting of enfuvirtide, sifuvtrtide, a!buvirtlde, FB006M, and TRM 144;
(8) the CXCR.4 inhibitor AMD-070;
(9) the entry Inhibitor SP01A; (10) the gp!20 inhibitor BMS-488043;
(11) the G6PD and NADH-oxidase inhibitor immuaitin;
(12) CCRS Inhibitors selected from the group consisting of aplaviroc, vicriviroc, maraviroc, ccnicriviroc, PRG-I 40, INCB.I 5050, PF-232798 (Pfizer), and CCR5mAb004;
(13) CD4 attachment inhibitors selected from the group consisting of ibalizumab (TMB-355) and BMS-068 (BMS-663068);
(14) phamiacokmetic enhancers selected from the group consisting of cobicistai and SPI-452; and
(15) other drugs for treating HIV selected from the group consisting of BAS-I00, SPI- 452, REP 9, SP-Gi A, TNX-3SS, DES6, ODN-93, ODN-112, VGV-1, PA-457 (bevMmat), HRG214, VGX-410, D-247, AMZ 0026, CYT 99007A-221 HIV, DEBiO-025, BAY 50- 4798, MDX010 (ipilimumab), PBS 119, ALG 889, and PA-1G5QQ40 (PA-040), and combinations thereof.
[0205] In certain embodiments, die additional therapeutic agent is selected from one or more of:
(I) Combination drugs selected from the group consisting of ATRIPLA®
(efavirenz+tenofovir disoproxil fumarate +emiricitabine), COMPLERA® or EYIPLERA® (rilpivlrffie-rterjofovir disoproxil fumarate - emiricitabine), STRIBILD®
(eh'itegfavir-fcobieistat+tenofoyir disoproxil fumarate Emtricitabine), do!utegravir ÷ abacavir sulfate -fdamiv dine, TRILMEQ® (dolutegravir + abaeavir + lamiivudine) , iamivudine + nevirapine + zidovudine, dointegravir+ri!pivirhie, dolntegravirfriipivirme hydrochloride, atazanavir sulfate + cobicistai, atazanavir + cobicistai, darimavir + cobicistai, etavirenz + Iamivudine + tenofovir disoproxil fumarate, tenofovir alafenamide hemifnmarate ■÷· emtricitabine ·÷ cobicistai + elvitegravir, tenofovir alafenamide hemifumarate + emtricitabine, tenofovir aiaienarnide + emtricitabine, tenofovir alafenamide hemifismarate + emtricitabine + rifpivtrisie, tenofovir alafenamide + emtricitabine + ri!pivirhie , Vacc-4x + romidepsiri, darunavir tenofovir alafenamide hemifumarate^ emtricitabine + cobicistai APH-0812, raltegravir + iamivudine, ALETRA® (ALUVIA®, lopinavir+ritcaiavir), atazanavir sulfate + ritonavir, COMB!YIR® (sdovudine+!amivudtee, AZT+3TC), EPZICOM® (Kivexa®, abacavir sulfate +lamivudme, ABC+3TC), TRIZIVIR® (abacavir sulfate+zidovudme+lamivudme, ABC+AZI+3TC), TRUVA.DA® (tenofovir disoproxil fumarate - emincitabme, TDF+FTC), doravirine + lamivudine + ienofovir disoproxil fumarate, doravirine + lamivudine + ienofovir disoproxil, tenofovir + lamivudine and lamivudine + tenofovir disoproxil fumarate;
(2) HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavir, fesamprenavir calcium, indinavir, indinavir sulfate, lopinavir, ritonavir, nelflnavir, nelfinavir mesylate, saquinavir, saquinavir mesylate, tipranavir, breeanavir, darunavir, DG-17, TMB-657 (FPL-100) and T C-31091 1 ;
(3) HIV non-nuc!eoside or non-nncleotide inhibitors of reverse transcriptase selected from the group consisting of delavirdine, delavirdine mesylate, nevirapine, etravirine, dapivirine, doravirine, rilpivirine, efavirenz, M-G23, VM-1500, lentinan and AIC-292;
(4) HIV nucleoside or nucleotide inhibitors of reverse transcriptase selected from the group consisting of VIDEX® and VIDEX® EC (didanasine. ddl)5 zido vudine, emtricitabine, dldanosine, stavudine, zalcitabine, lamivudine, censavudine, abacavir, abacavir sulfate, amdoxovir, el ucitabine, alovudine, phosphazid, fozivudine tidoxil, aprkitabine, amdoxovir . KP-1461, fosa!vudine tidoxil, ienofovir, tenofovir disoproxil, ienofovir disoproxil fumarate, tenofovir disoproxil hemifi.mia.rate, tenofovir alafenamide, tenofovir alafenamide
hemifumarate, tenofovir alafenamide fumarate, adefovir, adefovir dipivoxil, and festinavir;
(5) HIV integrase inhibitors selected from the group consisting of ewcwniin, derivatives of curcu in, chicoric acid, derivatives of chicoric acid, 3 ,5 -dicaffeoylquin ic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxyiic acid, derivatives of
aurintricarboxyiic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of lyrphostin, qoercetin, derivatives of quercetin, raltegravir, dvitegravir, do!utegravir and eabotegravir;
(6) HIV non-catalytic site, or allosterie, integrase inhibitors (NCINI) selected from the group consisting of CX-05168, CX-05045 and CX-14442;
(?) HIV gp4.l inhibitors selected from the group consisting of enfovixtide, sifuvirtide and albuvirtide; (8) HIV entry inhibitors selected from the group consisting of cenicriviroc;
(9) HIV g i O inhibitors selected from the group consisting of Radha-108 (Reeeptol) and BMS-663068;
(10) CCR.5 inhibitors selected from the group consisting of apiaviroc, vicriviroc, rnaraviroe, cemcriviroc, PRO- 140, Adaptavir (RAP-J O 1 ), nifeviroc (TD-0232), TD-0680, and vMIP (Haimipu);
(11) CD4 attachment inhibitors selected from the group consisting of ibalkuniab;
(12) CXCR4 inhibitors selected from the group consisting of plerixafor, ALT- 1188, vMIP and Hairnipu;
(13) Pharmacokinetic enhancers selected from the group consisting of cobicistat and ritonavir;
(14) Immune-based therapies selected from the group consisting of dermaVir, interkukin-7, plaqnenll (hydroxychloroquine), proleukin (aldesleukin, if . -2), interferon alia, interferon alfa-2b, interferon alfa-n3, pegylated interferon alfa, interferon gamma, hydroxyurea, myeoplienoiate mofetil (MP A) and its ester derivative mycoptienolate mofetil (MMF), WF-10, ribavirin, l'L-2, IL-12, polymer polyeffayleneimine (PEI), Gepon, VGV-1, MOR-22, BMS-936S59, toll-like receptors modulators (tkl, ilr2, tlr3, tir4, tlr5, tlr6, tlr7, tlrS, tir9, tlrlO, tir1 1 , tirl2 and tlr.13), rintatolimod and IR-103;
(15) HIV vaccines selected from the group consisting of peptide vaccines, recombinant subunit protein vaccines, live vector vaccines, DNA vaccines, virus-like particle vaccines (pseudovirion vaccine), CD4-derived peptide vaccines, vaccine combinations, rgpl2Q (AIDSVAX), ALVAC HIV (vCP! S2i)/AIBSVAX B/E (gp!20) (RV144), monomelic gp l 20 IHV-1 subtype C vaccine (Novartis), Remnne, ITV-l . Contre Vir, Ad5~ENVA-48, DCVax- 001 (CDX-24G1), PEP-6409,Vacc-4x. Vacc~C5, VAC-3S, muiticlade DNA recombinant adenovirus-5 (rAdS), Pennvax-G, V C-HIV MAB060-00-AB, AVX-101. Tat Oyi vaccine, AVX-201, HIV-LAMP-vax, Ad355 Ad35-GR1N, NAcGM3/VSSP ISA-51, poIy-ICLC adjuvanted vaccines, Tatlmmune, GTU-muMHIY (FIT-06), AGS-004, gpl40[deita]V2.TVl+ MF-59, rVSVIN HIV-1 gag vaccine, SeV-Gag vaccine, AT-20, DNK-4, Ad35-GPJN/ENV, TBC-M4, HIV AX , HIVAX-2, NYVAC-HIV-PTl , NY V AC-Ηί V-FN, D Α-ΗΪ V-PT 123, rAAV l -PG9DP, GOVX-B i 1 , GOVX-B21 , ThV-GL ΤϋΠ-16, VGX-3300. TVI-HIV-1, Ad-4 (Ad4-env Clade C + Ad4-mGag)s EN41-UGR7C, EM4I-FPA2, PrcVaxTat, TL-01, SAV- 001 , AE-H, MYM-YIGl, CombiHIVvac, ADVAX, MYM-V201, MVA-CMDR, ETV-01, CDX-I4G1, rcAd26.MOS l .HiV-Env and DNA~Ad5 gag/pof/nef/nev (HYTN505);
(16) HIV antibodies, bispecific antibodies and "antibody-ilke" therapeutic proteins (such as DARTs®, Duobodies®, Bites®, XmAbs®, TandAbs ®„ Fab derivatives) including BMS-936559, TMB-360 and those targeting HIV gp!20 or gp41 selected from the group consisting of bavituximab, UB-42L C2F5, C2G12, C4E10, C2F5+C2G12+C4E10, 3-BNC- 1 17 , PGT145, PGT12L DX01 Q (ipilim ma ). VRCOl, A32, 7B2, 1GE8, VRC-07-523 and VRC07;
(17) latency reversing agents selected from the group consisting of Histone deaeetylase inhibitor such as Romidepsin, vorinostai panobinostat; Proteasome inhibitors such as Yeleade; protein kinase C (PKC) activators such as indolaetam, Prostratin, Ingenol B and DAG-lactones, Ionomycin, GS -343, PMA, SAHA, BRD4 inhibitors, IL-15, JQI , disulfram, and amphotericin B;
(18) HIV nucleocapsid p7 (NCp7) inhibitors selected from the group cons sting of azod carbonamide
(19) HIV maturation inhibitors selected from the group consisting of BMS-955176 and GSK-2838232;
(20) PI3K inhibitors selected from the group consisting of ideialisib, AZD-8186, bupariisib, CLR-457, pictilisib, neratinib, rigosertib, rigosertib sodium, EN-3342, TGR-1202, aipelisib, duvelisib, UCB-5857, tase!isib, XL-765, gedatoiisib, VS-5584, copanlisib, CA? orotaie, perifosine, RG-7666, GSK-2636771 , DS-7423, panulisib, GS -2269557S GSK- 2126458, CUDC-907, PQR-309. iNCB-040093, pilaralisib, BAY- 1082439, puquitinib mesylate, SAR-245409, AMG-319, RP-6530, ZST -474, MLN-1117, SF-1126, RV-1729, sonolisib, LY-30234 4, SAR-260301 and CLR-1401 ;
(21) the compounds disclosed in WO 2004/096286 (Gilead Sciences), WO
2006/1 10157 (Gilead Sciences), WO 2006/015261 (Gilead Sciences), WO 2013/006738 (Gilead Sciences), US 2013/0165489 (University of Pennsylvania), US20140221380 (Japan Tobacco), US20140221378 (Japan Tobacco), WO 2013/006792 (Pharma Resources), WO 2009/062285 (Boehringer Ingelheim), WO 2010/130034 (Boehringer Ingelheim), WO 2013/091096A1 (Boehringer Ingelheim), WO 2013/159064 (Gilead Sciences), WO
2012/145728 (Gilead Sciences), WO2012/003497 (Gilead Sciences), WO2014/100323
(Giiead Sciences), WO2012/1 5728 (Gilead Scie ces), WO2013/159064 (Gilead Sciences) and WO 2012/003498 (Gilead Sciences); and
(22) other drugs for treating HIV selected from the group consisting of BanLee, MK~ 8507, AG- 1105, TR-452, M -8591 , REP 9, CYT-107, alisporivir, NOV-205, IND-02, metenkefalin, PGN-007, Aeemannan, Gamimune, Prolastin, 1,5-dicaiFeoylquinic acid, ΒΓΓ- 225, RPI-MN, VSSP. HI viral, IMC TIOO, SB-728-T, RPI-MN, V1R-576, HGTV-43, MK- 1376, rfflV7-shl-TAR-CCR5RZ, MazF gene therapy, BlockAk!e, ABX-464, SCY-635, naltrexone, AAV-eCD4-Ig gene therapy and PA- 1050040 (PA-040); and combinations thereof*
[0206] In certain embodiments, a compound disclosed herein Is combined with two. three, four or more additional therapeutic agents, in certain embodiments, a compound disclosed herein Is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein Is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein is combined with four additional therapeutic agents, The two, three four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, or they can be selected from different classes of therapeutic agents, in a specific embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV non~nucleoside inhibitor of reverse transcriptase, in another specific
embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound. In a further embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucieoside inhibitor of reverse transcriptase, and an HIV protease inhibiting compound. In an additional embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HTV non-nucleoside inhibitor of reverse transcriptase, and a
pharmacokinetic enhancer. In another embodiment, a compound disclosed herein is combined with two HIV nucleoside or nucleotide inhibitors of reverse transcriptase. [0207] In certain embodiments, a compound disclosed herein, is combined with one, two, three, four or more additional therapeutic agents. In certain embodiments, a compound dssciosed herein is combined with one additional therapeutic agent, In certain embodiments, a compound disclosed herein is combined with two additional therapeutic agents. In other embodiments, a. compound disclosed herein is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein is combined with four additional therapeutic agents. The one, two, three, four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, and/or they can be selected from d ifferent, classes of therapeutic agents. In a specific embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase, in another specific embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, and an HIV protease inhibiting compound, in a further embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, ami an HIV protease inhibiting compound. In an additional embodiment, a compound disclosed herein is combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer. In certain embodiments, a compound disclosed herein is combined with at least one HIV nucleoside inhibitor of reverse transcriptase, an integrase inhibitor, and a pharmacokinetic enhancer, In another embodiment, a compound disclosed herein is combined with two HIV nucleoside or nucleotide inhibitors of reverse transcriptase.
[0208] In certain embodiments, a compound disclosed herein is combined with at least one HIV nucleoside inhibitor of reverse transcriptase, an integrase inhibitor, and a pharmacokinetic enhancer.
[0209] In a particular embodiment, a compound disclosed herein is combined with abacavir, abacavir sulfate, tenofovir, tenofovir disoproxlf &marates tenofovir alafenamide, or tenofovir alafenamide hemifumarate,
[021.0] In a particular embodiment, a compound disclosed herein is combined with tenofovir, tenofovir disoproxil fimnarate, tenofovir alafenamide, or tenofovir alafenamide hemifumarate, [0211] In a particular embodiment a compound disclosed herein is combined with a first additional therapeutic agent selected from the group consisting of: abacavir, abacavi sulfate, tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenoibvir alafenamide hemifumaraie and a second additiosal therapeutic agent selected from the group consisting of emtricitibirse and lamivudine.
[0212] Irs a particular embodiment, a compound disclosed herein is combined with a first additional, therapeutic agent selected from the group consisting of: tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenofovir alafenamide hemlfumarate and a second additional therapeutic agent, wherein the second additional therapeutic agent is emtrichibine.
[0213] in a particular embodiment, a compound disclosed herein is combined with one, two, three, four or more additional therapeutic agents selected from Ttiumeq®
(dolutegravir+abacavir +3amivudine), doktegravir ÷ abacavir sulfate ~ lamivudine, raltegravk raltegravir * lamivudine, Truvada® (tenoibvir disoproxil fumarate
+emtricitabine, TDF-f-FTC), maraviroc, enfuvirtide , Epzieom® (Livexa®, abacavir sulfate +lamlvudine, ABC+3TC), Trizivir® (abacavir sulfate+zidovudme+Ianiivudine,
ABC+AZT -3TC), adefovir, adefovir dipivoxil, Stribild ® (eivitegraviH-cobicistat+tenofovir disoproxil fumarate +emtrlcitabine) rilpivirine, rilpivirine hydrochloride, Complera® (Evlpfera®, rilpivirine+tenofovir disoproxil fumarate - umtricitabmie), Cobicistat, atazanavir sulfate + cobicistat, atazanavir - cobicistat, darimavir -s- cobicistat, Atripia®
(efavirenz+tenofovir disoproxil fumarate +emtrieitabine), atazanavir, atazanavir_su!fate, dolutegravir, elvltegravk, Aluvia® (Kaletra®, lopinavir+ritonaw), ritonavir 5 emtrieitabine , atazanavirji lfate - ritonavir, darunavir, lamivudine, Proiasilm fosamprenavir, fosamprenavir calcium, efavirenz, Comblvir® (zidovudme+iamivudine, AZTf3TC), etravirine, nelfioavfc nelfmavir mesylate, interferon, didanosine, stavudme, indinavir, indinavir sulfate, tenofovir ÷ lamivudine, zidovudine, nevirapine, saquinavir, saquinavir mesylate, aldesleukin, zaieitabke, tipranavlr, araprenavir, delavirdine, delavirdlne mesylate, Radha-108 (Receptol), HI viral, lamivudine + tenofovir disoproxil femarate, efavirenz lamivudine tenofovir disoproxil fumarate , phosphazid, lamivudine -f- nevirapine ÷ zidovudine, abacavir, abacavir sulfate, tenofovir, tenofovir disoproxil, tenofovir disoproxil fumarate, darunavir ÷ cobicistat, atazanavir sulfate + cobicistat, atazanavir ÷ cobicistat, tenoibvir alafenamide and tenofovir alafenam Ide hem ifuniarate , [0214] In a particular embodiment, a compound disclosed herein is combined with abacavir, abacavir sulfate, tenofovir, tenofovir disoproxil, tenofovir disoproxil fumarate, tenofovir disoproxil hemlfumaraie, tenofovir aiafenamide or tenofovir aiafenamide hemifismarate,
[0215] la a particular embodiment, a compound disclosed herein is combined with tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide, or tenofovir aiafenamide hemifumarate.
[0216] in a particular embodiment, a compound, disclosed herein is combined with a first additional therapeutic agent selected from the group consisting of: abacavir sulfate, tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide. and tenofovir aiafenamide hemi fumaraie and a second additional therapeutic agent selected from the group consisting of erntrieitabine and laxnivudine.
[0217] In a particular embodiment, a compound disclosed herein Is combined with a -first additional therapeutic agent selected from the group consisting of: tenofovir, tenofovir disoproxil, tenofovir disoproxil fumaraie, tenofovir aiafenamide, and tenofovir aiafenamide hemifnmarate and a second additional therapeutic agent, wherein the second additional therapeutic agent, is emtricitablne.
[0218] In certain embodiments, a compound disclosed herein is combined with 5-30 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemifumarate, or tenofovir aiafenamide and 200 mg emtrlcitablne, in certain embodiments, a compound disclosed herein is combined with 5-10; 5-15; 5-20; 5-25; 25-30; 20-30; 15-30; or 10-30 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemi fumaraie. or tenofovir akfenamide and 200 mg emtricitablne. In certain embodiments, a compound disclosed herein is combined with 10 mg tenofovir aiafenamide fumaratc, tenofovir aiafenamide hemlfumaraie, or tenofovir aiafenamide and 200 mg emtricitablne. in certain embodiments, a compound disclosed herein is combined with 25 mg tenofovir aiafenamide fumaraie, tenofovir aiafenamide hemifumarate, or tenofovir aiafenamide and 200 mg emtricitablne. A compound as disclosed herein (e.g., a compound of Formulas (I), (II), and/or (III)) may be combined with the agents provided herein in any dosage am ount of the compound (e.g., from 50 mg to 500 nig of compound) the same as if each combination of dosages were specifically and individually listed. [0219] In certain embodiments, a compound disclosed herein is combined with 200-400 nig tenofovir disproxIL tenofovir disoproxil fumarate, or tenol vir disoproxii hemifurnarate and 200 mg emtrieitabine. In certain embodiments, a compound disclosed herein is combined with 200-250; 200-300; 200-350; 250-350; 250-400; 350-400; 300-400; or 250» 400 mg tenofovir disoproxii, tenofovir disoproxii fumarate, or tenofovir disoproxil hemifurnarate and 200 nig emtrieitabine, In certain embodiments, a compound disclosed herein is combined with 300 mg tenofovir disoproxii fumarate, tenofovir disoproxii hemifumarate, or tenofovir disoproxii and 200 mg emtrlcitahlne. A compound as disclosed herein (e.g., a compound of Formulas (I), (11), and/or (III)) may be combined with the agents provided herein m any dosage amount of the compound (e.g., from 50 mg to 500 mg of compound) the same as if each combination of dosages were specifically and individually listed.
[0220] In certain embodiments, when a compound disclosed herein is combined with one or more additional therapeutic agents as described above, the components of the composition are administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
[0221] In certain embodiments, a compound disclosed herein is combined with one or more additional therapeutic agents in a unitary dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration.
[0222] In certain embodiments, a compound disclosed herein is administered with one or more additional therapeutic agents. Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of the compound disclosed herein and one or more additional therapeutic agents are both present in the body of the patient.
[0223] Co-administration includes administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents, For example, in some embodiments, a unit dose of a compound disclosed herein is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents. Alternatively, In other embodiments, a unit, dose of one or more additional therapeutic agents is administered first, followed fay administration of a unit dose of a compound disclosed herein within seconds or minutes. In some embodiments, a unit dose of a compound disclosed herein is administered first, followed, after a period of hours {e.g.. I -12 hours), by administration of a unit dose of one or more additional therapeutic agents, In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound disclosed herein.
XRPD Data
[0224] In certain embodiments, the crystalline forms are characterized by the interiattlce plane intervals determined by an X-ray powder diffraction pattern (XRPD). The
diffractogram of XR PD is typically represented by a diagram plotting the intensity of the peaks versus the location of the peaks, i.e., diffraction angle 20 (two-theta) in degrees, The intensities are often given in parenthesis with the following abbreviations; very strong - vst; strong - st; medium - m; weak ~ w; and very weak - vw. The characteristic peaks of a given XRPD can be selected according to the peak locations and their relative intensity to conveniently distinguish this crystalline structure from others.
[0225] Those skilled in the art recognize that the measurements of the XRPD peak locations and/or intensity for a given crystalline form of the same compound will vary within a margin of error. The values of degree 2Θ allow appropriate error margins. Typically, tha error margins are represented by For example, the degree 20 of about "8.7±G.3" denotes a range from about 8,7- 0.3» i.e., about 9.0, to abont 8.7-0.3, i.e., about 8.4, Depending on the sample preparation techniques, the calibration techniques applied to the instruments, human operational variation, and etc, those skilled in the art recognize that the appropriate error of margins for a XRPD can be ±0.5; ±0.4; ±0.3; ±0.2; ±0.1; ±0.05; or less. In certain embodiments of the invention, the XRPD margin of error is ±0.2,
[0226] Additional details of the methods and equipment used for the XRPD analysis are described in the Examples section.
[0227] The XRPD peaks for the crystalline forms of (2R,SS, 13aR) -8-hydroxy-7;9-dioxo~
N-(2,456 rifiuorobenzyl)~2s3,4s5,7!9,i3s13a-oetahydrO"2,5- msihar:op>T?do[ 2';4>S]p\Tszi^ (Formula I) Form I. is below in Table 1A.
Table 1 A; XRPD peaks for crystalline forms of Formula 1 Form I
Figure imgf000065_0001
[0228] The XRPD peaks for the crystalline form of (2R,5S,13aR)~8-hydroxy-7,9~dioxo~ N~(2A6 riiiuoFobenzyl)-2A
methanop ridofl^'^Sj razino^^^ (Formula Ϊ) Form II s below in Table IB.
Table IB: XRPD peaks for crystalline fos-ms of Formula I Form II
Figure imgf000066_0001
[0229] The XRPD peaks for the crystalline form of {2R}5S;13aR)-8-hydroKy-7?9»dioxo~ N-(2,4s6-irIfluoroben.zyl)~253,4,5,7,9; 13, 13a-octahydn 255~
methanopyrido[1^2':4,5]pyrazmo[2 "¾^ (Formula I) Farm ΙΠ is below in Table ί C,
Table IC: XRPD peaks for crystalline forms of Formula I Form ill
Figure imgf000066_0002
[0230] The XRPD peaks for the crystalline form of (2R,5S,13a )-8-hydroxy-7,9-dioxo-
'N~(2;4,6 rifiuorobenzyi)-2,3s4,5,7,9513,13a»octahyclro~2:i5- msthaEopyrido[l 2,:4,5]p}'Tazino[2,l-b][li3]oxazepine~10-carboxamide (Formula Ϊ) Form IV is below m Table I D.
ID: XRPD peaks for crystalline forms of Formula I Form IV
Figure imgf000067_0001
Figure imgf000067_0002
[0231 ] The XRPD peaks for the crystalline form of sodium (2R,5S, 13aR.)~7,9-dioxo-l 0» ((25456-trifluorobesizyI)earbamoyl)-2,3s4,5s7,9> 13,13a-octahydro~2,5~
meihanopyrido[i^2':4s5]pyrazino[2,l-b][l,3]oxazepin-8~olate (Formula II) is below in Table IE.
Table IE; XRPD peaks for crystalline forms of Formula If Form 1
Figure imgf000068_0001
Figure imgf000068_0002
[0232] The XRPD peaks for the crystalline form of (2R,5S,13aR)-8~hydroxy-7,9"dioxo- N-(2,4,6-trifIuorobenzy!)-2s354}5,7,9J13,13a-ociahy(ko-2J5- methanopyrido[1^2,:4,5]p Tazino[2;l"b][l,3]oxazepme~10~carboxamide oxalic acid co- crystal Form I is below in Table IF,
Table IF; XRPD peaks for crystalline forms of Formula ϊ Oxalic Acid Co-crystal
Form I
Figure imgf000069_0001
Preparation of the Polymorphs
Formula I
[0233] One method of synthesizing Ο.Ά ΐύΙ)^νύί χγ--7 -·άί κο·-Ν-(;6-- tr iiluQroben2yI)-2,3,4,5»7s9, 13,1 Sa-ocia ydro^jS-metfaaaopyridof ^'i^SJpyrazinoP, I - fa] [ 1 ,3] oxazepine- 10-carboxara ide (e.g. a compound of Formula (I)) has been previously described in PCX Publication No. WO2014/100323. This reference is hereby incorporated herein by reference in its entirety, and specifically with respect to the synthesis of
(2RS5S, OaR)-S»hydroxy-7,9~dIoxo» -(2,4;6~triiluorofaenz 'i)-253!4)5>7s9, 3,13a-octahydro- 2,5-methanopyrMo[1^2,:4,5]pyraano[25 l-b][ l,3]oxazepine~10-carboxamide.
[0234] For example, in one aspect, provided is a method of producing a composition comprising one or more polymorphs of {2R,5S,13a )-§~Hydroxy-7>9-dioxo- -{2,4)6~ trifi uof oben¾i)"2 A ^ hjP.SJoxazepinc^O-carboxamide, wherein the method comprises combining a compoimd of Formula (I) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the. compound of Formula (I). In another aspect, provided is another method of producing a composition comprising one or more polymorphs of (2R,5S, i :¾R)-8-iiydroxy.7s9"dioxo~N 2,4>6 rifluorobenzyl).2.;3>45S5?,9a3, !3a- octahyd ro~2, 5-meth anopyrido [ 1 ',2':4,5 ]pyrazino[251 -b][l ,3]oxazepine~ 1 O-carboxamide, wherein the method comprises combining (2R,5S43aR)-8-Hydroxy~7,9~dioxo-N^ iriiiuorobeni^l)-2,354A
b][ls3]oxa2:epme-10-carboxamide with a suitable solvent or a mixture of suitable solvents.
[0235] The choice of a particular solvent or combination of solvents affects the formation favoring one polymorphic form of (2R,5S JSa ^^Iydro ^^-dioxo-N-^Ad-
Figure imgf000070_0001
b][l 53]oxazepme-10-carboxamide over another. Solvents suitable for polymorph formation may include, for example, methanol, ethanoi, water, isopropyl acetate, acetonitrile, tetrahydrofurao, methyl isobutyl ketone, and any mixtures thereof,
[0236] In another aspect, provided is also one or more polymorphs of (2R,SS, 13aR)-8- Hydroxy-7,9~dioxo-N-(2A6- rlfto
methanopyrido[l\2';4,5]p} ajzino[2J-b][1 ]ox&zepine-10~carboxamide produced according to any of the methods described herein.
[0237] It should be understood thai the methods for preparing the polymorphs described herein (including any one or more of polymorphic Forms I to Viil) may yield quantity and quality differences compared to the methods for preparing (2R,SS,13aR)-8-Hydroxy-759~ dioxo-N-(25456"trifluoroben¾4)-23s4,5:,759313,13a-octahYdrQ-2)5- methanopyrido[ 1 °s2!:435]pyrazino[2, 1 -b] [ l 53] ixa/epine-' I G-earboxamide produced on laboratory scale.
Formula I Forms I and II
[0238] In one embodiment, provided is a method of producing a composition comprising polymorphic Form I, Form Π, or a mixture thereof, of (2R,5S, 13aR}-8- Iiydroxy-7,9~dioxo~ - (2A6 rifiuorabenzyl)~2;3,4,5,7,9, 13, ! 3a-octahydro~2,5~
methanopyrido[l',2!:4s5]pyTazino[2,l-b][1 ,3]oxazepke«I0~earboxarnide, wherein the method comprises combining (2R,5S,13aR)~8-Hydroxy-7s9- lioxo-N--(2,4s6-ififluorobenzyl).
2,3 A557,9/D, 13a~octahydro-2.,5^
carboxamide with a solvent to produce a composition comprising polymorphic Form, i, Form II or a mixture thereof, of the (2R,5S, 13aR)-8 iydroxy-7,9^ioxo^
Figure imgf000071_0001
carboxamide, wherein the solvent is isopropyl acetate,
[0239] Provided is a polymorphic Form I, Form II, or a mixture thereof, of (2R35Ss13aR)- 8-Hydroxy-7,9-dioxo-N-(2f4,6"trifluorobet32^ l)-2,3 AS A 13, 13a >ciahydro~2,S~
methanopyrido[l :4, S]pyrazino[2, l-b][l ,3]oxazepine- 1 G-carboxamide produced by combining (2R,5S, 13aR.}~8 i.ydfoxy-7J9-dioxo-N-{2,4!6 ri.tl¾orobenEyI)-2,3,45557,9J 13,13a- octahydro-295-meihanopyr ido[ i ',2';455]pyrazino[25 l-b][ ! JJoxazepme- 10-carboxa ide with a solvent, wherein the solvent is isopropyl acetate.
Formula I, Form III
[0240] in one embodiment, provided is a method of producing a composition comprising polymorphic Form III of (2RJ5S^3aR)-8~Hydroxy~759-dioxo-N-(2>456~ fluoroberizyl)-
Figure imgf000071_0002
carboxamidc, wherein the method comprises combining (2R,5S,I3aR)~8~Hydroxy"7,9-dioxo- N-(2,4,6' rifdiorobeB S'l)-2,354,5,7,9:i 13,I3a-octahydro--2,5- methanopyrido[1^2!:4,5]p)ra ino[2, i-b][l,3]oxazepms~10-carboxaniide with a solvent to produce a composition comprising polymorphic Form HI of the (2R,5Ss13aR)-8-Flydfoxy- 7,9^ioxo- ~(2, ,6 rifluoroben2yi)-2 i ,4,5,7,9, 13, 13a-octahydro~2,5~
metha^opyrido[li,2!;4,5]pyra ino[2Jl-b][l,3]oxazepine-I 0-carboxa5nide, wherein the solvent is methyl isobutyl ketone.
[0241] Provided is a polymorphic Form III of {2R,5Ss13aR)-8»Hydroxy--759-dioxo-- - {2,4,6-t?if=uorob inzyl)~2,3,4,5,7.9, 13, 13a~octahydro-2.S ·
methanop>Tido[ 1^2'>4}5]p Ta¾ino[2,l-b][l,3]oxa2;epine-10 arboxamide produced by combining {2R,5S,13aR)-8 Iydroxy~7^^^
QCtahyd∞~2,5-methanopyrido[1^2!:4?5]pyr^^ with a solvent wherein the solvent is methyl isobutyl ketone. Formula I Forms IV, VII, and VIII
[0242] In one embodiment, provided is a method of producing a composition comprising polymorphic Form IV, Form VII, and Form VIII, or a mixture thereof, of (2R55S,!3aR)-8- Hydroxy-7.9^ioxo-N-(2A6-^^
rr!ethanopyrido[r?2';4»5]pyrazirio[2,l--b][1 ,3]oxazepme-10~carboxamide, wherein the method comprises combining (2R,5S, 13aR)-8-Hydroxy~7,9-dioxo-'N-(2,4,6-^
Figure imgf000072_0001
carboxamide with a solvent to produce a composition comprising polymorphic Form fV, Form VII, and Form VIII, or a mixture thereat; of the (2R)5SJ 3a )-8-Hydroxy»759"dioxo-N-
Figure imgf000072_0002
n)e&aoopyrido[rs2r:4,5)p razino^ wherein the solvent is methanol
[0243 ] Provided is a polymorphic Form IV, Form VII, and Form VIII. or a mixture thereof, of (2R.5S, 13aR)-8-Ilydroxy-7s9-dioxo~NH'2,4!6-trifiuorobesKyl)~2,354!557i9513,13a- ociahydro-2,5-meihaaopyrido[ I ',2!:4,5 ]pyrazino[2, 1 ~b] [ 1. s3]oxazepme- 10 -carboxamide produced by combining (2R,SS>13aR}-8-lIydroxy~?, Hlioxo-N-(2,4,6-L^ifluorobenz>4)- 2,3,4,5.7,9, 13,1 :k¾-octahydro-2,5•n-;eihanopyrido[1 ^2!: ;5 ]pyrazino(2, i -b][i Jjoxazepine-f 0- carboxamide with a solvent, wherein the solvent is methanol.
Formula I, Form V
[0244] In one embodiment, provided is a method of producing a composition comprising polymorphic Form V of (2R,5SS 13aR)-8-Hydroxy-7,9^toxo-N 2A^
2 ,3,4,5 ,7, ,13, 13a-octahydro-2,5~meihanopyndo[l ',2Η5] τ3Ζ3ηο[2, 1 -b][l,3]oxa2epine~1 Q- carboxamlde, wherein the method comprises combining (2R!5S513aR)-8-Hydroxy-7,9-dloxo- N-{2s4i6-tri.fluoroberrzyl)-2,3s4,5,7J9) 13, 13a-octahyd.ro-2,5- methanopyrido[ 1 ',2':4,5]pyrazina[2, < - ][ i ,3 joxazepine- 10-earboxamide with a solvent to produce a composition comprising polymorphic Form V of the (2R55S,13a )-S-Hydroxy~7s9- dloxo~N-(2,456~tritluorobe∞.y!)~253,4J5,7s9,13!!3a--octaliydro~2s5- msthanopyridoj i ',2':4,5]pyrazinof2, 1 -b][ IJjoxazepine-lO-carboxantide, wherein the solvent is water.
[0245] Provided is a polymorphic Form V of (2R,5S,13aR>H-Hydroxy»7,9-dioxo~N~ (2!4,6-iritluorobeiizyl)-253,4s5s7s9, 13, 13a-ociahydro~2s5- rnethanopyrido[ 1 !,2t:455]pyra ieo[2s 1 -b i [ joxazepine- 1 G~earboxami.de produced by combining (2R;5S, 13aR)-S 1ydroxy~759-dioxo-N«(2,4,6 rM^ 13, 13a- cctahydro~25S-mefeanopyrIdG[ri2':4?5]^^^ with a solvent, wherein the solvent is water,
Formula I, Fm~m VI
[0246] In one embodiment, provided is a method of producing a composition comprising polymorphic Form VI of(2R,5S, 13aR 8 :iydroxy-7,9Hiioxo-Ni:2; ^^
2,3A5,75, ! 3J 3a~ocia ydro-2^
carboxamide, wherein the method comprises combining (2R, 5S. I
Figure imgf000073_0001
N-{2,4,6-ti-ifluorobenzyi)~25354,537,9313,I3a~octahydro-2s5-- mct anop}'Tido[r52^'4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide with a solvent to produce a composition comprising polymorphic Form VI of the (2R,5S, 1 aR)-8 -I f ydroxy · 759-dioxo-N-{2,4,6-trifluoroberszyi)-2,354J,7,9, 13, 13a~octahydro~2?5~
metha»opyrido[ r,2!:4,5]pyra2iEo[2, 1 -b] [ 1 ,3]oxazepme- 10-carboxamide, wherein the solvent is selected from the group consisting of methanol, water, and any mixtures thereof, In an embodiment, the solvent is a mixture of water and methanol.
[0247] Provided is a polymorphic Form VI of (2R, S, 13aR)~8~H droxy~7,9~dioxo-N- (2,4,6-trifiuorobenzyl)~2,3 ,4,5,7,9, 13,13 a -octahydro-2,S- methanopyrido[1^2':4s5jpyrazmo[2sl^ by combining
Figure imgf000073_0002
oetahydro«2,5-metfaanopyrido[i ',2!:4,,5 jpyrazino[2, 1 ~h][ 1 ,3 joxazepine- 10-carboxamide with a solvent, wherein the solvent is selected from the group consisting of methanol, water, aad any mixtures thereof, Irs an embodiment, the solvent is a mixture of water and methanol
Formula II
[0248] One method of synthesizing (2R.5S 3aR)-8-hydroxy-7^
triflucfGberiwi)-2,3,4,557,9513,B
b][l ,3joxazepine-10-carboxaniMe (e.g. a compound of Formula (I)) has been previously described in PCX Publication No. WO2014/100323. This reference Is hereby incorporated herein by reference in its entirety, and specifically with respect to the synthesis of (2R,5SJ3aR)-8-hydrox ^
2s5-methanopyrido[ I :4,5]p>Tazmo[2, 1 ~b][ 1 ,3]oxazcpine- ) O-earboxamide. One method of synthesizing sodium (2 , 5 S, I 3aR)~7,9~dioxo- 10-((2,4,6-trifluorobenzyr)caTbamoyi)-
2^4,5J, s!3J 3a~0cia ro-2,3-met^^^
olate (e.g. a compound of Formula (If)) is described herein.
[0249] For example, in one aspect, provided is a method of producing a composition comprising one or more polymorphs of sodium (2RS5S,13aR.)~7,9~dioxo-10-((2,4s6- xrit1uorobenzyi>carbainoyi)-2,3,4,5,7!9J 13,13a-octaiiydfo-255~
methaoop τid [1 2':455] yraziBo 2 --b][I,3]ox zepm-8-oiat > wherein the method comprises combining a compound of Formula (II) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the compound of Formula (II), In another aspect, provided is another method of producing a composition comprising one or more polymorphs of sodium (2Rs5S,13aR)-7>9-dioxo-10"((2,4s6- trifluorolx2n?.y!)carbanioyI)-2,3,4,S,7,9, 13, 13a~octahydro-2,S- methanopyrido[ j2,:4}S]p>Tazino[2s i--b][l:,3]oxazepk-8"Olate, wherein the method comprises combining sodium (2R.SS, 13aR)~7,9-dioxo- 10-^^.e^Tit uor benz ljcarbant !)- 2.3,4,5,7,9, 13, 13a-oc†ahydro-2J5-methanopyridoi 1 ',2::4,5 ]pyra ino(2, 1 -b}[ ] ,3 joxazepin-S- olate with a suitable solvent or a mixture of suitable solvents,
[0250] The choice of a particular solvent or combination of solvents affects the formation favoring one polymorphic form of sodium (2R,5S,!3aR)-?.9-diox.a-iO~((2,4,6~
trill uorcbenzy l)carbamoyl)~2 , 3 ,4,5,7,9,13, 13 a-octahydro~2,S - nlethffirø ido[I^2 *4J] azin [2ϊl-b] L3]oxa2εpk"8- l te over another. Solvents suitable for polymorph formation may include, for example, methanols etharsoL water, isopropyl acetate, acetonitrile, tetrahydrofuran, methyl isohutyl ketone, and any mixtures thereof.
[0251 ] In another aspect, provided is also one or more polymorphs of sodium
(2R,5S,13aR)-7,9H*ioxo-10 (2A6-^^
2}5-methanopyrido[r>2,:4,5]p}'ra2;ino[2,l»b][13]oxa2:epiK~8~olate produced according to any of the methods described herein.
[0252] It should be understood that the methods for preparing the polymorphs described herein (including any polymorphic Form I) may yield quantity and quality differences compared to the methods for preparing sodium (2R,5S513aR)-7,9~dioxo-IO-((2,4.6- trifIuoFobenzyl)carhamoyl)-2,3,4,5,7s9, 13J 3a-0€iahydfo-2,5-
Figure imgf000075_0001
produced on laboratory seals.
Formula II, Form I
[0253] In one embodiment, provided is a method of producing a composition comprising polymorphic Form ! of sodium (2R,5S, 13aR}-759-dio¾>i0-{(2;456- trifluorobenzyl)carbamoyl)-2,3 ,4,5,7,9, 13, 13a-octa ydro-2s5- metbanopyrido r,2!:4,5]pyraz;ino[25l~b][l ,3]osa^pin"8' >lates wherein the method comprises combining (2R,5S, 13aR)-8-Hydroxy~759~dioxo-N-(254;6-trifluor benzyl)"253,4,5!7,9> 13, 13a- octahydro~255~nietbanopyrido[r,2 ,5^ a sodium base (e.g. sodium hydroxide) in a solvent to produce a composition comprising polymorphic Form I of sodium (2R,5S5i3aR)-7J-dioxo-10-((2s4,6- trifloorobc;nz 'l)carbamoyI)-2,3,4,5,7,9! 13.13a-octahydro-255- m.etka^opyr o[1^2':455]pyraz½o[2,l-b][l53]oxa2;epin~8-oiateJ wherein the solvent is selected from the group consisting of ethanol, diraethylformam ids, and any mixture thereof. In an embodiment the solvent is a mixture of ethanol and dimeftylforrnamide.
[0254] Provided is also polymorphi Form 1 of sodium (2R,5S, I3aR)-7,9-dloxo-10- ((25456»trifluorobenzyl)earbamoyl)--2J3s4,S57,9J13s i3a-octahydro-2>5~
roetham>pvrido 1 ^2 V4 S jpyraz;no[2, 1 -b) f 1 ,3]oxa2epin~8~oiate prepared by combining
(2R,5S, 13&R)~8~Hydroxy~7,9»dioxo-N^ 13,13a-octahydro-
2,5-methanopwido[rs2[:4J5]pyrazino 2,i~b][l,3]oxaxepine-10-earboxamide with a sodium base (e.g. sodium hydroxide) in a solvent, wherein the solvent is selected from the group consisting of eifianol, dimedi ifornsara ide, and any mixture ihereof. ϊη an embodiment the solvent is a mixture of ethanol and dimethylformarnide.
Formula III
[0255] One method of synthesizing (2R,5S,13aR}- g-hydroxy~7,9--dioxcv -(2,4,6-- trifiuorobenzyl)-2s3A5 ,9,i 3, i3a-GCtah^
b] 1 J joxazepine- i 0-carboxam ide (e.g. a compound of Formula (I)) has been previously described in FCT Publication No. WO201.4 100323. This reference is hereby incorporated herein by reference in its entirety, and specifically with respect to the synthesis of
(2¾5SJ3aR)-8-hydroxy-7,9^o^^ 2,5~met¾aiiQpyr!do[rs2,:4s5]p>Tazino^ One method of synthesizing potassium (2R,5S, 13aRV7,9-dioxo-- i 0 (2.4,0-tjifluorobenzyi)carbarnoyl) -
2,3,4,5 ,7,9,13J3a >ctahydro-2,5~meihanopyrM^
o!at (e.g. a compound of Formula (III)) is described herein,
[0256] For example, in one aspect, provided is a method of producing a composition comprising one or more polymorphs of potassium (2R.5S, 13aR)~7,9~dioxo~l 0~((2,4,6~ irit1uoa>benzyi)carbaTnoyi)-2,3.4 5.7,9, 13, 13a-octa ydro-255- metha o ido[1 2!:4,5]p ziu [2J -b][l !3]o¾ e in-8-ol te, wherem the method comprises combining a compound of Formula (III) with a suitable solvent or a mixture of suitable solvents to produce a composition comprising one or more polymorphs of the compound of Formula (III). In another aspect provided is another method of producing a composition comprising one or more polymorphs of potassium (2R,5S,13aR)-7,9-dioxo-10-((2,4,6- tri fluoroberszy S)carbamoyi)-2, 3,4,5,7,9,13, 13 a~octahydro-2,S - m.ethanopyrido[r,2,:4,5]p}irazino[2,l-b][l}3]oxazepin~8-olateJ wherein the method comprises combining potassium (2R(5S,13aR)-7,9-dioxo-10-((2s4,6-h"jfiyorobenzyl)carbamoy1)~
2,3,4,S,7,9,13,13a-octahydrO"2,5~metha^
ol te with a suitable solvent or a mixture of suitable solvents,
[0257] The choice of a particular solvent or combination of solvents affects the formation favoring one polymorphic form of potassium (2RS5S, 13aR)-7,9-dioxo- 10~((2,4,6- trifluorobenzyI)earbamoyi)-2,3 ,4,5,7,9, 13,13a-octahydro-2,5- niethanopyrido[!'s2':455]pyrazir3o[2, l-b][l,3]oxazepm-¾-olate over another. Solvents suitable for polymorph formation, may include, for example, methanol, etlianol, water, isopropyl acetate, aceionitrile, tetrahydrofuran, methyl isobutyl ketone, and any mixtures thereof.
[0258] In another aspect, provided is also one or more polymorphs of potassium
(2R,5S,13aR)-7,9<lioxo Q~((2A6- riffe^
2,5-memanopyrido[1^2':4,5]pyra?j.n«[2, l -b][l ,3]oxazepin-8-oiate produced according to any of the methods described herein.
[0259] It should be understood that the methods for preparing the polymorphs described herein (including any one or more of polymorphic Forms I-III) may yield quantity and quality differences compared to the methods for preparing potassium (2R,5S,13aR)-7,9-d oxo~l 0- {(2s4,6 rifIuorobenzyl)carbamoyi)~2s3,455J7,9JI 3J 3a--octahydro-2,5- meihanop rido[r52!:4JS]pyraziao[2,l-b][l,3]oxazspin~8~olate produced on laboratory scale.
Formula III Form I
[0260] Is one em odimen s provided is a method of producing a composition comprising polymorphic Form Ϊ of potassium (2ILSSs13a )-7,9~dioxo-10-((2,4,6- trifluoroberi2y])carbamoyi)-2,3s4,557,9, 13, 13a~oetahydro-2,5~
roethanopyrtdof. I ',2::4S5 Jpyrazinoj 2, i ~b j[ I ,3 joxa epin-S-oia e, wherein the meihod comprises combining (2R,5S513aR)-g~Hydroxy~7s9~dioso-N-(2;4i6-trifii5orobenzyi)-2,3s4,557s9, 13,13a- oetahydro-2,5-methanopyndo[l^^ with a potassium base (e.g. potassium acetate) in a solvent to produce a compos tion comprising polymorphic Form I of potassium (2R,5S,I 3a )»759-dioxo-10-((2s4,6- trifluorobenzyl)carfaaaioyl)~253, JS,7,9s 13, 13a~ootahydro-2:i5- niethanopyrido[ ,2':4,5jpyrazino[2,l"b][lJ3]oxa?,epin-S"Olaie) wherein the solvent is selected from the group consisting of eihanol, water, and any mixtures thereof In an embodiment, the solvent is a mixture of ethanol and water,
[0261 ] Provided is also polymorphic Form I of potassium (2R,5S,13aR)-7,9"dioxo~10- ((2s4,6 riflnorobersy'l)carbamoyl)-2J54,5,7s9,i3s13a-octahydro-2,5- methanopyrido[r52!:4J5]pyrazirio[2Ji-b]|l33]oxa¾epm--8~olate prepared by combining
(2R,5S 3aR)-8-Hydraxy-7,^
2,S~methanopyrido[1 ^2!:455]p Tazino[2J-b][l,3]oxa^epine-10 ai"bosan;iide with a potassium base (e.g. potassium acetate) in a solvent, wherein the solvent is selected from the group consisting of eihanol, water, and any mixtures thereof,
Formula III, Form II
[0262] in one embodiment, provided is a method of producing a composition comprising polymorphic Form ΙΪ of potassium (2Rs5SJ 3a )-7,9-dioxO"10-{{2,4,6~
trifluorobenzyl)carbarnoyS)-2,354,5,7,9s 13,13a-octahydro~2,5- methanopyrido[rs2f:4J5]pyrazino[2,l --b] l,3]oxazepin"8»o1ate, wherein the method comprises combining (2R,5S43aR)-8-Hydroxy-7s9^to^^
oetahydro-2,S~methanopyrMo[1^2!:4.5]pyr^ a potassium base (e.g. potassium acetate) is a solvent to produce a composition comprising polymorphic Form II of potassium (2R,5S513aR)~7,9-dioxo~10~((254,6- trifiuorobenzyl)carbamoy!)-2s3,4s557,9s13,13a--octahydrO'-2,5- methanopyrido[rs2,:4,5]p>TazIsio[2sl--bJ[li3]oxa¾epk-8~o!ate) wherein the solvent is selected from the group consisting of acetonitriie, water, and any mixtures thereof. la an embodiment, the solvent is a mixture of acetonitriie and water.
[0263] Provided is also polymorphic Form II of potassium (2R,5S.13aR)-7,9-dioxo~ I0~ {{2A04rs !uGrobenzyi}earbam
methan pyrido ,2':4}5]pyra2mo[2,l43][1 jSjoxazepm-S-oiate prepared by combining (2R,5 Ss 13aR)-8 iydroxy-7:!9-dioxo» -(2s4,64rifiuoroben2yi}--253 ,4,5,7,9, 13 , 13 a-octahydro- 2,5- sthanopyrido[ 1 ',2!:4,5 jpyrazino] 2, 1 -b][1 ,3]ox.azepinc- 1 O-carboxarnide with a potassium base (e.g. potassium acetate) in a solvent, wherein the solvent is selected from the group consisting of acetonitriie, water, and any mixtures thereof.
Formula Ml Form III
[0264] In one embodiment, provided is a method of producing a composition comprising polymorphic Form III of potassium (2R;5S, 1 aR)-7, -dioxo- 10-{(2,4,6- trifiuorobenzyI)carbamoyi)-2Js4,5,7s9J 3,13a-oetahyd!O-2,5~
methanop rido[ 1 2' :4, 5]pyrazino[2? 1 -b] [ 1 ,3]oxazepin-8-olate, wherein the method comprises combining (2R,5S, 13aR)~8-iiydroKy»7,9"dioxo4\7--(2s4,64rlI-luorobenzyl)-2J3,4,5,7,9J 13s13a- i Hahydro--2,5--ii(Kitha.riOpyrido[ ! ',2':4,5]p Tazino 2, ! ~h][ 1 ,3 loxazepine- 10-carboxamide with a potassium base (e.g. potassium phosphate) in a solvent to produce a composition comprising polymorphic Form II of potassium (2R, 5 S, 1 aRV7,9-dioxo- 1 £>-{( 2,4,6- tritluoro benzy l)carbamoyi)~253 ,4, 5, 7,9, 13,13 a-ociahydro-2,5 - metham-)pyrido[i !,2f:4,5]pyfazko[2Jl4^][i ,3]oxazepin"8-olateJ wherein the solvent is methanol,
[0265] Provided is also polymorphic Form III of potassium (2R,5S,13aR.)-7,9-dioxo-10- ((2J4,64rifIuorobenzyl)carbamoyi)-2,3,4,5,759! 13, 13a~octahydro-2,5- methanopyrido 1 ',2' :4,5]pyrazmo[2s 1 )] [ 1 ,3]oxazepin-8-olate prepared by combining
(2R,5S 3aR)-8-Hy<-roxy-7,^
2,5 -methanopyrido [Γ,2' :4, 5] pyrazino[2, 1 -b] [ 1 , 3]oxazepine- 1 ϋ-earboxamide with a potassium base (e.g. potassium phosphate) in a solvent, wherein the solvent is methanol. Formula I Co-Crystals
[0266] In one embodiment, pro ided is & method of producing a composition comprising polymorphic ciiric acid co-crystal, fbmaric acid co-crystal oxalic acid co-crystal, or a mixture thereof, of (2R3S, 1 Sal^-g-H dro y-T^-dioso-N-C ^^-triflu r benEy [)- 2,3,4,5 ,7,9,13 3a~oetahydro~2,5~rnet¾^
carboxamide, wherein the method comprises combining (2R,55, 13a )~8~Hyd.roxy~7,9-dioxo-
Figure imgf000079_0001
methanop\Tido iy2^4,5]pyrazino with an acid (e.g. citric acid, fumaric acid, or oxalic acid) in a solvent to produce a composition comprising polymorphic citric acid co-crystal, farrsarie acid co-crystal, oxalic acid co-crystal, or a mixture thereof, of the C2R.58J 3aR)~8~Hydroxy-7,9-diox0^
2,3,4,5 ,9J3, i3a-octahydro-2,5-methaiiopyrido[r!2,:4s5]pyrazmo[2 , 1 -b][l ,3]oxazepine-10- carboxamide, wherein the solvent is tetrahydrofuran,
[0267] Provided is a polymorphic citric acid co-crystal, fumaric acid co-crystal, oxalic acid co-crystal, or a mixture thereof, of(2R,5S,13aR)-8 'iydroxy-7,9^to^
Tji]uorGbenz i)-2 s4
b j [1 , 3 joxazepirie · 10 --carboxarn ide produced by combining (2R,5S, i 3aRV8~Hydroxy-7,9- dioxO"N-(2,4s6-trifluorobenz>'l)-2,3s4,5, 7,9,13 J3a-ociahydro-2,5- mcthanopyrido[I,,2,;4s5]pyrarino[2,i~b][i53]oxa epine-10~earboxamide with an acid {e.g. eitxic acid, fumaric acid, or oxalic acid) in a solvent, wherem the solvent is tetrahydrofuran,
Uses in Manufacturing of .Drug Product Formula 1
[0268] Provided are also a use of the polymorphs described herein in the manufacture of a drag product The one or more of the polymorphic forms described herein {e.g., one or more of polym orphic Forms I, IL III, IV, Vs VI, VII and VIIJ) may be used as an intermediate in the manufacturing process to produce the drug product.
[0269] in certain embodiments. Forms I to VIII of (2R,5S, 13aR}-8-!iydroxy-?,9~dioxo- N"(2,4,6 TifliJorobenz>d)-2,3,4,557,9J3,33a-octahydro-2,5"
methanop iido[r,2f:4,5]pyrazino[2J -fo][l,3]oxazepme~10-carboxamide are used in the manufacture of an active pharmaceutical ingredient, fa certain embodiments. Form Ϊ of
(2R^S 3aR)-8-Hydroxy-7,9~ta
2,5-methanopyrido[ 1^2!:4,5]p>^a^ is used in the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form II of (2R.5S, 1 :½R)-8 Jydroxy-7,9-dioxo-N--(254J6-triflirorobenz)'-!)~2!3>4s55759513,13 ~octahyd.ro-
Figure imgf000080_0001
is used in the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form III of (2R,5S 3aR)-8-Hydroxy-7,9^
2s5=metliaMOpyrido[1^2':4,3]pyrazm^ is used in the manufacture of an active pharmaceutical ingredient, fa certain embodiments, Form IV of (2¾5S 3aR)-8-Hydraxy^
2!5~me†haao yrido 1^2¾55] > a£m^ is used in the manufacture of m active pharmaceutical ingredient, fa certain embodiments, Form V of (2R,5S 3aR)-8-Hydroxy~7,9^to
2>5~meteano rido[1^2i:4s5] r^ino^ is used in the manufacture of an active pharmaceutical Ingredient. In certain embodiments. Form VI of (2 ,5S, ! 3aR)-8~Hydrox .y~739-d50xo- -(254,6 rifluoroben2}'l)~253;45557,9; 13,1 a-octah>dro- 2,5~methatto Tido[1^2':4,53p∞^ is used In the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form VII of
Figure imgf000080_0002
2,5~me haBOpyrido[1^2,:4,5]pyrazino[2,l-b][ls3]oxazepine-10-carboxaniide is used in the mannfacture of an active pharmaceutical ingredient. In certain embodiments. Form VIII of (2R,5S, 13aR)~8 1ydroxy*7,9-dioxo~N-(2.4J6-trif=uorobenzyi)-2,3,4 5!7 9$ i3,13a-octahydro~ 2,5 -methanopyrido[ 1 2' :4, 5] pyrazino [2, l~b][l ,3 joxazepine- 10-carboxaraideis used in the manufacture of an active pharmaceutical ingredient.
Formula II
[0270] Provided are also a use of the polymorphs described herein in the manufacture of a drug product. The one or more of the polymorphic forms described herein (e.g., polymorphic Form I) may be used as an intermediate in fee manufacturing process to produce the drug product.
[0271] In certain embodiments, Form I of sodium (2RS5S, 13 R)-7,9-dIoxo-10-((2A6~ trifluorobenzy 1 )earbamoy ί )- 2,3 ,4,5 ,7.9, 13J 3a-octalrydro~2,5- methanopyridoi I ^2':4,5]pyra2inoi2, 1 -b j ( 1 ,3 ]oxazepin-8-oIate arc used In the manufacture of an active pharmaceutical ingredient.
Formula III
[0272] Provided are also a use of the polymorphs described herein in the manufacture of a drug product. The one or more of the polymorphic forms described herein (e.g., one or more of polymorphic Forms I, II, and HI) may be used as an intermediate in the
manufacturing process to produce the drug product,
[0273] In certain embodiments, Farms I-IIX of potassium (2R55SsI3aR)-799-dioxo~I0~ ^^^-irifliior b njy' c rb r o l)^^^^,?^, 13, 13a-octahydro~2s5- methanopyrido[Ii J2t;4s5]pyrazino[2,i-b [l,3]oxa2Spin--8~oiate are used in the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form I of potassium
(2R3S, 13aR)-7J-dioxo 0~((2,4!6 rifl oroben2yl)carbainoyi)-2s3,4?5,799J 13,13a»octahydro- 2,5"m.ethanopyrido[1^2t:455]pyrazino[2,l"b][l,3]oxazepm~8~oIate is used in the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form II of potassium
(2R,5S 3aR)-7,9-dioxo-10-((2A6-^
2,5-methanopyrido[i^2 ,5]pyrazino[2J-b][is3]oxazepin-8 ilatc is used in the manufacture of an active pharmaceutical ingredient. In certain embodiments, Form III of potassium (2R,5S? 13a )-?,9~d.IoxG-l 0-((2;4,6 ri.ii uorobenzyl)caf bamoyl)-2,3 ,43,7,9, 13,13a~octa ydro- 2,5-methanopyrido( 1^2':4,S]pyra2ino(2, l ~b][I,3]oxazepin-8-oiaie is used in the manufacture of an active pharmaceutical ingredient.
Articles of Manufacture, and Ki ts
[0274] Compositions comprising one or more of fee polymorphic forms described herein {e.g., one or more of polymorphic Forms I to VIII of {2R,5SJ3aR)~8~Hydroxy~7s9~dioxo-N~ (2J4)6~p-ifiuorobenzyi)-2s3,455J7>9, 13, 13a-octahydro-2,5- methanopyrido[ I !,2':4,5]p>Tazino 2s 1 -b] [ 1 ,3]oxa epine- 10-carboxamide; polymorphic Form I of sodium (2R,5S,l3aR)-7,9-dioxo~10-((2J4,6 riiluoroben¾d)carbamoyl)~2 3)4,5J:!9J 13,13a. oetahydro-255~methanopyrido[i^2!:455]pyraz;ino[2,l-b][L3]oxazspm"8--oiate; and
polymorphic Forms I, II and III of potassium (2R,5S, i3aR)~7,9~dioxo 0-({2,,4,6" tri fluorobenzy l)carbamoyl)-2 , 3 ,4, 5 ,7,9, 13,13 a-octahyd ro~2,i> - metharii pyrido[l,,2i:4J]pyrazino[2J -b][i ,3]oxazepm-8-oIate) and formulated in one or more pharmaceutically acceptable carriers, excipients or other ingredients ear* be prepared, placed in an appropriate container, arid labeled for treatment of an indicated condition. Accordingly there also is contemplated an article of manufacture, such as a container comprising a dosage form of one or more of the polymorphic forms described herein (e.g., one or more of polymorphic Forms I to VIE of {2 35S>13aR)»8-Hydrox5'-7,9-dioxo-N-{2,4J6.
irifluorobeKyl)-253 ,4,5,7,9, 13, ! 3a-octahydfO"255~methanopyrido[ s2t:4,5]pyrazmo[2, \- b][ ! ,3]oxazepine- 10-carboxamide; polymorphic Form I of sodium (2R55S,13a )-7,9~dioxo~ 10-((2,456-tiif!«orobenz 4)carbamoyl)~253,4,5J59313J3a-octahydro»2;5~
methanopyridop '52::45S)pyrazmo[2J-b][L3]oxa7epin-8-ola e; and polymorphic Forms I, II» and III of potassium ( ^S^Sa J-TJ-dio o-lO^ia^^-trifluoroben ^ca b moyl)- 23,4,5,7,9, 13, 13a-octaliydro~2,5-methariOpyrido[ r,2f:4,S]pyra2;ino[2, 1 ~b][ls3]ox.a?.epin-8- olate), and a label containing instructions for use of the compound^.
[0275] In some embodiments, the article of manufacture is a container comprising a dosage form, of one or more of the polymorphic forms described herein (e.g., one or more of polymorphic Forms I to VIII of (2R..5S, 13aR)-8-Hydroxy-7,9-dioxo-N-(2 A6- trIfluorohenzyl)-2,3 ,4,5,7,9, ! 3„ 13a-octahydro--2;,5»methai'iopyTido[r>2,:4>5]pyrazino[2s 1~ b][l,3]oxazspine-1.0-carboxamide; polymorphic Form 1 of sodium (2R,5S, i3aR}-7,9-dIoxo- 1.0-({2,4,64rifli5orobe!izyl)carbamoyi)-2,3,4,5,759,13313a-octahydro-2s5~
niethanop rido[r,2':4,5]pyrazino[2,l"b][l ,3]oxazepin-S-oiate; and polymorphic Forms I, II, and III of potassium (2R,5S, i 3aR)-'7.9-dioxo- ! 0~((254,6^^
2,3,4,5,7,9 J3,13aH3etahydro-2,S~mete
oiaie), and one or more pharmaceutically acceptable carriers, excipients or other ingredients. In one embodiment of the articles of manufacture described herein, the dosage form is a tablet,
[027 1 Kits also are contemplated. For example, a kit can comprise a dosage form of a pharmaceutical composition and a package insert, containing instructions for use of the composition in treatment of a medical condition. The instructions for use in the kit may be for treating HIV. In certain embodiments, the instructions for use in the kit may be for treating HIV,
[0277] In certain embodiments, the polymorphic, salt, co-crystal and solvate forms described herein may potentially exhibit Improved properties. For example, in certain embodiments, the polymorphic, salt, co-crystal and solvate forms described herein may potentially exhibit improved stability. Such improved stability could have a potentially beneficial impact on the manufacture of the Compound of Formulas L II, and/or ill, such as for example offering the ability to store process intermediate for extended periods of time. Improved stability could also potentially benefit a composition or pharmaceutical composition of the Compound of Formulas I, II, and/or ill. In certain embodiments, the polymorphic, salt and. solvate forms described herein may also potentially result in improved yield of the Compound of Formulas I, II, and/or III, or potentially result in an improvement of the quality of the Compound of Formulas 1, 11, and/or HI. In certain embodiments, the polymorphic, salt and solvate forms described herein may also exhibit improved
pharmacokinetic properties and/or potentially improved bioavailability.
Methods
Formula 1 Form I
[0278] (2R.5S, 13aR)-8-hydroxy-7s9-dioxo-N~(254,6-trifl oroben2yl)-2,3s4,5s7,9513,13a- octahydro-2,5-methanopyrido[ 1 !,2':4,5jpyrazino[2, 1 -b] [ 1 ,3joxazepin< 1 O-carboxaniide (73 rag) was added to a glass vial. Isoproparsol (1 mL) was added, the vial was capped, and the suspension was stirred at about 21 ''€. for not less than 5 days. Formula I Form I was isolated as a solid from the suspension by centrifuge/filtration md characterized as discussed below.
Formula I Form II
[0279] (2 ,5S, 13aR) ¾droxy-7,9-dioxo-N 2,4,6-tiiflisoroben¾d)-2,3,435,759, 13,13a- QctahydrQ~255~methanop rido[ Ϊ V^4,5]pyrazinc[2, l-b][i JJoxazepine- 10-carboxamide (60 mg) was added to a glass vial Methyl tert-butyl ether (I mL) was added, the vial was capped, and the suspension was stirred at about 21 °C for not less than 5 days. Formula I Form 11 was isolated as a solid from the suspension by centrifuge/fiitraiion and characterized as discussed below.
[0280] (211,58, 13aR)~8¾droxy~7,9~dioxo»N-(2s436.tsiftuoroben2y1)-253,4;5,7.,9, 13,13a- octa ydro-2,5-methanopvxido[ 1 ^2':4,5 jpyrazino[2, 1 -b]( i ,3]oxazepine~l Q-carboxamide (98 mg) was suspended in ethanol/water (1 mL, aw 0.7-0.8) for 5 days with agitation. Formula 1 Form II was isolated as a solid from the suspension by centrifuge/filtration and characterized as discussed below. Formula I Form HI from Compound ?-!«
Figure imgf000084_0001
Compound G1~a
[0281 J A solution of about 10% (w w) Compound G-la (2.5 g Compound 1001) k methylene chloride was concentrated to a residue under vacuum. LiCl (2.6 g, 7 equiv) followed by N-methyl-2~pyrroiidone (12,5 mL) was added to the resulting residue, The method of preparing compound Gt-a can be determined by one skilled on the art, for example, as described in co-pending PCX Serial No, US2013/076367, filed December 19, 2013 entitled, "FOLYCYCLIC-CA BAMOYLPYRIDONE COMPOUNDS AND THEIR PHARMACEUTIC ΑΙ.· USE." The mixture was heated to an internal temperature of approximately 75 °C. After 2,5 hours, the reaction was cooled to approximately 20 °C. Dichloromethane (12.5 mL) and 0.SM hydrochloric acid (12.5 mL) was added, and the resulting mixture stirred for 5 minutes. The phases were separated, and the organic layer was washed with 10% aqueous sodium chloride solution (twice) followed by water. This solution was concentrated while gradually adding 3 volumes of ssopropyl alcohol portionwise (40 °C bath temperature, 200-230 torr vacuum). The resulting slurry was slowly cooled to 2-4 °C. The product was filtered and deliquored and dried. Formula I Form III was solated and characterized as discussed below.
Formula I Form 111 from Formula I [0282] (2 ,5S5I3aR)-8¾droxy-7,9~dioxo~^
octahydro~255~methanopyrido[ 1 ',2* ;455]pyrazino[2, 1. -b] [ i ,3 joxazepme- 1 O-carboxarrude (lOOmg) and etbanol (0.5 mL) were added to a reaction vessel and seeded with Formula I Form III. The slurry was allowed to age at room temperature for about 18 hours. Formula I Form III was isolated as a solid from the suspension by centrifuge/filtration and characterized as discussed below. Formula I Form IV
[0283] (2R55S43a )-8¾'droxy-7J"dioxo~N^2)4,6 rii]uorobesi2yl)~2 455J59J3si3a- octaliydro~2,5-meihanopyrido[ 1 ! 52!:4,S]pyradno[2, 1 ~b][l s3]oxazepine- 1 G-carboxamide (406 mg) and potassium acetate (200 rng) were added to a glass vial. Methanol (5 mL) was added, the vial was capped, and the suspension was stirred at about 21 °C, After 11. days the solids were isolated from fee suspension by centrifuge/filtration and the filtrate retained, Afler slow evaporation over 16 days, large crystals of Formula I Form IV were found in the filtrate vessel.
Formula I Form ¥
[0284]
Figure imgf000085_0001
2,3S4,SS7,9 J 3, 13a-octahydro~2,5-mette
carboxamide (25,2 mg) was mixed with 1.21 grams of water at room temperature, forming a solution. After several days, crystals suitable for single crystal X-ray crystallography were found and analyzed,
Formula I Form VI
[0285] 173 mg of (2R>SSJ3aR}-8-hydroxy-^
2 4^7,9> ! 5 ! a-octah drO"255^
carboxamide (25.2 mg) was suspended in 2 mL of rae hanol water solution (aw 0.5), The suspension was filtered the next day and several days after that, crystals suitable for single crystal X-ray crystallography were found in the filtrate and analyzed.
Formula I Form VII
[0286] 112,3 rng C2 43aR)~S~h rc -^
2^4I5 ,9 3,13a-ociahydro-2,5-raeihanopy
carboxamide (25.2 mg) suspended in 0.95 grams of methanol at room temperature. A sample of the suspension was filtered for testing after several days, and several days post-fiitration, solids suitable for single crystal X-ray crystallography were found in the filtrate and analyzed. Formula I Form VIII
[0287] 53 g (2R,5S 3aIi)-8-hydrox -7J~dioxo-N-(2 4i6-t ifluoroben¾ [)-
2,3,4.5,7,9,13 J 3aK>etahydro-2sS-meta
carhoxamide (25.2 nig) suspended in SO mL of methanol at room temperature. To the mixture was slowly added 0,70 grams of KO'H dissolved in 20 mL methanol. Full addition of KOH solution resulted m a solution, A suspension formed over the course of two days and a sample was filtered for testing after ten days. After several days, solids suitable for single crystal X-ray crystallography were found in the filtrate and analyzed.
Formula II Form I
[028S] (2R,5S, 13aR}-§~hydrosy-7. -dIoxo-N~(254,6 ritluoroben2 'i)-253!435J;9513,13a- octahydro-2,5~methanopyrido[l. 's2!:4,5]pyrazino[2, 1 -b] [ 1 ,3]oxazepine-l 0-earhoxamide (20 g) ars.d ethanol (80 mL) were added to a reaction vessel and warmed to about 75 CC. Aqueous sodium hydroxide (22 mL 2 M solution) was added over approximately 30 minutes, after which the slurry was cooled to approximately 20 °C over approximately one hour. Sodium
(2R,SSs 13aR)-7,9-dioxo-10H;(2,4,6-irifluorobefl^
25S-m.ethanopyrido[I! 52':435]pyra2ino[2,l -bl[L3]oxazepin-8-oiate Form I was collected by filtration, washed with EiOI ! (SO mL) and dried under vacuum.
[0289] 1H MR (400 MHz, DMSO-d6) 8 10.63 (fc, I - 5,8 Hz, 1H), 7.88 (s, Hi), 7,29 ~ 7,07 (m, 2H), 5,20 (dd, I - 8.6, 3.6 Hz, 1H), 5.09 (t, J - 4.1 Hz, 1 H)S 4.52 (m, 3H), 4,35 (dd, J = 12.8, 3.6 Hz, 1H), 3.87 (dd, I = 12.7, 8,7 Hz, 1H), 2.03 - Ϊ .80 (m, 3H), 1.76-1.64 (m, 2H), 1.50 - 1.40 (m, 1H).
Formula I Citric Acid Co-crystal Form I [0290] C2R,5S,i3aR 8-hydroxy-?59-diox^^
octahydro-2!5-methanopwido[ I f,2':4,S]pyrazino[2, ί ~b] [ l,3]ox.azepir.e- 1 G-carboxamide (131 nig) and citric acid (148 rng) were added to a glass vial Tetrahydro&ran (1 mL) was added, the vial was capped, and the mixture was stirred at about 21 "C for two days. The vial was vented and the solvent was allowed to evaporate at about 21 °C unassisted. After eight weeks at room temperature, crystals were found in the vessel and were identified as a 1:2 Formula I citric acid co-crystal.
§4 Formula I Fumaric Acid Co-crystal Form 1 [0291] (2R,5S 3a )-8~hydroxy-^^
octahydro"2,5-metliaiTopyrkk)[1 ^2::4.5 lpyrazino[251 -b][ tJJoxazepirse- 10-carboxamide (131 mg) aod fumaric acid (103 mg) were added to a glass vial. Te rahydrofura (1 mL) was added, the vial capped, and the suspension stirred at about 21 °C for two days. An additional 1 mL of tetrahydrofuran was added, and the mixture was heated to 45 °C. After about 12 hours at 45 ftC the mixture was fou d to be fully dissolved wd was removed from the heat bath. The vial was vented, and the solvent left to evaporate at room temperature. After one day solids were observed m the vial and it was re-capped. After eight weeks large crystals were found in the capped vial and identified as a ! : 1 Formula I fomaric acid co-crystal
Formula I Oxalic Acid Co-crystal Form I [0292] (2R,5SJ3aR)-8-hydrox -7,9^^
octahydro-2>5-me&anop}Tido[l^ (147 mg) arid oxalic acid (134 mg) were added to a glass vial, Tetrahydrofuran (1. mL) was added, the vial capped, and the resulting solution was stirred at about 21 °C for about two days. The vessel was vented, and the solvent was allowed to evaporate at room temperature unassisted. After ne day solids were observed m " the vial and it was re-capped. Two days later large crystals were found in the vial and were identified as a 1 :1 Formula I oxalic acid co-crystal
Formula EI Form I
[0293] 406 mg
Figure imgf000087_0001
23,4,5,7,9 3, 13a~octahydro~255-metim^
carboxamsde (25.2 mg) was combined with 200 mg potassium acetate and 5 mL of methanol at room temperature, resulting in a suspension. A sample of the suspension was filtered the next day for testing. After several days, solids suitable for single crystal X-ray
crystallography were found In the filtrate and analyzed.
Figure imgf000087_0002
octahydro~255~methanopyTido[r,2t:4^ (1.96 g) was charged to a reaction flask and stirred, followed by etbanol (20 mL), The mixture was stirred at room temperature resulting in a suspension. In a separate vessel potassium hydroxide (253 mg) was dissolved in deionized water (5 ml,). The potassium hydroxide solution was transferred by syringe pump to the stirring suspension over about 2.5 hours followed by a rinse into the reactor with water (2 mL) after the base solution transfer was completed.
[0295] A sample of about 0,5 mL of the resulting suspension was centrifuge/filtered. The filtrate was retained and stored at room temperature. After several weeks it had evaporated and the vessel contained large crystals, which were Identified by single x-ray crystallography as Formula III Form 11.
Formula III Form II (Dimer)
[0296] 94,8 mg of (2R,5S 3aR)~8-hydroxy^
2,3,4,.V<v,9,13,I a-octohydro~2 5~meto^
carboxamide (25,2 mg) was suspended In I mL of aceionitri!e/water (1:1). After one week a sample was filtered for testing. After several days, solids suitable for single crystal X-ray crystallography were found in the filtrate and analyzed.
Formula 111 Form III
[0297] A 4 mL glass vial was charged with 140 mg of (2Rs5S,13aR)-S-hydroxy-7i9- dio o-N-C ^^-trifluorobenzylJ^.S^^J , 13S 13 -octahydro-2,S- inethanopyrido[rs2!:4,5]pyrazim¾[2J »b][1 ]o¾azepirse--10-carboxarnide, and 73 mg of potassium phosphate and 2 ml, of methanol. The vial was capped and placed in a rotating mixer to provide gentle and constant mixing. The experiment was carried out at room temperature. More than a week later and more than a month later solids were isolated by centrifuge/filtration and examined by XRPD. Large crystals suitable for Single Crystal X~ray Crystallography were found and analyzed.
[0298] The crystalline forms of the present invention were characterized by various analytical techniques, including X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), and dynamic vapor sorption (DVS) using the procedures described below,
[0299] X-R y Powder Diffraction', XRPD analysis was conducted on a dlffractometer
§6 (PA analyticai XPERT-PRO, PANanaiytical B.V., Almeio, Netherlands) using copper radiation (Cu Κα, λ = 1.5418 A). Samples were prepared for analysis by depositing the powdered sample in the center of an aluminum holder equipped with a zero background plate. The generator was operated at a voltage of 45 kV and amperage of 40 mA. Slits used were Sailer 0,02 rad„ antiscatter L0°, and divergence. The sample rotation speed was 2 see. Scans were performed from 2 to 40° 20 during 15 min with a step size of 0.0167° 29. Data analysis was performed by X'Pert Highscore version 2,2c (PANalytical B.V., Aknelo, Netherlands) and X'Pert data viewer version 1.2d (PANalytical B.Y., Almeio, Netherlands),
[0300] The XRPD pattern for (2RJS, 13aR)-8~hydroxy-7,9-dioxo-N-{2J456- irifhsor he z l)-2,3AS,7,943 3 ~o^^
bj 13 joxazepine- .1 O-curboxamide Form I is represented in Figure I .
[0301] The XRPD pattern for (2R,5Sa3aR)-S~hyd=Oxy-7,9-d;oxo~N-(2!4,6- trif]uoroben2}4)-2>334,5!,7,95 ! 3, 13a-oelahydro~2!5-mediariopyrido[r,2':4,S]pyrazino[2, 1- b][l s3]oxa2epine- 10~earboxa?nide F orm II is represented in Figure 2.
[0302] The XRPD pattern for (2R;5S?13aR)~8~hydroxy~759-dioxo~N~^
trifluorob nzyl^l^ASJ^JJ Sa-oc^
b][l 53]oxaz.epme 0 aiix>xamide Form III is represented in Figure 3.
[0.303] The XRPD pattern for (2R,5S, 13aR)-8-hydroxy-7,9^ox^
triiluoroben;¾ l)-2 3,4,5,7s9, 13, 13a-cctahydro~2>5-meih&rsopyrido[ 1 ',2!:4S5 ]pyrazino[2.1 - b] 1.3 joxazepine- 10~earboxarn kie Form IV is represented in Figure 4.
[0304] The XRPD pattern for sodium (2R,5SJ 3a )-?59~dioxo~lG~{(254,6~
trifluorober!2yl)carbainoy[)-2.3,4,5,7,9, 13,13a~oetahydro-2.,S~
methaROpyrido[ 1 ',2' :4, 5]pyrazmo[2, 1 -b] [ 1 ,3]Gxazepiri~8~oiate Form 1 is represented in Figure 5. The calculated XRPD pattern for sodium (2R,5 S, 13aR)~7,9-dioxo~ 10-((2,4,6- trifiif.orobenzyl)earbamoyI)-253,4i5,7s9, 1.3 , 13a~octahydro-2,5-- raethanop>Tido[r,2f:4;5]pyrazino[2,l--b][L3]oxazepin-8-oIate Form Ϊ represented in Figure 5 was calculated by using Mercury 3.1 Development (Build RCS), Single crystal data for sodium (2R,5S, 13aR)-7,9<lIoxo~l 0-((254,6 riil«orohenz>'l)carbamoyl)~2>3,4i5,7J9:,l 3, 13a~ octahydro~255~methanopyrido[1^2,;4;5]pyrazino[2 I~b][l.s3]oxazepin"8-olate Form I was input into Mercury 3.1 Development (Build RCS) to calculate the XRPD pattern for sodium C2R55SJ3aR)-7! ~dioxG 0 i2A6~irifiuorobenCT^^ 2,5~metlianopyrido[ ,2!:4,5}py!'azirio[2J 3][] ,3joxazepm~8~olate Form L Bulk material, such as sfcoichioraetry arity between the temperature was obtained on a Rigaku Miniflex II XRD usin power settings of 40kV, ISniA, scan speed of 2,0000 degrees per minute, a Miniflex 300/600 goniometer and an ASC-6 attachment, a scan range of 3.000 to 40.000 degrees, an incident slit of 1.250 degress, a length limiting slit of 10,0 mm, and SC-70 detector, a receiving slit #1 of 1 ,250 degrees, continuous scan mode, and a receiving slit #2 of 0.3mm. The sample was prepared by smoothing about 20 mg of solids on a silicon disk mounted in a metal holder. Acquisition temperature was -21 °C.
[0305] The XRPD pattern for sodium (2R,5S,13aR)-739-dioxo-10-({254,6- triilucHTtbenzyl)carbamo>'l)-2,3,4<5,7,9, 13 J 3a-oeteriydro-2,5- met¾anop} Mo[r52!:455]p Tazino[2,I~b][l53]ox^«pffl-8-olate Form I is further represented in Figure 16. The calculated XRPD pattern for sodium (2R,5S,13aR)»7i9-dioxo-10-((2,4,6- trifluosx5benz ])carbamoyl)-2,3,4,5t7 9, 13, 1 a-octahy d ro-2,5 - methanopyrido[l',2,:4,5]pyrazino[2,l.-b][l !3]oxazepm-8-oIate For ί represented in Figure 1.6 was calculated by using Mercury 3.1 Development (Build RC5). Single crystal data for sodium (2R ,5 S, 13aR)~7,9-dioxo- 10-((2A6-trifluorobenzyi)carbamoy! 253 ,4,5,7,9, 13, 13a- octahydro-2s5-methanopyrido[ V, :4,5]pyrazmo[2, 1 ~b] [ 1 ,3]oxa;iepm-8-olate Form I was input into Mercury 3.1 Development (Build RC5) to calculate the XRPD pattern for sodium <2R55S,i:½R}-7,9~dioxo 0-iC2,4}6 rifm^
2,5-med*anopyrido[ 1 !.2':4,S]pyn¾zino(2, 1 -b][! ,3]oxazepin-S-oIate Form I. Bulk material, such as stoichiometry arity between the temperature was obtained on a Rigaku Mini ile II XRD using power settings of 40kV, 15mA, scan speed of 2,0000 degrees per minute, a Miniflex 300/600 goniometer and an ASC-6 attachment, a scan range of 3.000 to 40.000 degrees, an incident slit of 1.250 degress, a length limiting slit of 1.0,0 mm, and SC-70 detector, a receiving slit #1 of 1.250 degrees, continuous scan mode, and a receiving slit #2 of 0.3mm, The sample was prepared by smoothing about 20 rag of solids on a silicon disk mounted in a metal holder. Acquisition temperature was -21 °C.
[0306] Figure 16 compares the calculated XRPD pattern of sodium {2R,5S,13aR}~7!9- io o 0H(2A6-tfiflu roben¾1)c ^
metba»opyrido[r,2':4,5]pyrazmo[2,l-b][1 ]oxazepiri~8~olate Form I to the experimental XRPD pattern of sodium (2R,5SJ
Figure imgf000090_0001
2,3AS!7, 3J3a-oetahydriv-2,S~m^ elate Form I. The comparison shows the degree to which the calculated XRPD and experimental XRPD agree. Strong agreement indicates the solved crystal structure is also the crystal stmciure of the material analyzed directly by XRPD. This determination can support orthogonal data about the composition of the bulk material, such as stoichiom etry.
[0307] The actual and calculated XRPD pattern for (2R-5S, 13aR)-8-hydroxy-7(9-dioxo-
Figure imgf000091_0001
methanopyrktoi 1 ',2' :4, 5]pyrazino[2, i ~b] [ 1 ,3 joxazepine- 1 O-carboxaniide oxalic acid co- crystal Form Ϊ is represented in Figure 6,
[0308] Differential scanning calorimetry Thermal properties were evaluated using a Differential Scanning Calorimetry (DSC) instrument (TA QIGGO, TA instruments, New Castle, DE, USA). Approximately 1 to 10 mg of solid sample was placed in a standard aluminum pan vented with a pinhole for each experiment and heated at a rate of 5 to 10 °C/min under a 50 mL/miii nitrogen purge. Data analysis was conducted using Universal Analysis 2000 Version 4.7A. (TA Instruments, New Castle, DE, USA), Heat of fusion analysis was conducted by sigmoidal integration of the endother ic melting peak.
[0309] The DSC for (IRJSJSa i-g^h dro y-T^-dio o-N-Cl^^-t iil orobenz l)-
Figure imgf000091_0002
e-arboxatmde Form I is represented in Figure 7.
[0310] The DSC for {2R,5S,13aR)-8»hydroxy.7,9~dioxo-N-{2,436.tfifluorobenzyi)~
253AS A13,13a-octahydro-2>5-m^
carboxarnide Form III is represented in Figure 8,
[031 1 ] The DSC for sodium (2RJ5S/!:kiR}-?,9-dioxo- 10~((2>4,6- ti1fluorok;nzyl}carbamoyl)~2>3.455,7?9513, 13a-ociahydro-2,S- meihanop3' ido[i^2!:435]pyra^ino[2J1 i][l ,3]oxazepin-8-olaie Form I is represented in Figure 9.
[0312] Thermogr itmtric analysis: Thermogravlmetric analysis (TGA) was performed on a TGA instrument (TA Q5G0, TA Instruments, New Castle, DE, USA). Approximately 1 to 10 mg of solid sample was placed in an open aluminum pan for each experiment and heated at a rate of 5 to 10 °C/min under a 60 mL/min nitrogen purge using. Data analysis was conducted using Universal Analysis 2000 Version 4.7A (TA Instruments, New Castle, DE, USA),
[0313] The TGA for (2R;5S, 13aR}~8-hydroxy-7,9-diox >N-(2; >6 rifhiorab.inz l)~ 2.3,4,5,7,9, 13, 13a-ociahydro-2,5-s¾ethanopyrido[ 1 \2 ]pyrazino[2,, 1 -bji 1 ,3]oxazepine- 10- carboxamide Form I is represented in Figure 10,
[0314] The TGA for (2R,5S, 13aR)~8~hydroxy-7,9-dioxo~N~(2A6«
2,3,4.5,7,9, 13,13a >ctahydro-2}5~meth^
carboxasnide Form ΠΪ is represented in Figure 11.
[0315] The TGA for sodium (2R,SS5 i3aR)-7>9-dioxo-10-{{2,4,6- riiluoro eri y c amoyi)^^^^,?^:, 13. i 3a-octa ydro-2.5- methanopyrido[ 1 ,,2!;4s5]pyrazino[2, 1 -b j [ 153]oxazepin~8~olate Form I is represented in Figure 12,
[0316] Dynamic vapor sorption: The hygroscopicity was evaluated at room temperature using a dynamic vapor sorption (DVS) instrument (TGA Q5000 TA Instruments, New Castle, DE), Water adsorption and desorption were studied as a function of relative humidity (RH) over the range of 0 to 90% at 25 8C. The relative humidity in the chamber was increased by 10% RH and held until the so lid and atmosphere reached equilibration. The equilibrium test was continued until passed or expired after 5 or 10 hours. At this point, RH was raised 10% higher and the process was repeated until 90% RH was reached and equilibrated. During this period, the water sorption was monitored. For desorption, the relative humidity was decreased in a similar manner to measure a foil sorption/desorptlors cycle. The cycle was optionally repeated. All experiments were operated in dm/dt mode (mass variation over time) to determine the equilibration endpoiut. Approximately 5-10 g of solid was used, Data analysis was conducted using Universal Analysis 2000 Version 4.7A (TA Instruments, New Castle, DE, USA),
[0317] The DVS for (2R ,5S, 13aR)~8¾droxy-7,9~dIoxo~N 2 ,4,6-trifiuarote
2,3,4,5,7,9,13, 13a-<x>iJ dro-295~me^ l-b][l,3]oxa2epme-10- carboxamide Form I is represented in Figure 13,
[0318] The DVS for (2R,5S.13aR)-8-hydroxy-7,9-dioxo-^
2,3A5,7,9913s13a~octahydfo~2,5~^^
carboxamide Form TIT is represented in Figure 14, [0319] The DVS for sodium (2R.5S, 13aR)-7,9-dioxo- 10-((2,4,6~
trif liiorobenzyl)earbamoy !)~233,4,5,7,9,13,13 a~octahydro-2, 5- meihanopyridof I ',2':4,5]pyrazino[2, 1 -b|( 1 ,3]oxazcpi.n-8-olatc Form I is represented in Figure 15.
[0320] The single crystal X-ray diffraction studies were carried out on a Broker APEX II Ultra difrractometer equipped with Mo Ka radiation (e.g. Wavelength). Crystals of the subject compound were cut into a 0.22 x 0.18 x 0.04 mm section and mounted on a Cryoloop with Paratone-N oil. Data were collected in a nitrogen gas stream at a particular temperature as shown in the Tables below (e.g. 1.00(2) K or 200(2) K). A total number of reflections were collected covering the indices, (e.g. -9<~ά<-ί Gs -I <==k<^l6, -37<=¾k-=36). Certain reflections were found to be symmetry independent with a Rmi value. Indexing and unit-ceil refinement indicated a crystal system (e.g. monochnic, tridinic, or orthorhombic lattice). The space group, which was uniquely defined by the systematic absences in the data, was found (e.g. Pi , P2(l), C2, and P21212). The data were integrated using the Broker SAINT software program and scaled using the SADABS software program. Solution by direct methods (SHELXT) produced a complete phasing model compatible with the proposed structure.
[0321] All nonhydrogen atoms were refined anisotropicall by full-matrix least-squares (SHELXL-201 ), All hydrogen atoms were placed using a riding model. Their positions were constrained relative to their parent atom using the appropriate HFIX command in SHELXL- 2014, Crystal iographic data are summarized in tables below. The absolute stereochemistry was set to conform to previously studied samples of the same compound,
[0322] The single crystal X-ray crystallography data for Formula I Forms I -IV are summ rized in Table 2A below. The single crystal X-ray crystallography data for Formula I Forms V-VIII are summarized in Table 2Α-Ϊ below. The indexing data for Formula II Form ! is summarized in Table 2C below. The single crystal X-ray crystallography data for the co- crystals of the present, in vention summarized in Table 2C below. The single crystal X-ray crystallography data for Formula III Forms I ΙΙΪ is summarized in Table 2D-I below. Data from furt her characterization of the crystals ate summarized in Tables 3 A and 38 below. Data from further characterization of the crystals are also summarized in Tables 3A and 3C~i below.
Table 2 A: Single Crystal Data for Formula I Forms I - IV and Formula II Form I
Figure imgf000094_0001
 Table 2Α-Ϊ: Single Crystal Data for Formula I Forms V - VIII
Figure imgf000095_0001
Table 2 : indexing Data for Formula II Form I
Figure imgf000095_0002
[0323] The single crystal X-ray crystallography data for Formula ΙΪ Form I is summarized in Table 2B-I below.
Table 2I : Single Crystal Data for Formula II, Form I
Acquisition Space
Z Unit Call Dimensions
C42 K34 F6 N6 Temp. Group
Na2 O10
100(2) K P2I2121 4 Distance (A) Angle f )
Form and Solvent in Density
Solvent a b C a identification lattice ( g/at3) β y
Formula ΪΪ 8.9S6I 13,9202 31.115
Ethanol/DMF none 1.614 90 90 90 Form. I (10) (14) (3)
Table 2€: Single Crystal Data for Formula I Co-Crystals
Figure imgf000096_0001
Single Crystal Data for Fonnuk III, Forms I-III
Figure imgf000097_0001
Table 3A: Crystal Data and Structure Refinement for Formula I Forms ί
Figure imgf000097_0002
Table 3A-I: Crystal Data and Structure Refinement for Formula Ϊ Forms V - Vill
Figure imgf000098_0001
Table 3B: Crystal Data and Structure Refinement for Formula I Co-Crystals
Figure imgf000098_0002
Table 3C-h Crystal Data and Structure Refinement for Formula ΪΠ Form I, Formula HI Form II (Dimer), arid Formula III, Form III
Figure imgf000099_0001
[0324] In certain embodiments of the invention, Formula III is bydrated. In certain embodiments, Formula III is hydrated with five to six water molecules.
[0325] Each of tire references including all patents, patent applications and publications cited in the present application is incorporated herein by reference in its entirety, as if each of them is individually incorporated. Farther, it would be appreciated that, in the above teaching of invention, the skilled in the ar could make certain changes or modifications to the invention, and these equivalents would still be wi thin the scope of the invention defined by the appended claims of the application. Each of the references including all patents, patent applications and publications cited in the present application is incorporated herein by reference in its entirety, as if each of them is individually incorporated. Further, it would be appreciated that, in the above teaching of invention, the skilled in the art could make certain changes or modifications to the invention, and these equivalents would still be within the scope of the invention defined by the appended claims of the application.

Claims

What is Claimed: 1. A compound selected from the group consisting of:
Figure imgf000100_0001
octahydrO"2)5~me&aiiopyrido[l,,2\'4,5]pyra£3¾o 2J ^][l,3]oxa¾epme-10- carboxamide Form I;
^SSJS R^g^lydro -T^dio o-N^
ociahydiT>~2,5-methao.opyrido[ 1 ',2':4,5]pyra2mo[2, 1 -b]f 1 ,3}oxazepine- 10- carboxamide Form ΪΪ;
{2R,SSJ3aR)-8.eydroxy-7J9-dioxo-N 25456 Tif! ofobenzyr)«2 54s5 s9a3J3a- ociabydro~2)5~ ethanopyrido[ 1 ',Ι'Ά, Sjpyrazmop., i -h] [1 ,3 ]oxazepine- 10~ carboxarr de Form III;
(2R,5S 3aR)-8-Hydroxy-7,9<fc^^
octahydro-2>5-methanopyrido[rj2,:4>5]p>Ta¾mo[2s 1-b] [1 , ]o s2isp e- 10- carboxamide Form IV;
(2R,5S, 1 ;¼R}»8-Hydroxy-7!9~dloxO"N-(2s4,6-iriiluorobenz> )-253J4,S57;9, 13, 13a- octahydro-2,5-methanopyrido[l ',2' :4,5]pyrazino[2, 1 ~b] [ 1. ,3]oxazepine- 10- carboxaraide Form V ;
(2R5SSa3aR)~8~Hydroxy-7>9~dioxo-N-(2)456 rifiuorobenxyl)-2 ^;5 759513J3a-- ociahydro-2,5-meiliariOpyrido[ .1 !,2':4,5]pyra5;irjo[251 -b'j { 1 ,3]oxazepme~ 1.0~ carboxamide Form VI;
(2R.5S, J.3aR)-8 4ydro¾y-7,9-dioxo-N"(2i456 rii1uofobeszyi).253,455,7,9513,1.3a- octahydr o-2,5~methanopyrido[ 1 ;4s5]pyrazin.o[2s 1 -b] [ 1 ,3]oxazepiEe~ 10~ carboxamide Form VII; (2R ,5S ,13aR.)~8~Hydtosy"7 -dioxo-N-(2s456-tiit¾oroben¾yI)-2,354,5,7J9, 13, 13a~
Figure imgf000101_0001
carboxamide Form VIII;
(2R,5S!13aR)"8-Hydroxy»7,9-dIoxo.N-(2,456 rifiuorob n¾yl)-2 ,4J5J59,l 3,13a- octahydro~2sS-metfhanopyrido[ 1 !,2':4,5 ]pyrazino[2, 1 -b [[ 1 ,3]oxaxepf ne~ 10~ carboxamide fumaric acid co-crystal;
(211,58, 13aR}.8»Hydroxy~7s9-dk>xo-N-(234;6 riflisorobeo2yl)-2,3s4!3s7,,9s 13, ! 3a- ociahydfG-23S-methaaopyrido[ ,2?:4,5Jp
carboxamide citric acid co-crystal;
(2R,5S, OaR)-8-eydfoxy-759-dioxO"N-(254,6-trifiuoroben¾4).2,3,45557,9, 13, 3a- octahydro-2,5~methariapyrida I !,2,:4,5]pyrazino[2, 1 -b][l ,3]oxazepme~1.0~ carboxamide oxalic acid co-crystal; potassium (2R,53, 13 aR}-7,9~dioxo- 10"((2,4?6-iriiluoroben2yr}carbamoyi}- 2,3,4.5,7,9 J3 3a-octahydro-2,S-Bietl5aiiopyrido[ s2::4J5]pyra Mo[25l- b] 1 ,3]oxazepin-8-oiate Form L potassium (2RJ5S,13aR)-7J9-dioxo 0~((2s4;6 rifluorobenzyl)cai sainoyl)- 2,3,4,5,7,9, 13, 3a"OCtabydro~2,5»rrtethanopyrido[r,2,;4s5]pyra2;ko[251 - b][ 1 ,3joxazepin-8-olatc Forra II; arid potassium (2R,5SJ3aR)-7,9Klioxo-.[0-({2J456"irifiuoroberi2yI)carbamoyI)- 2,3,4,5,7,9, 13, 13a~ociahydra~2s5-methanopyrido[ 1 ',2':4,5}pyrazma[2, 1 - b][l,3]Gxazepm~8~okte Form III.
2. A crystalline form of (2R,SS,13aR)4¾^ydroxy~7s9~dioxo-N' 2,456 rii¾orob!5nzyl)~ 2,3,4,5,7,9,13,13a etaaydro-2,5~mette^
carboxamide, wherein the crystalline form is Form I.
3. The crystalline form, of claim 2, characterized by an x-ray powder diffraction (XRPD) pattern having peaks at about 13. , 16.2°, and 20.6° 2-Θ ± 0.2° 2-Θ
4. The crystalline form of claim 3, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 25.0° and 27.4° 2-Θ ± 0.2° 2-Θ.
5. The crystalline form of claim 3 or 4, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 9.3° and 10.5° 2-θ ± 0.2ΰ 2-8,
6. The crystalline form of any one of claims 3 to 5, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 11.4°, 13,9°, 17.4°, and 22.3° 2-Θ ± 0.2° 2-Θ.
7. The crystalline form of any one of claims 3 to 6, characterized by an x-ray powder diffraction (XRPD) pattern substantially as set forth in Figure 1.
8. The crystalline form of arty one of claims 3 to 7, characterized by differential scanning calorimetry (DSC) pattern substantially as set forth in Figure 7.
9. The crystalline fonts of any one of elaims 3 to 8, characterized by a dynamic vapor sorption (DVS) pattern substantially as set forth m Figure .13.
I.0. A crystalline form of (2R.5S 3aR>8-hydroxy-7,9^^^
2,3,4,5 ,7,9,13, 13a~ocmhydro~2J~methanop^
carboxamlde, wherein the crystalline form is Form !L
I I , The crystalline form of claim 10. characterized by an x-ray powder diffraction (XRPD) pattern having peaks at about 6.6°, 10.6°, and 12,5° 2-Θ ± 0.2° 2-Θ
12, The crystalline form of claim 11, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 16.2" and 21.4° 2-Θ ± 0.2° 2-Θ.
13. The crystalline form of any one of claims 1 1 or 12, wherein the x-ray powder diffraction (XRPD) pattern has further peaks ai about 14.3°, 23,5°, and 25.6° 2-0 ± 0.2° 2-0.
14. The crystalline form of any one of claims 1.0 to 13, characterized by an x-ray powder diffraction (XRPD) pattern substantially as set forth Figure 2,
15. A crystalline form of (2R,5SJ3aR)~8~hydroxy-?,9-dioxo-N~(2s4,6~trifluorobenzyl)- 2,3,4,5,7,9, 13,13a-octaliydro-2,5-metiianopyrido[1^2i:4,5]pyrazmo[2; 1 ~b][is3]oxazepke-10- earbcxarrssde, wherein the crystalline form is Form 111,
16. The crystalline form of claim 15, characterized by an x-ray powder diffraction (XRPD) pattern having peaks at about 9.6°, 14.0% and 18.5° 2-Θ ± 0.2° 2-Θ
17. The crystalline form of claim 16, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 20.G* and 22.5° 2-8 ± 0.2° 2-0,
18. The crystalline form of claim 16 or 17, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 12, 1 ° and 16.2° 2-Θ ± 0.2° 2-Θ.
1 . The crystalline form of any one of claims 16 to 18, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 25.0°, 27.0°, and 29.0° 2-0 ± 0.2° 2-Θ.
20. The crystalline form of any one of claims 15 to 1 , characterized by an x-ray powder diffraction (XRPD) pattern substantially as set forth in Figure 3,
21. The crystalline form of any one of claims 15 to 20, characterized by differential scanning ealorinietry (DSC) pattern substantially as set forth in Figure 8.
22. The crystalline form of any one of claims 15 to 2 !s characterized by a dynamic vapor sorption (DVS) pattern substantially as set forth in Figure 14.
23. A crystalline form of(2R,5SJ 3aR)-8½xlroxy~7,9-diox^
2,3,4,5 J,9 3, 13a"Ocmhydro-2J5-methanopyrido[1^2i:4,5]pyrazlno[25l-b][ls31oxazepIne-10- earboxarrside, wherein the crystalline form is Form IV.
24. The crystalline form of claim 23, characterized by an x-ray powder diffraction (XRPD) pattern having peaks at out 6,2°, 163°, and 22.3° 2-Θ ± 0.2° 2-Θ.
25 , The crystalline form of claim 24, where!?} the x~ray powder diffraction (XRPD) pattern has further peaks at about 22.7° and 25 2-Θ ± 0,2° 2-Θ.
26. The crystalline form of claim 24 or 25, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 8.6" and 13.2° 2-0 ± 0,2° 2-8.
27. The crystalline form of any one of claims 24 to 26, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 18.7°, 20,0°, and 27.7° 2-Θ 0.2° 2-Θ.
28. The crystalline form of any one of claims 23 to 27, characterized by an x-ray powder diffraction (XRPD) pattern substantially as set forth in Figure .
29. A crystalline form of (2R,5S, 13aR)-8-hydroxy-7,9-dioxo~N-(2I4:,6-trlfiuorobenzyl)~ 23,4,5,7,9, i 3, 13a-octahydro«2s5-methanopyrido[ 1 ',2':4,5]pyrazmo[2, l-b][l,3]oxazepine-10- carboxamuk, wherein the crystalline form is Font). V.
30. A crystalline form of (2R,5S, 13aRV8-hydroxy-7,9-dioxo~^
Figure imgf000104_0001
carboxamides wherein the crystalline form is Form VI.
31. A crystalline form of (2R,5SS1 :¾R)~8-hydroxy-7,9 ioxo-N-(2,4s6-triiluoro'beQ2yl). 23,4,5,7,9, 13.1 a-oci?ihy^O-2.5--methauop rido ] ^2':4 5]p ra?jno[2, 1 -b][ 1 ,3]oxazepine- 1 G- carhoxam ide, wherein the crystalline form is Form VII,
32. A crystalline form of (2R,5S,13aR)-8-hydroxy-7,9-^^
2 A5s7,9,13,13a >ctahydro~2,5-me^^
carfaoxamide, wherein the crystalline form is Form VTiX33. A crystalline form of
(2¾SSJ3aR)-8-hydra^
2,5-methanopyrido[ ,2,;4551pyra irjo 2, 1 -b|[! ^Joxazspme-l 0-carboxamide oxalic- acid co- crystal.
33, The crystalline form of claim 32, characterized by an x-ra powder diffraction (XRPD) pattern having peaks at about 145°, 17/1°, and 19.1° 2-Θ * 0.2° 2-0.
34, The crystalline form of claim 34, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 21.8° and 26.5° 2-0 ± 0.2° 2-Θ.
35. The crystalline form of claim 33 or 34, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 7.6° and 9. 2-Θ ± 0,2° 2-8.
36, The crystalline .form of any one of claims 33 to 35, wherein the x-ray powder diffraction (XRPD) pattern has further peaks at about 11.6 29,7°> and 39.4° 2-Θ * 0.2° 2-Θ.
37. The crystalline form of any one of claims 32 to 36, characterized by an x-ray powder diffraction (XRPD) pattern substantial ly as set forth in Figure 6.
38, A pharmaceutical composition comprising a therapeutically effective amount of tbe forms of any one of claims 1 to 37 and a pharmaceutically acceptable carrier or excipient.
39, The pharmaceutical composition of claim 38, further comprising one to three additional therapeutic agents,
40. The pharmaceutical composition of claim 39, wherein the additional therapeutic agents are each anti-HIV drugs.
41. The pharmaceutical composition of claim 39 or claim 40, wherein the additional therapeutic agents are each independently selected from the group consisting of HIV protease inhibitors, HIV non-rt cleoside inhibitors of reverse transcriptase, HIV nucleoside i hibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, and other drags for treating HIV.
42. The pharmaceutical composition of any one of claims 39 to 41, wherein at least two of the additional therapeutic agents are each ΗΓΥ nucleotide or nucleoside inhibitors of reverse transcriptase.
43. The pharmaceutical composition of claim 38, further comprising tenofovir disoproxil furnarate and emtridtabine,
44. The pharmaceutical composition of claim 38, further comprising tenofovir aiafenamide and emtrkitahirjc,
45. The pharmaceutical composition of claim 38, further comprising tenofovir aiafe armde hemifumarate and emtridtabine.
46. The pharmaceutical composition of any one of claims 38 to 45, wherein the pharmaceutical composition is in a unit dosage form.
47. The pharmaceutical composition of claim 46, wherein the unit dosage form is a tablet.
48. A pharmaceutical composition prepared by combining a therapeutically effective amount of a form of any one of claims ί to 37 with a pharmaceutical ly acceptable carrier or exeipient.
49. Use of a form of any one of claims 1 to 37 for treating or prophylactically preventing an HIV infection.
50. Use of a form of any one of claims 1 to 37 for the manufacture of a medicament for treating or prophylactically preventing an HIV infection,
51. A form of any one of claims 1 to 37 for use in a method for treating or
prophylactically preventing an HIV infection.
52. A method for treating or prophylactically preventing an HIV infection in a human in need thereof, comprising administering to the human a. therapeutically effecti ve amount of a form of any one of claims 1 to 37.
PCT/US2015/036784 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide WO2015196137A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP19167626.1A EP3564244B1 (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
CA2950309A CA2950309C (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
NZ726732A NZ726732A (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1’,2’:4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide
EP15734504.2A EP3157931A1 (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
AU2015276881A AU2015276881B2 (en) 2014-06-20 2015-06-19 Crystalline forms of (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide
JP2016574182A JP6386104B2 (en) 2014-06-20 2015-06-19 (2R, 5S, 13AR) -8-hydroxy-7,9-dioxo-N- (2,4,6-trifluorobenzyl) -2,3,4,5,7,9,13,13A-octahydro- Crystal form of 2,5-methanopyrido [1 ′, 2 ′: 4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide
EP23195199.7A EP4309736A3 (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5]pyrazino [2,1-b][1,3]oxazepine-10-carboxamide
AU2018203737A AU2018203737A1 (en) 2014-06-20 2018-05-28 Crystalline forms of (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462015238P 2014-06-20 2014-06-20
US62/015,238 2014-06-20
US201462017183P 2014-06-25 2014-06-25
US62/017,183 2014-06-25

Publications (1)

Publication Number Publication Date
WO2015196137A1 true WO2015196137A1 (en) 2015-12-23

Family

ID=53514405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/036784 WO2015196137A1 (en) 2014-06-20 2015-06-19 Crystalline forms of (2r,5s,13ar)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido [1',2':4,5] pyrazino [2,1-b] [1,3] oxazepine-10-carboxamide

Country Status (12)

Country Link
US (4) US9682084B2 (en)
EP (3) EP3157931A1 (en)
JP (4) JP6386104B2 (en)
AU (2) AU2015276881B2 (en)
CA (1) CA2950309C (en)
ES (1) ES2962291T3 (en)
MA (1) MA40236A (en)
NZ (2) NZ726732A (en)
PL (1) PL3564244T3 (en)
TW (1) TW201613936A (en)
UY (1) UY36176A (en)
WO (1) WO2015196137A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017083304A1 (en) * 2015-11-09 2017-05-18 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US9732092B2 (en) 2012-12-21 2017-08-15 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydropyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]OXAZEPINES and methods for treating viral infections
WO2018064071A1 (en) * 2016-09-27 2018-04-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2019113462A1 (en) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2019207602A1 (en) * 2018-04-26 2019-10-31 Mylan Laboratories Limited Polymorphic forms of bictegravir and its sodium salt
WO2020003151A1 (en) * 2018-06-28 2020-01-02 Honour Lab Limited Process for the preparation of sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1', 2':4,5]pyrazino[2,1-b] [1,3] oxazepin-8-olate and its polymorphic form
EP3653629A1 (en) 2018-11-16 2020-05-20 Sandoz AG Acid addition salts of an integrase strand transfer inhibitor
WO2020161744A1 (en) 2019-02-07 2020-08-13 Cipla Limited Novel polymorphs of integrase inhibitor
CN111978333A (en) * 2019-09-30 2020-11-24 常州制药厂有限公司 Crystal form A of Bictegravir sodium salt, preparation method and application
WO2021233434A1 (en) * 2020-05-22 2021-11-25 上海迪赛诺生物医药有限公司 New crystal form of bictegravir sodium and preparation method therefor
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2023248240A1 (en) * 2022-06-21 2023-12-28 Mylan Laboratories Limited Polymorphic forms of bictegravir sodium

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NO2717902T3 (en) 2014-06-20 2018-06-23
TWI744723B (en) 2014-06-20 2021-11-01 美商基利科學股份有限公司 Synthesis of polycyclic-carbamoylpyridone compounds
TW201613936A (en) * 2014-06-20 2016-04-16 Gilead Sciences Inc Crystalline forms of(2R,5S,13aR)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
TWI794171B (en) 2016-05-11 2023-03-01 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-l1 inhibitors
TWI808055B (en) 2016-05-11 2023-07-11 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-1 inhibitors
DK3749673T3 (en) * 2018-02-09 2022-06-20 Sandoz Ag Crystalline form of bictegravir sodium
CN112996517A (en) * 2018-09-19 2021-06-18 吉利德科学公司 Integrase inhibitors for the prevention of HIV
WO2021236944A1 (en) 2020-05-21 2021-11-25 Gilead Sciences, Inc. Pharmaceutical compositions comprising bictegravir
US20240208995A1 (en) * 2021-04-19 2024-06-27 Honour Lab Limited Polymorphic forms of bictegravir potassium

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006116764A1 (en) * 2005-04-28 2006-11-02 Smithkline Beecham Corporation Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
WO2014100323A1 (en) * 2012-12-21 2014-06-26 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use

Family Cites Families (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE788516A (en) 1971-09-10 1973-03-07 Lonza Ag METHOD OF MANUFACTURING ALCOXYACETYLACETIC ESTERS
GB1528382A (en) 1974-12-26 1978-10-11 Teijin Ltd Cyclopentene diols and acyl esters thereof and processes for their preparation
DE2658401A1 (en) 1976-12-23 1978-07-06 Merck Patent Gmbh CYCLOPENTAN-1-AMINE, METHOD FOR THE PRODUCTION THEREOF AND AGENTS CONTAINING THESE COMPOUNDS
US4575694A (en) 1984-03-05 1986-03-11 Allied Corporation Coaxial connector
DE3900736A1 (en) 1989-01-12 1990-07-26 Hoechst Ag POSITIVELY WORKING RADIATION-SENSITIVE MIXTURE CONTAINING A MULTI-FUNCTIONAL (ALPHA) -DIAZO- (BETA) -KETOESTER, METHOD FOR THE PRODUCTION THEREOF AND RADIATION-SENSITIVE RECORDING MATERIAL COMPRISING THIS MIXTURE
US6703396B1 (en) 1990-02-01 2004-03-09 Emory University Method of resolution and antiviral activity of 1,3-oxathiolane nuclesoside enantiomers
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US5914331A (en) 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
DE4014649A1 (en) 1990-05-08 1991-11-14 Hoechst Ag NEW MULTIFUNCTIONAL CONNECTIONS WITH (ALPHA) -DIAZO-SS-KETOESTER AND SULPHONIC ACID UNIT UNITS, METHOD FOR THEIR PRODUCTION AND USE THEREOF
GB9301000D0 (en) 1993-01-20 1993-03-10 Glaxo Group Ltd Chemical compounds
US5922695A (en) 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
SE9702772D0 (en) 1997-07-22 1997-07-22 Astra Pharma Prod Novel compounds
US5935946A (en) 1997-07-25 1999-08-10 Gilead Sciences, Inc. Nucleotide analog composition and synthesis method
AU1403099A (en) 1997-11-14 1999-06-07 Merck & Co., Inc. Alpha-1a adrenergic receptor antagonists
JP2002529455A (en) 1998-11-09 2002-09-10 ジェームズ・ブラック・ファウンデーション・リミテッド Gastrin and cholecystokinin receptor ligands
GB2345058A (en) 1998-12-01 2000-06-28 Cerebrus Pharm Ltd Hydroxypyridone compounds useful in the treatment of oxidative damage to the central nervous system
DE69939749D1 (en) 1998-12-25 2008-11-27 Shionogi & Co AROMATIC HETEROCYCLES WITH HIV INTEGRASE INHIBITING PROPERTIES
AU2001262732A1 (en) 2000-06-14 2001-12-24 Shionogi And Co., Ltd. Inhibitor for enzyme having two divalent metal ions as active centers
CN101513402B (en) 2001-08-10 2012-03-21 盐野义制药株式会社 Antiviral agent
IL160642A0 (en) 2001-10-03 2004-07-25 Ucb Sa Pyrrolidinone derivatives
BRPI0213522C1 (en) 2001-10-26 2021-05-25 St Di Ricerche Di Biologia Molecolare P Angeletti S P A hydroxypyrimidinone derivative compounds, pharmaceutical composition, and use of a compound
US7109186B2 (en) 2002-07-09 2006-09-19 Bristol-Myers Squibb Company HIV integrase inhibitors
AU2003267098B2 (en) 2002-09-11 2008-11-20 Merck & Co., Inc. Dihydroxypyridopyrazine-1,6-dione compounds useful as HIV integrase inhibitors
EP4059923A1 (en) 2002-11-20 2022-09-21 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as hiv integrase inhibitor
US20040224917A1 (en) 2003-01-14 2004-11-11 Gilead Sciences, Inc. Compositions and methods for combination antiviral therapy
ATE490788T1 (en) 2003-04-25 2010-12-15 Gilead Sciences Inc ANTIVIRAL PHOSPHONATE ANALOGUE
TW200510425A (en) 2003-08-13 2005-03-16 Japan Tobacco Inc Nitrogen-containing fused ring compound and use thereof as HIV integrase inhibitor
TW200528440A (en) 2003-10-31 2005-09-01 Fujisawa Pharmaceutical Co 2-cyanopyrrolidinecarboxamide compound
AU2005211349A1 (en) 2004-01-30 2005-08-18 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. N-benzyl-3,4-dihyroxypyridine-2-carboxamide and N-benzyl-2,3-dihydroxypyridine-4-carboxamide compounds useful as HIV integrase inhibitors
CN101014574A (en) 2004-03-09 2007-08-08 默克公司 HIV integrase inhibitors
WO2005110399A2 (en) 2004-04-29 2005-11-24 The Regents Of The University Of California Zinc-binding groups for metalloprotein inhibitors
EP1755586A2 (en) 2004-04-29 2007-02-28 The Regents of the University of California Hydroxypyridinone, hydroxypyridinethione, pyrone, and thiopyrone metalloprotein inhibitors
WO2005110414A2 (en) 2004-05-07 2005-11-24 Merck & Co., Inc. Hiv integrase inhibitors
US7273859B2 (en) 2004-05-12 2007-09-25 Bristol-Myers Squibb Company HIV integrase inhibitors: cyclic pyrimidinone compounds
US7531554B2 (en) 2004-05-20 2009-05-12 Japan Tobacco Inc. 4-oxoquinoline compound and use thereof as HIV integrase inhibitor
MY134672A (en) 2004-05-20 2007-12-31 Japan Tobacco Inc Stable crystal of 4-oxoquinoline compound
EP2229945A1 (en) 2004-05-21 2010-09-22 Japan Tobacco, Inc. Combinations comprising a 4-isoquinolone derivative and anti-HIV agents
ES2720618T3 (en) 2004-07-27 2019-07-23 Gilead Sciences Inc Phosphonate analogs of HIV inhibitor compounds
EP1790638B1 (en) 2004-09-15 2013-04-03 Shionogi Co., Ltd. Carbamoylpyridone derivative having hiv integrase inhibitory activity
JP2006118669A (en) 2004-10-25 2006-05-11 Sanoh Industrial Co Ltd Resin tube
CA2634499A1 (en) 2004-12-23 2006-06-29 Virochem Pharma Inc. Hydroxydihydropyridopy razine-1,8-diones and methods for inhibiting hiv integrase
EP1852434B1 (en) 2005-02-21 2011-07-13 Shionogi Co., Ltd. Bicyclic carbamoylpyridone derivative having hiv integrase inhibiting activity
EP1888581A2 (en) 2005-05-16 2008-02-20 Gilead Sciences, Inc. Hiv-integrase inhibitor compounds
CA2616314A1 (en) 2005-07-27 2007-02-01 Gilead Sciences, Inc. Antiviral phosphonate conjugates for inhibition of hiv
CA2626956A1 (en) 2005-10-27 2007-05-03 Shionogi & Co., Ltd. Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
EP2308490A1 (en) 2005-12-30 2011-04-13 Gilead Sciences, Inc. Methods for improving the pharmacokinetics of hiv integrase inhibitors
US7601844B2 (en) 2006-01-27 2009-10-13 Bristol-Myers Squibb Company Piperidinyl derivatives as modulators of chemokine receptor activity
JP2009525261A (en) 2006-02-01 2009-07-09 日本たばこ産業株式会社 6- (3-Chloro-2-fluorobenzyl) -1-[(2S) -1-hydroxy-3-methylbutan-2-yl] -7-methoxy-4-oxo for the treatment of retroviral infections Use of -1,4-dihydroquinoline-3-carboxylic acid or a salt thereof
PL1992607T3 (en) 2006-03-06 2015-05-29 Japan Tobacco Inc Method for producing 4-oxoquinoline compound
CN101437801B (en) 2006-03-06 2013-02-06 日本烟草产业株式会社 Process for production of 4-oxoquinoline compound
US7893055B2 (en) 2006-06-28 2011-02-22 Bristol-Myers Squibb Company HIV integrase inhibitors
WO2008010953A2 (en) 2006-07-19 2008-01-24 University Of Georgia Research Foundation, Inc. Pyridinone diketo acids: inhibitors of hiv replication in combination therapy
MX2009002689A (en) 2006-09-12 2009-03-26 Gilead Sciences Inc Process and intermediates for preparing integrase inhibitors.
EP2084160A1 (en) 2006-10-18 2009-08-05 Merck & Co., Inc. Hiv integrase inhibitors
ES2603617T3 (en) 2007-02-23 2017-02-28 Gilead Sciences, Inc. Modulators of pharmacokinetic properties of therapeutics
US20080280945A1 (en) 2007-05-09 2008-11-13 Sachin Lohani Crystalline forms of an HIV integrase inhibitor
CN103480000A (en) 2007-06-29 2014-01-01 吉里德科学公司 Therapeutic compositions and the use thereof
WO2009006203A1 (en) 2007-06-29 2009-01-08 Gilead Sciences, Inc. Therapeutic compositions and the use thereof
WO2009018350A1 (en) 2007-07-31 2009-02-05 Limerick Biopharma, Inc. Pyrone analog compositions and methods
AR068403A1 (en) 2007-09-11 2009-11-18 Gilead Sciences Inc PROCESS AND INTERMEDIARIES FOR THE PREPARATION OF INTEGRASA INHIBITORS
EA201200631A1 (en) 2007-11-16 2012-11-30 Джилид Сайенсиз, Инк. Inhibitors of Human Immunodeficiency Virus Replication
GB0803019D0 (en) 2008-02-19 2008-03-26 Btg Int Ltd Fluorinated compounds
US8129398B2 (en) 2008-03-19 2012-03-06 Bristol-Myers Squibb Company HIV integrase inhibitors
US20100272811A1 (en) 2008-07-23 2010-10-28 Alkermes,Inc. Complex of trospium and pharmaceutical compositions thereof
EP2320909B8 (en) 2008-07-25 2016-03-30 VIIV Healthcare Company Chemical compounds
WO2010011819A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
WO2010011818A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
WO2010011816A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
WO2010011815A1 (en) 2008-07-25 2010-01-28 Smithkline Beecham Corporation Chemical compounds
PT2320908E (en) 2008-07-25 2014-03-06 Shionogi & Co Dolutegravir prodrugs
MX2011006241A (en) 2008-12-11 2011-06-28 Shionogi & Co Synthesis of carbamoylpyridone hiv integrase inhibitors and intermediates.
ES2964383T3 (en) 2008-12-11 2024-04-05 Viiv Healthcare Co Processes and intermediates for carbamoylpyridone HIV integrase inhibitors
TWI518084B (en) 2009-03-26 2016-01-21 鹽野義製藥股份有限公司 Process for pyrone and pyridone derivatives
US8835461B2 (en) 2009-03-26 2014-09-16 Shionogi & Co., Ltd. Substituted 3-hydroxy-4-pyridone derivative
US8338441B2 (en) 2009-05-15 2012-12-25 Gilead Sciences, Inc. Inhibitors of human immunodeficiency virus replication
PT2444400T (en) 2009-06-15 2018-06-06 Shionogi & Co Substituted polycyclic carbamoylpyridone derivative
KR101280198B1 (en) 2009-09-02 2013-06-28 이화여자대학교 산학협력단 pyrazole derivatives, preparation thereof and composition comprising the same for prevention and treatment of osteoporosis
ES2688925T3 (en) 2010-01-27 2018-11-07 Viiv Healthcare Company Antiviral treatment
WO2011105590A1 (en) 2010-02-26 2011-09-01 日本たばこ産業株式会社 1,3,4,8-tetrahydro-2h-pyrido[1,2-a]pyrazine derivative and use of same as hiv integrase inhibitor
TWI582097B (en) 2010-03-23 2017-05-11 Viiv醫療保健公司 Process for preparing carbamoylpyridone derivatives and intermediates
US20130165489A1 (en) 2010-05-03 2013-06-27 The Trustees Of The University Of Pennsylvania Small Molecule Modulators of HIV-1 Capsid Stability and Methods Thereof
CA2798735A1 (en) 2010-05-19 2011-11-24 Haruki Shibata Prophylactic and/or therapeutic agent for non-alcoholic steatohepatitis
JP5806735B2 (en) 2010-07-02 2015-11-10 ギリアード サイエンシーズ, インコーポレイテッド 2-Quinolinyl-acetic acid derivatives as HIV antiviral compounds
US9102614B2 (en) 2010-07-02 2015-08-11 Gilead Sciences, Inc. Naphth-2-ylacetic acid derivatives to treat AIDS
EP2595986A2 (en) 2010-07-14 2013-05-29 Addex Pharma SA Novel 2-amino-4-pyrazolyl-thiazole derivatives and their use as allosteric modulators of metabotropic glutamate receptors
CN106083891B (en) 2010-08-05 2018-03-23 盐野义制药株式会社 The manufacture method of compound with hiv integrase inhibitory activity
SI2620436T1 (en) 2010-09-24 2018-08-31 Shionogi & Co., Ltd. Substituted polycyclic carbamoyl pyridone derivative prodrug
WO2012106534A2 (en) 2011-02-02 2012-08-09 The Regents Of The University Of California Hiv integrase inhibitors
AP2015008931A0 (en) 2011-04-21 2015-12-31 Gilead Sciences Inc Benzothiazole compounds and their pharmaceutical use
US20140213553A1 (en) 2011-05-03 2014-07-31 Concert Pharmaceuticals Inc. Carbamoylpyridone derivatives
US9328075B2 (en) 2011-05-05 2016-05-03 St. Jude Children's Research Hospital Pyrimidinone compounds and methods for treating influenza
US9121496B2 (en) 2011-06-29 2015-09-01 Arvinmeritor Technology, Llc Drive axle system and a method of control
US9540343B2 (en) 2011-07-06 2017-01-10 Gilead Sciences, Inc. Compounds for the treatment of HIV
CN102863512B (en) 2011-07-07 2016-04-20 上海泓博智源医药技术有限公司 Antiviral compound
US9206197B2 (en) 2011-09-14 2015-12-08 Mapi Pharma Ltd. Amorphous form of dolutegravir
US9200009B2 (en) 2011-10-12 2015-12-01 Shionogi & Co., Ltd. Polycyclic pyridone derivative having integrase inhibitory activity
WO2013087581A1 (en) 2011-12-12 2013-06-20 Bayer Intellectual Property Gmbh Amino-substituted imidazopyridazines
WO2013091096A1 (en) 2011-12-20 2013-06-27 Boehringer Ingelheim International Gmbh Condensed triclyclic compounds as inhibitors of hiv replication
UY34750A (en) 2012-04-20 2013-11-29 Gilead Sciences Inc ? COMPOUNDS FOR HIV TREATMENT, COMPOSITIONS, PREPARATION METHODS, INTERMEDIARIES AND THERAPEUTIC METHODS ?.
WO2014008636A1 (en) 2012-07-11 2014-01-16 Merck Sharp & Dohme Corp. Macrocyclic compounds as hiv integrase inhibitors
WO2014014933A1 (en) 2012-07-20 2014-01-23 Merck Sharp & Dohme Corp. Hiv treatment with amido-substituted pyrimidinone derivatives
EP2877469A4 (en) 2012-07-25 2016-04-06 Merck Sharp & Dohme Substituted naphthyridinedione derivatives as hiv integrase inhibitors
US8877931B2 (en) 2012-08-03 2014-11-04 Gilead Sciences, Inc. Process and intermediates for preparing integrase inhibitors
WO2014074675A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company Heteroaryl substituted pyridyl compounds useful as kinase modulators
JP2016503030A (en) 2012-12-14 2016-02-01 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC Pharmaceutical composition
WO2014099586A1 (en) 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. 4-pyridinonetriazine derivatives as hiv integrase inhibitors
EP2934482A4 (en) 2012-12-21 2016-07-20 Merck Sharp & Dohme Gastro-retentive formulations
US20140221355A1 (en) 2012-12-21 2014-08-07 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
US20140221380A1 (en) 2012-12-27 2014-08-07 Japan Tobacco Inc. SUBSTITUTED SPIROPYRIDO[1,2-a]PYRAZINE DERIVATIVE AND PHARMACEUTICAL USE OF SAME AS HIV INTEGRASE INHIBITOR
EP3008044B1 (en) 2013-06-13 2018-11-21 Merck Sharp & Dohme Corp. Fused tricyclic heterocyclic compounds as hiv integrase inhibitors
WO2015039348A1 (en) 2013-09-23 2015-03-26 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds useful as hiv integrase inhibitors
EA030695B1 (en) 2013-09-27 2018-09-28 Мерк Шарп И Доум Корп. Substituted quinolizine derivatives useful as hiv integrase inhibitors
WO2015089847A1 (en) 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. Spirocyclic heterocycle compounds useful as hiv integrase inhibitors
EP3096763B1 (en) 2014-01-21 2019-12-25 Laurus Labs Limited Novel process for the preparation of dolutegravir and pharmaceutically acceptable salts thereof
WO2015138933A1 (en) 2014-03-13 2015-09-17 Assia Chemical Industries Ltd. Solid state forms of dolutegravir sodium
TW201613936A (en) * 2014-06-20 2016-04-16 Gilead Sciences Inc Crystalline forms of(2R,5S,13aR)-8-hydroxy-7,9-dioxo-n-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
NO2717902T3 (en) 2014-06-20 2018-06-23
TWI744723B (en) 2014-06-20 2021-11-01 美商基利科學股份有限公司 Synthesis of polycyclic-carbamoylpyridone compounds
TWI737647B (en) 2015-11-09 2021-09-01 美商基利科學股份有限公司 Therapeutic compositions for treatment of human immunodeficiency virus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006116764A1 (en) * 2005-04-28 2006-11-02 Smithkline Beecham Corporation Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
WO2014100323A1 (en) * 2012-12-21 2014-06-26 Gilead Sciences, Inc. Polycyclic-carbamoylpyridone compounds and their pharmaceutical use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CAIRA: "Crystalline Polymorphism of Organic Compounds", TOPICS IN CURRENT CHEMISTRY, SPRINGER, BERLIN, DE, vol. 198, January 1998 (1998-01-01), pages 163 - 208, XP008166276, ISSN: 0340-1022 *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10689399B2 (en) 2012-12-21 2020-06-23 Gilead Sciences, Inc. Substituted 3,4,5,6,8,10,14,14a-octahydro-2h-2,6-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazocines and methods for treating viral infections
US9732092B2 (en) 2012-12-21 2017-08-15 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydropyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]OXAZEPINES and methods for treating viral infections
US10035809B2 (en) 2012-12-21 2018-07-31 Gilead Sciences, Inc. Substituted 2,3,4,5,7,9,13,13a-octahydro-1,5-methanopyrido[1′,2′:4,5]pyrazino[1,2-a][1,3]diazepines and methods for treating viral infections
US11548901B2 (en) 2012-12-21 2023-01-10 Gilead Sciences, Inc. Substituted 1,4-methanopyrido[1′,2′:4,5]pyrazino[1,2-a]pyrimidines for treating viral infections
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
AU2016354007C1 (en) * 2015-11-09 2020-09-10 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2017083304A1 (en) * 2015-11-09 2017-05-18 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2016354007B2 (en) * 2015-11-09 2019-11-14 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2020200995B9 (en) * 2015-11-09 2022-04-28 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US10548846B2 (en) 2015-11-09 2020-02-04 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
EP3632415A1 (en) * 2015-11-09 2020-04-08 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2020200995B2 (en) * 2015-11-09 2022-04-07 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
EP3346995B1 (en) 2015-11-09 2019-08-28 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
US11744802B2 (en) 2015-11-09 2023-09-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
AU2016354007C9 (en) * 2015-11-09 2020-10-01 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2018064071A1 (en) * 2016-09-27 2018-04-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
WO2019113462A1 (en) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2019207602A1 (en) * 2018-04-26 2019-10-31 Mylan Laboratories Limited Polymorphic forms of bictegravir and its sodium salt
US11623933B2 (en) 2018-06-28 2023-04-11 Honour Lab Limited Process for the preparation of sodium (2R,5S,13AR)-7,9-dioxo-10-((2,4,6-trifluorobenzyl) carbamoyl)-2,3,4,5,7,9,13,13A-octahydro-2,5-Methanopyrido[1 ′,2′:4,5]pyrazino[2,1 -b] [1,3] oxazepin-8-olate and its polymorphic form
WO2020003151A1 (en) * 2018-06-28 2020-01-02 Honour Lab Limited Process for the preparation of sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1', 2':4,5]pyrazino[2,1-b] [1,3] oxazepin-8-olate and its polymorphic form
EP3653629A1 (en) 2018-11-16 2020-05-20 Sandoz AG Acid addition salts of an integrase strand transfer inhibitor
WO2020161744A1 (en) 2019-02-07 2020-08-13 Cipla Limited Novel polymorphs of integrase inhibitor
CN111978333A (en) * 2019-09-30 2020-11-24 常州制药厂有限公司 Crystal form A of Bictegravir sodium salt, preparation method and application
CN113698420A (en) * 2020-05-22 2021-11-26 上海迪赛诺生物医药有限公司 Novel crystal form of bicalutavir sodium and preparation method thereof
WO2021233434A1 (en) * 2020-05-22 2021-11-25 上海迪赛诺生物医药有限公司 New crystal form of bictegravir sodium and preparation method therefor
WO2023248240A1 (en) * 2022-06-21 2023-12-28 Mylan Laboratories Limited Polymorphic forms of bictegravir sodium

Also Published As

Publication number Publication date
MA40236A (en) 2017-04-26
UY36176A (en) 2016-01-08
CA2950309C (en) 2019-04-30
AU2015276881A1 (en) 2016-12-15
US11202780B2 (en) 2021-12-21
EP4309736A3 (en) 2024-05-08
AU2018203737A1 (en) 2018-06-21
US10098886B2 (en) 2018-10-16
US20200390775A1 (en) 2020-12-17
AU2015276881B2 (en) 2018-06-07
JP6386104B2 (en) 2018-09-05
CA2950309A1 (en) 2015-12-23
US20150366872A1 (en) 2015-12-24
US9682084B2 (en) 2017-06-20
EP3564244A1 (en) 2019-11-06
JP2017518357A (en) 2017-07-06
JP2022153400A (en) 2022-10-12
JP6606692B2 (en) 2019-11-20
EP3157931A1 (en) 2017-04-26
JP2018199687A (en) 2018-12-20
TW201613936A (en) 2016-04-16
EP4309736A2 (en) 2024-01-24
EP3564244C0 (en) 2023-09-13
JP2020023509A (en) 2020-02-13
US20170333438A1 (en) 2017-11-23
PL3564244T3 (en) 2024-04-08
ES2962291T3 (en) 2024-03-18
EP3564244B1 (en) 2023-09-13
NZ726732A (en) 2018-05-25
US20190015420A1 (en) 2019-01-17
NZ736644A (en) 2023-06-30

Similar Documents

Publication Publication Date Title
US11202780B2 (en) Crystalline forms of (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1′,2′:4,5]pyrazino[2,1-b][1,3]oxazepine-10-carboxamide
AU2018203175B2 (en) Sodium (2r,5s,13ar)-7,9-dioxo-10-((2,4,6-trifluorobenzyl)carbamoyl)-2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[1&#39;,2&#39;:4,5]pyrazino[2,1-b][1,3]oxazepin-8-olate
AU2018317833B2 (en) Solid forms of an HIV capsid inhibitor
KR102497604B1 (en) Choline salt forms of an hiv capsid inhibitor

Legal Events

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

Ref document number: 15734504

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2950309

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015276881

Country of ref document: AU

Date of ref document: 20150619

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016574182

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015734504

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015734504

Country of ref document: EP