WO2004078163A2 - Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen - Google Patents

Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen Download PDF

Info

Publication number
WO2004078163A2
WO2004078163A2 PCT/US2004/006288 US2004006288W WO2004078163A2 WO 2004078163 A2 WO2004078163 A2 WO 2004078163A2 US 2004006288 W US2004006288 W US 2004006288W WO 2004078163 A2 WO2004078163 A2 WO 2004078163A2
Authority
WO
WIPO (PCT)
Prior art keywords
crystal
acid
degrees
ray diffraction
diffraction pattern
Prior art date
Application number
PCT/US2004/006288
Other languages
French (fr)
Other versions
WO2004078163A3 (en
Inventor
Örn ALMARSSON
Magali Bourghol Hickey
Matthew Peterson
Michael J. Zaworotko
Brian Moulton
Nair Rodriguez-Hornedo
Original Assignee
Transform Pharmaceuticals, Inc.
University Of South Florida
The Regents Of The University Of Michigan
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
Priority claimed from PCT/US2003/006662 external-priority patent/WO2003074474A2/en
Priority claimed from US10/449,307 external-priority patent/US7078526B2/en
Priority claimed from US10/601,092 external-priority patent/US20050025791A1/en
Priority claimed from PCT/US2003/027772 external-priority patent/WO2004078161A1/en
Priority claimed from US10/660,202 external-priority patent/US7927613B2/en
Priority claimed from PCT/US2003/041273 external-priority patent/WO2004061433A1/en
Application filed by Transform Pharmaceuticals, Inc., University Of South Florida, The Regents Of The University Of Michigan filed Critical Transform Pharmaceuticals, Inc.
Priority to US10/546,963 priority Critical patent/US20070059356A1/en
Priority to CA002514733A priority patent/CA2514733A1/en
Priority to JP2006508979A priority patent/JP2007524596A/en
Priority to EP04715190A priority patent/EP1631260A2/en
Priority to PCT/US2004/009947 priority patent/WO2004089313A2/en
Priority to US10/551,014 priority patent/US20060223794A1/en
Priority to US10/926,842 priority patent/US7446107B2/en
Priority to PCT/US2004/028456 priority patent/WO2005092884A1/en
Priority to EP04782868A priority patent/EP1718640A4/en
Priority to JP2007500742A priority patent/JP2007525502A/en
Priority to JP2006525508A priority patent/JP4842819B2/en
Priority to EP20040783308 priority patent/EP1670753A4/en
Publication of WO2004078163A2 publication Critical patent/WO2004078163A2/en
Publication of WO2004078163A3 publication Critical patent/WO2004078163A3/en
Priority to IL173575A priority patent/IL173575A0/en
Priority to NO20060669A priority patent/NO20060669L/en
Priority to US12/234,420 priority patent/US20090088443A1/en
Priority to IL199140A priority patent/IL199140A/en
Priority to US12/792,415 priority patent/US20100311701A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1617Organic compounds, e.g. phospholipids, fats
    • A61K9/1623Sugars or sugar alcohols, e.g. lactose; Derivatives thereof; Homeopathic globules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/145Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic compounds

Definitions

  • the present mvention relates to co-crystal API-containing compositions, pharmaceutical compositions comprising such APIs, and methods for preparing the same.
  • APIs Active pharmaceutical ingredients (API or APIs (plural)) in pharmaceutical compositions can be prepared in a variety of different forms. Such APIs can be prepared so as to have a variety of different chemical forms including chemical derivatives or salts. Such APIs can also be prepared to have different physical forms. For example, the APIs may be amorphous, may have different crystalline polymorphs, or may exist in different solvation or hydration states. By varying the form of an API, it is possible to vary the physical properties thereof. For example, crystalline polymorphs typically have different solubilities from one another, such that a more thermodynamically stable polymorph is less soluble than a less thermodynamically stable polymorph. Pharmaceutical polymorphs can also differ in properties such as shelf-life, bioavailability, morphology, vapour pressure, density, colour, and compressibility. Accordingly, variation of the crystalline state of an API is one of many ways in which to modulate the physical properties thereof.
  • APIs that have improved properties, in particular, as oral formulations. Specifically, it is desirable to identify improved forms of APIs that exhibit significantly improved properties including increased aqueous solubility and stability. Further, it is desirable to improve the processability, or preparation of pharmaceutical formulations. For example, needle-like crystal forms or habits of APIs can cause aggregation, even in compositions where the API is mixed with other substances, such that a non- uniform mixture is obtained. It is also desirable to increase or decrease the dissolution rate of API-containing pharmaceutical compositions in water, increase or decrease the bioavailability of orally-administered compositions, and provide a more rapid or more delayed onset to therapeutic effect.
  • APIs can be obtained which improve the properties of APIs as compared to such APIs in a non-co-crystalline slate (free acid, free base, zwitter ions, salts, etc.).
  • the present invention provides a co-crystal pharmaceutical composition
  • a co-crystal pharmaceutical composition comprising an API compound and a co-crystal former, such that the API and co- crystal former are capable of co-crystallizing from a solid or solution phase under crystallization conditions.
  • Another aspect of the present invention provides a process for the production of a pharmaceutical composition, which process comprises:
  • an API which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
  • a co-crystal former which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
  • a further aspect of the present invention provides a process for the production of a pharmaceutical composition, which comprises:
  • the present invention provides a process for the production of a pharmaceutical composition, which comprises:
  • the present invention provides a process for modulating the solubility of an API, which process comprises:
  • the present invention provides a process for modulating the dissolution of an API, whereby the aqueous dissolution rate or the dissolution rate in simulated gastric fluid or in simulated intestinal fluid, or in a solvent or plurality of solvents is increased or decreased, which process comprises:
  • the present invention provides a process for modulating the bioavailability of an API, whereby the AUC is increased, the time to T max is reduced, the length of time the concentration of the API is above ' _ T max is increased, or C max is increased, which process comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
  • the present invention provides a process for improving the linearity of a dose response of an API, which process comprises:
  • the present invention provides a process for improving the stability of a pharmaceutical salt, which process comprises:
  • the present invention provides a process for making co-crystals of difficult to salt or unsaltable APIs, which process comprises:
  • the present invention provides a method for decreasing the hygroscopicity of an API, which method comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
  • the present invention provides a process for crystallizing an amorphous compound, which process comprises:
  • the present invention provides a process for reducing the form diversity of an API, which process comprises:
  • the present invention provides a process for modifying the morphology of an API, which process comprises:
  • the present invention provides a co-crystal composition
  • a co-crystal composition comprising a co-crystal, wherein said co-crystal comprises an API compound and a co-crystal former.
  • the co-crystal has an improved property as compared to the free form (including a free acid, free base, zwitter ion, hydrate, solvate, etc.) or a salt (which includes salt hydrates and solvates).
  • the improved property is selected from the group consisting of: increased solubility, increased dissolution, increased bioavailability, increased dose response, decreased hygroscopicity, a crystalline form of a normally amorphous compound, a crystalline form of a difficult to salt or unsaltable compound, decreased form diversity, more desired morphology, or other property described herein.
  • Figs. I A-B PXRD diffractograms of a co-crystal comprising celecoxib and nicotinamide, with the background removed and as collected, respectively.
  • Fig. 2 DSC thermogram for a co-crystal comprising celecoxib and nicotinamide.
  • Fig. 3 TGA thermogram for a co-crystal comprising celecoxib and nicotinamide.
  • Fig. 4 Raman spectrum for a co-crystal comprising celecoxib and nicotinamide.
  • Figs. 5A-B PXRD diffractograms of a co-crystal comprising celecoxib and 18-crown-6, with the background removed and as collected, respectively.
  • Fig. 6 DSC thermogram for a co-crystal comprising celecoxib and 18-crown-6.
  • Fig. 7 TGA thermogram for a co-crystal comprising celecoxib and 18-crown-6.
  • Figs. 8A-B PXRD diffractograms of a co-crystal comprising topiramate and 18-crown-6, with the background removed and as collected, respectively.
  • Fig. 9 DSC thermogram for a co-crystal comprising topiramate and 18-crown-6.
  • Figs. 10A-B PXRD diffractograms of a co-crystal comprising olanzapine and nicotinamide (Form
  • Fig. 11 DSC thermogram for a co-crystal comprising olanzapine and nicotinamide (Form I).
  • Fig. 12 PXRD diffractogram of a co-crystal comprising olanzapine and nicotinamide (Form II).
  • Figs. 13A-B PXRD diffractograms of a co-crystal comprising olanzapine and nicotinamide (Form
  • Figs. 14A-D Packing diagrams and crystal structure of a co-crystal comprising olanzapine and nicotinamide (Form III).
  • Fig. 15 PXRD diffractogram of a co-crystal comprising c ⁇ -itraconazole and succinic acid.
  • Fig. 16 DSC thermogram for a co-crystal comprising c ⁇ -itraconazole and succinic acid.
  • Fig. 17 PXRD diffractogram of a co-crystal comprising c/s-itraconazole and fumaric acid.
  • Fig. 18 DSC thermogram for a co-crystal comprising CM-itraconazole and fumaric acid.
  • Fig. 19 PXRD diffractogram of a co-crystal comprising czAitraconazole and L-tartaric acid.
  • Fig. 20 DSC thermogram for a co-crystal comprising c ⁇ -itraconazole and L-tartaric acid.
  • Fig. 21 PXRD diffractogram of a co-crystal comprising cw-itraconazole and L-malic acid.
  • Fig. 22 DSC thermogram for a co-crystal comprising c/ ' s-itraconazole and L-malic acid.
  • Fig. 23 PXRD diffractogram of a co-crystal comprising cw-itraconazoleHCl and DL-tartaric acid.
  • Fig 24 DSC thermogram for a co-crystal comprising c ⁇ -itraconazoleHCl and DL-tartaric acid.
  • Fig. 26 DSC thermogram for a co-crystal comprising modafinil and malonic acid (Form I).
  • Fig. 27 Raman spectrum for a co-crystal comprising modafinil and malonic acid (Form I).
  • Fig. 28 PXRD diffractogram of a co-crystal comprising modafinil and malonic acid (Form II).
  • Figs. 29A-B PXRD diffractograms of a co-crystal comprising modafinil and glycolic acid, with the background removed and as collected, respectively.
  • Figs. 30A-B PXRD diffractograms of a co-crystal comprising modafinil and maleic acid, with the background removed and as collected, respectively.
  • Figs. 31 A-B PXRD diffractograms of a co-crystal comprising 5-fluorouracil and urea, with the background removed and as collected, respectively.
  • Fig. 32 DSC thermogram for a co-crystal comprising 5-ftuorouracil and urea.
  • Fig. 33 TGA thermogram for a co-crystal comprising 5-fluorouracil and urea.
  • Fig. 34 Raman spectrum for a co-crystal comprising 5-fluorouracil and urea.
  • Figs. 35 A-B PXRD diffractograms of a co-crystal comprising hydrochlorothiazide and nicotinic acid, with the background removed and as collected, respectively.
  • Figs. 37A-B PXRD diffractograms of a co-crystal comprising hydrochlorothiazide and piperazine, with the background removed and as collected, respectively.
  • Figs. 38A-B An acetaminophen 1-D polymeric chain and a co-crystal of acetaminophen and 4,4'- bipyridine, respectively.
  • Figures 40C and 40D show the supramolecular entity containing the synthon and corresponding co-crystal of aspirin and 4,4'-bipyridine, respectively.
  • FIGs. 41A-D Pure ibuprofen and the corresponding crystal structure are shown in Figures 41A and 4 IB, respectively.
  • Figures 41C and 41D show the supramolecular entity containing the synthon and corresponding co-crystal of ibuprofen and 4,4'-bipyridine, respectively.
  • FIGs. 2A-D Pure flurbiprofen and the corresponding crystal structure are shown in Figures 42A and 2B, respectively.
  • Figures 42C and 42D show the supramolecular synthon and corresponding co-crystal of flurbiprofen and 4,4'-bipyridine, respectively.
  • Figs. 43A-B The supramolecular entity containing the synthon and the corresponding co-crystal structure of flurbiprofen and trans- l,2-bis(4-pyridyl)ethylene, respectively.
  • Figs. 44A-B The crystal structure of pure carbamazepine and the co-crystal structure of carbamazepine and -phthalaldehyde, respectively.
  • Fig. 45 A packing diagram of the co-crystal structure of carbamazepine and nicotinamide.
  • Fig. 46 PXRD diffractogram of a co-crystal comprising carbamazepine and nicotinamide.
  • Fig. 47 DSC thermogram for a co-crystal comprising carbamazepine and nicotinamide.
  • Fig. 48 A packing diagram of the co-crystal structure of carbamazepine and saccharin.
  • Fig. 49 PXRD diffractogram of a co-crystal comprising carbamazepine and saccharin.
  • Fig. 50 DSC thermogram for a co-crystal comprising carbamazepine and saccharin.
  • Figs. 51 A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 2,6-pyridinedicarboxylic acid, respectively.
  • Figs. 52A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 5-nitroisophthalic acid, respectively.
  • Figs. 53 A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 1,3,5,7-adamantanetetracarboxylic acid, respectively.
  • Figs. 54A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepme and benzoquinone, respectively.
  • Figs. 55A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and trimesic acid, respectively.
  • Fig. 56 PXRD diffractogram of a co-crystal comprising carbamazepine and trimesic acid.
  • Fig. 57 Dissolution profile for a co-crystal of celecoxib :nicotinamide vs. celecoxib free acid.
  • Fig. 58 Dissolution profile for co-crystals of itraconazole: succinic acid, itraconazle:tartaric acid and itraconazole:malic acid vs. itraconazole free base.
  • Fig. 61 Dissolution profile of several formulations of modafinil free form and modafinikmalonic acid (Form I).
  • co-crystal as used herein means a crystalline material comprised of two or more unique solids at room temperature, each containing distinctive physical characteristics, such as structure, melting point and heats of fusion, with the exception that, if specifically stated, the API may be a liquid at room temperature.
  • the co-crystals of the present invention comprise a co-crystal former H-bonded to an API.
  • the co- crystal former may be H-bonded directly to the API or may be H-bonded to an additional molecule which is bound to the API.
  • the additional molecule may be H-bonded to the API or bound ionically or covalently to the API.
  • the additional molecule could also be a different API.
  • Solvates of API compounds that do not further comprise a co-crystal former are not co-crystals according to the present invention.
  • the co-crystals may however, include one or more solvate molecules in the crystalline lattice. That is, solvates of co-crystals, or a co-crystal further comprising a solvent or compound that is a liquid at room temperature, is included in the present invention, but crystalline material comprised of only one solid and one or more liquids (at room temperature) are not included in the present invention, with the previously noted exception of specifically stated liquid APIs.
  • the co-crystals may also be a co-crystal between a co-crystal former and a salt of an API, but the API and the co-crystal former of the present invention are constructed or bonded together through hydrogen bonds.
  • Other modes of molecular recognition may also be present including, pi-stacking, guest-host complexation and van der Waals interactions.
  • hydrogen-bonding is the dominant interaction in the formation of the co-crystal, (and a required interaction according to the present invention) whereby a non-covalent bond is formed between a hydrogen bond donor of one of the moieties and a hydrogen bond acceptor of the other. Hydrogen bonding can result in several different intermolecular configurations.
  • hydrogen bonds can result in the formation of dimers, linear chains, or cyclic structures. These configurations can further include extended (two-dimensional) hydrogen bond networks and isolated triads (Fig. 60).
  • An alternative embodiment provides for a co-crystal wherein the co-crystal former is a second API. In another embodiment, the co-crystal former is not an API. In another embodiment the co-crystal comprises two co-crystal formers.
  • the chemical and physical properties of an API in the form of a co-crystal may be compared to a reference compound that is the same API in a different form.
  • the reference compound may be specified as a free form, or more specifically, a free acid, free base, or zwitterion; a salt, or more specifically for example, an inorganic base addition salt such as sodium, potassium, lithium, calcium, magnesium, ammonium, aluminum salts or organic base addition salts, or an inorganic acid addition salts such as HBr, HCl, sulfuric, nitric, or phosphoric acid addition salts or an organic acid addition salt such as acetic, propionic, pyruvic, malanic, succinic, malic, maleic, fumaric, tartaric, citric, benzoic, methanesulfonic, ethanesulforic, stearic or lactic acid addition salt; an anhydrate or hydrate of a free form or salt, or more specifically, for example, a hemihydrate, monohydrate, dihydrate, trihydrate, quadrahydrate, pentahydrate, sesquihydrate; or a solvate of a free form or salt.
  • the reference compound for an API in salt form co-crystallized with a co-crystal former can be the API salt form.
  • the reference compound for a free acid API co-crystallized with a co-crystal former can be the free acid API.
  • the reference compound may also be specified as crystalline or amorphous.
  • the co-crystals can include an acid addition salt or base addition salt of an API.
  • Acid addition salts include, but are not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid, and organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, madelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2- hydroxyethanesulfonic acid, benzenesulfonic acid,/?-chlorobenzenes
  • Base addition salts include, but are not limited to, inorganic bases such as sodium, potassium, lithium, ammonium, calcium and magnesium salts, and organic bases such as primary, secondary and tertiary amines (e.g. isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, and N-ethylpiperidine).
  • inorganic bases such as sodium, potassium, lithium, ammonium, calcium and magnesium salts
  • organic bases such as primary, secondary and tertiary amines (e.g. isopropylamine,
  • the ratio of API to co-crystal former may be stoichiometric or non-stoichiometric according to the present invention. For example, 1 :1, 1.5:1, 1 :1.5, 2:1 and 1 :2 ratios of API:co-crystal former are acceptable.
  • a co-crystal form of an API is particularly advantageous where the original API is insoluble or sparingly soluble in water.
  • the co-crystal properties conferred upon the API are also useful because the bioavailability of the API can be improved and the plasma concentration and/or serum concentration of the API can be improved. This is particularly advantageous for orally-administrable formulations.
  • the dose response of the API can be improved, for example by increasing the maximum attainable response and/or increasing the potency of the API by increasing the biological activity per dosing equivalent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a co-crystal of an API and a co-crystal former, such that the API and co-crystal former are capable of co-crystallizing from a solution phase under crystallization conditions or from the solid-state, for example, through grinding, heating, or through vapor transfer (e.g., co-sublimation).
  • the API has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine and a co-crystal former which has at least one functional group selected from ether, thioether,
  • co-crystals of the present invention are formed where the API and co-crystal former are bonded together through hydrogen bonds.
  • Other non-covalent interactions including pi-stacking and van der Waals interactions, may also be present.
  • the co-crystal former is selected from the co-crystal formers of Table I and Table II. In other embodiments, the co-crystal former of Table I is specified as a Class 1, Class 2, or Class 3 co-crystal former (see column labeled "class" Table I).
  • the difference in pK a value of the co-crystal former and the API is less than 2. In other embodiments, the difference in pK a values of the co- crystal former and API is less than 3, less than 4, less than 5, between 2 and 3, between 3 and 4, or between 4 and 5. Table I lists multiple pK a values for co-crystal formers having multiple functionalities. It is readily apparent to one skilled in the art the particular functional group corresponding to a particular pK a value.
  • the particular functional group of a co-crystal former interacting with the API is specified (see for example Table I, columns labeled “Functionality” and “Molecular Structure” and the column of Table II labeled "Co- Crystal Former Functional Group”).
  • the functional group of the API interacting with the co-crystal former functional group is specified (see, for example, Tables II and III).
  • the co-crystal comprises more than one co-crystal former.
  • two, three, four, five, or more co-crystal formers can be incorporated in a co-crystal with an API.
  • Co-crystals which comprise two or more co-crystal formers and an API are bound together via hydrogen bonds.
  • incorporated co- crystal formers are hydrogen bonded to the API molecules.
  • co- crystal formers are hydrogen bonded to either the API molecules or the incorporated co- crystal formers.
  • co-crystal formers can be contained in a single compartment, or kit, for ease in screening an API for potential co-crystal species.
  • the co-crystal kit can comprise 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or more of the co-crystal formers in Tables I and II.
  • the co-crystal formers are in solid form or in solution and in an array of individual reaction vials such that individual co-crystal formers can be tested with one or more APIs by one or more crystallization methods or multiple co-crystal formers can be easily tested against one or more compounds by one or more crystallization methods.
  • the crystallization methods include, but are not limited to, melt recrystallization, grinding, milling, standing, co-crystal formation from solution by evaporation, thermally driven crystallization from solution, co-crystal formation from solution by addition of anti-solvent, co-crystal formation from solution by vapor- diffusion, co-crystal formation from solution by drown-out, co-crystal formation from solution by any combination of the above mentioned techniques, co-crystal formation by co-sublimation, co-crystal formation by sublimation using a Knudsen cell apparatus, co- crystal formation by standing the desired components of the co-crystal in the presence of solvent vapor, co-crystal formation by slurry conversion of the desired components of the co-crystal in a solvent or mixtures of solvents, or co-crystal formation by any combination of the above techniques in the presence of additives, nucleates, crystallization enhancers, precipitants, chemical stabilizers, or anti-oxidants.
  • kits can be used alone or as part of larger crystallization experiments.
  • kits can be constructed as single co-crystal former single well kits, single co- crystal former multi-well kits, multi-co-crystal former single well kits, or multi-co-crystal former multi-well kits.
  • High-throughput crystallization e.g., the CrystalMaxTM platform
  • Multi-well plates e.g., 96 wells, 384 wells, 1536 wells, etc.
  • the API is selected from an API of Table IV or elsewhere herein.
  • co-crystals can comprise such APIs in free form (i.e. free acid, free base, zwitter ion), salts, solvates, hydrates, or the like.
  • the API can either be of the form listed in Table IV or its corresponding free form, or of another form that is not listed.
  • Table IV includes the CAS number, chemical name, or a PCT or patent reference (each incorporated herein in their entireties).
  • the functional group of the particular API interacting with the co-crystal former is specified.
  • a specific functional group of a co-crystal former, a specific co-crystal former, or a specified functional group or a specific co-crystal former interacting with the particular API may also be specified. It is noted that for Table II, the co-crystal former, and optionally the specific functionality, and each of the listed corresponding interacting groups are included as individual species of the present invention. Thus, each specific combination of a co-crystal former and one of the interacting groups in the same row may be specified as a species of the present invention. The same is true for other combinations as discussed in the Tables and elsewhere herein.
  • the co-crystal comprises an API wherein the API forms a dimeric primary amide structure via hydrogen bonds with an R 2 2 (8) motif.
  • the dimeric primary amide structure further comprises one, two, three, or four hydrogen bond donors.
  • the dimeric primary amide structure further comprises one or two hydrogen bond acceptors.
  • the dimeric primary amide structure further comprises a combination of hydrogen bond donors and acceptors.
  • the dimeric primary amide structure can further comprise one hydrogen bond donor and one hydrogen bond acceptor, one hydrogen bond donor and two hydrogen bond acceptors, two hydrogen bond donors and one hydrogen bond acceptor, two hydrogen bond donors and two hydrogen bond acceptors, or three hydrogen bond donors and one hydrogen bond acceptor.
  • Two non- limiting examples of APIs which form a dimeric primary amide co-crystal structure include modafinil and carbamazepine.
  • APIs which include a primary amide functional group include, but are not limited to, arotinolol, atenolol, carpipramine, cefotetan, cefsulodin, docapromine, darifenacin, exalamide, fidarestat, frovatriptan, silodosin, levetiracetam, MEN- 10700, mizoribine, oxiracetam, piracetam, protirelin, TRH, ribavirm, valrecemide, temozolomide, tiazofurin, antiPARP-2, levovirin, N- benzyloxycarbonyl glycinamide, and UCB-34714.
  • each process according to the invention there is a need to contact the API with the co-crystal former. This may involve grinding or milling the two solids together or melting one or both components and allowing them to recrystallize.
  • the use of a granulating liquid may improve or may impede co-crystal formation.
  • Non-limiting examples of tools useful for the formation of co-crystals may include, for example, an extruder or a mortar and pestle.
  • contacting the API with the co-crystal former may also involve either solubilizing the API and adding the co-crystal former, or solubilizing the co-crystal former and adding the API. Crystallization conditions are applied to the API and co-crystal former.
  • This may entail altering a property of the solution, such as pH or temperature and may require concentration of the solute, usually by removal of the solvent, typically by drying the solution. Solvent removal results in the concentration of both API and co-crystal former increasing over time so as to facilitate crystallization. For example, evaporation, cooling, co-sublimation, or the addition of an antisolvent may be used to crystallize co-crystals. In another embodiment, a slurry comprising an API and a co-crystal former is used to form co-crystals. Once the solid phase comprising any crystals is formed, this may be tested as described herein.
  • co-crystals on a large and/or commercial scale may be successfully completed using one or more of the processes and techniques described herein.
  • crystallization of co-crystals from a solvent and grinding or milling are conceivable non-limiting processes.
  • co-crystals with stoichiometries of 1:1, 2:1, or 1 :2 can be produced by adding co-crystal former in an amount that is 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 50, 75, 100 times or more than the stoichiometric amount for a given co- crystal.
  • Such an excessive use of a co-crystal former to form a co-crystal can be employed in solution or when grinding an API and a co-crystal former to drive co-crystal formation.
  • the present invention provides for the use of an ionic liquid as a medium for the formation of a co-crystal, and can also be used to crystallize other forms in addition to co-crystals (e.g., salts, solvates, free acid, free base, zwitterions, etc.).
  • This medium is useful, for example, where the above methods do not work or are difficult or impossible to control.
  • ionic liquids useful in co-crystal formation are: l-butyl-3-methylimidazolium lactate, 1-ethyl- 3-methylimidazolium lactate, and 1-butylpyridinium hexafluorophosphate.
  • compositions in general are discussed in further detail below and may further comprise a pharmaceutically-acceptable diluent, excipient or carrier.
  • the present invention provides a process for the production of a pharmaceutical composition, which process comprises:
  • an API which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine or of Table II or III;
  • a co-crystal former which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O- heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine or of Table I, II, or III; (3) grinding, heating or contacting in solution the
  • the present invention provides a process for the production of a pharmaceutical composition, which comprises:
  • Assaying the solid phase for the presence of co-crystals of the API and the co- crystal former may be carried out by conventional methods known in the art. For example, it is convenient and routine to use powder X-ray diffraction techniques to assess the presence of co-crystals. This may be affected by comparing the spectra of the API, the crystal former and putative co-crystals in order to establish whether or not true co- crystals had been formed. Other techniques, used in an analogous fashion, include differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), solid state NMR spectroscopy, and Raman spectroscopy. Single crystal X-ray diffraction is especially useful in identifying co-crystal structures.
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • TGA solid state NMR spectroscopy
  • Raman spectroscopy Raman spectroscopy
  • the present invention therefore provides a process of screening for co-crystal compounds, which comprises:
  • APIs and co-crystal formers of the present invention have one or more chiral centers and may exist in a variety of stereoisomeric configurations. As a consequence of these chiral centers, several APIs and co-crystal formers of the present invention occur as racemates, mixtures of enantiomers and as individual enantiomers, as well as diastereomers and mixtures of diastereomers. All such racemates, enantiomers, and diastereomers are within the scope of the present invention including, for example, cis- and tr ⁇ s-isomers, R- and S-enantiomers, and (D)- and (L)-isomers.
  • Co-crystals of the present invention can include isomeric forms of either the API or the co-crystal former or both.
  • Isomeric forms of APIs and co-crystal formers include, but are not limited to, stereoisomers such as enantiomers and diastereomers.
  • a co-crystal can comprise a racemic API and/or co-crystal former.
  • a co-crystal can comprise an enantiomerically pure API and/or co-crystal former.
  • a co-crystal can comprise an API or a co-crystal former with an enantiomeric excess of about 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 96 percent, 97 percent, 98 percent, 99 percent, greater than 99 percent, or any intermediate value.
  • stereoisomeric APIs include modafinil, cw-itraconazole, ibuprofen, and flurbiprofen.
  • stereoisomeric co-crystal formers include tartaric acid and malic acid.
  • Co-crystals comprising enantiomerically pure components (e.g., API or co-crystal former) can give rise to chemical and/or physical properties which are modulated with respect to those of the corresponding co-crystal comprising a racemic component.
  • the modafinil :malonic acid co-crystal from Example 10 comprises racemic modafinil.
  • Enantiomerically pure R-modafinil:malonic acid can conceivably be synthesized via the same or another method of the present invention and is therefore included in the scope of the invention.
  • enantiomerically pure S- modaf ⁇ nikmalonic acid can conceivably be synthesized via a method of the present invention and is therefore included in the scope of the invention.
  • a co-crystal comprising an enantiomerically pure component can give rise to a modulation of, for example, activity, bioavailability, or solubility, with respect to the corresponding co-crystal comprising a racemic component.
  • the co-crystal R-modafinil:malonic acid can have modulated properties as compared to the racemic modafinikmalonic acid co-crystal.
  • racemic co-crystal refers to a co-crystal which is comprised of an equimolar mixture of two enantiomers of the API, the co-crystal former, or both.
  • a co-crystal comprising a stereoisomeric API and a non-stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the API enantiomers.
  • a co-crystal comprising a non-stereoisomeric API and a stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the co-crystal former enantiomers.
  • a co-crystal comprising a stereoisomeric API and a stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the API enantiomers and of the co-crystal former enantiomers.
  • enantiomerically pure co- crystal refers to a co-crystal which is comprised of a stereoisomeric API or a stereoisomeric co-crystal former or both where the enantiomeric excess of the stereoisomeric species is greater than or equal to about 90 percent ee.
  • the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the bioavailability is modulated with respect to the racemic co-crystal.
  • the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the activity is modulated with respect to the racemic co-crystal.
  • the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the solubility is modulated with respect to the racemic co-crystal.
  • enantiomerically pure includes a composition which is substantially enantiomerically pure and includes, for example, a composition with greater than or equal to about 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent enantiomeric excess.
  • the present invention provides a process for modulating the solubility of an API, which process comprises:
  • solubility of the API is modulated such that the aqueous solubility is increased.
  • Solubility of APIs may be measured by any conventional means such as chromatography (e.g., HPLC) or spectroscopic determination of the amount of API in a saturated solution of the API, such as UV-spectroscopy, IR-spectroscopy, Raman spectroscopy, quantitative mass spectroscopy, or gas chromatography.
  • the API may have low aqueous solubility.
  • low aqueous solubility in the present application refers to a compound having a solubility in water which is less than or equal to 10 mg/mL, when measured at 37 degrees C, and preferably less than or equal to 5 mg/mL or 1 mg/mL.
  • Low aqueous solubility can further be specifically defined as less than or equal to 900, 800, 700, 600, 500, 400, 300, 200 150 100, 90, 80, 70, 60, 50, 40, 30, 20 micrograms/mL, or further 10, 5 or 1 micrograms/mL, or further 900, 800, 700, 600, 500, 400, 300, 200 150, 100 90, 80, 70, 60, 50, 40, 30, 20, or 10 ng/mL, or less than 10 ng/mL when measured at 37 degrees C.
  • Aqueous solubility can also be specified as less than 500, 400, 300, 200, 150, 100, 75, 50 or 25 mg/mL.
  • solubility can be increased 2, 3, 4, 5, 7, 10, 15, 20, 25, 50, 75, 100, 200, 300, 500, 750, 1000, 5000, or 10,000 times by making a co-crystal of the reference form (e.g., crystalline or amorphous free acid, free base or zwitter ion, hydrate or solvate), or a salt thereof.
  • aqueous solubility can be measured in simulated gastric fluid (SGF) or simulated intestinal fluid (SIF) rather than water.
  • the pH of the solvent used may also be specified as 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, or 14 or any pH in between successive values.
  • Examples of embodiments includes: co-crystal compositions with an aqueous solubility, at 37 degrees C and a pH of 7.0, that is increased at least 5 fold over the reference form, co-crystal compositions with a solubility in SGF that is increased at least 5 fold over the reference form, co-crystal compositions with a solubility in SIF that is increased at least 5 fold over the reference form.
  • the dissolution profile of the API is modulated whereby the aqueous dissolution rate or the dissolution rate in simulated gastric fluid or in simulated intestinal fluid, or in a solvent or plurality of solvents is increased.
  • Dissolution rate is the rate at which API solids dissolve in a dissolution medium.
  • the rate-limiting step in the absorption process is often the dissolution rate. Because of a limited residence time at the absorption site, APIs that are not dissolved before they are removed from intestinal absorption site are considered useless. Therefore, the rate of dissolution has a major impact on the performance of APIs that are poorly soluble. Because of this factor, the dissolution rate of APIs in solid dosage forms is an important, routine, quality control parameter used in the API manufacturing process.
  • Dissolution rate K S (C 3 -C)
  • K dissolution rate constant
  • S is the surface area
  • C s is the apparent solubility
  • C is the concentration of API in the dissolution medium.
  • C 3 -C is approximately equal to C s .
  • the dissolution rate of APIs may be measured by conventional means known in the art.
  • the increase in the dissolution rate of a co-crystal, as compared to the reference form (e.g., free form or salt), may be specified, such as by 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100%, or by 2, 3, 4, 5 ,6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500, 1000, 10,000, or 100,000 fold greater than the reference form (e.g., free form or salt form) in the same solution.
  • Conditions under which the dissolution rate is measured is the same as discussed above
  • the increase in dissolution may be further specified by the time the composition remains supersaturated before reaching equilibrium solubility.
  • Examples of above embodiments include: co-crystal compositions with a dissolution rate in aqueous solution, at 37 degrees C and a pH of 7.0, that is increased at least 5 fold over the reference form, co-crystal compositions with a dissolution rate in SGF that is increased at least 5 fold over the reference form, co-crystal compositions with a dissolution rate in SIF that is increased at least 5 fold over the reference form.
  • the methods of the present invention are used to make a pharmaceutical API formulation with greater solubility, dissolution, and bioavailability. Bioavailability can be improved via an increase in AUC, reduced time to T ma , (the time to reach peak blood serum levels), or increased C max ,.
  • the present invention can result in higher plasma concentrations of API when compared to the neutral form or salt alone (reference form).
  • AUC is the area under the plot of plasma concentration of API (not logarithm of the concentration) against time after API administration. The area is conveniently determined by the "trapezoidal rule": The data points are connected by straight line segments, perpendiculars are erected from the abscissa to each data point, and the sum of the areas of the triangles and trapezoids so constructed is computed. When the last measured concentration (C Thread, at time t n ) is not zero, the AUC from t n to infinite time is estimated by C Cincinnati/k eI .
  • the AUC is of particular use in estimating bioavailability of APIs, and in estimating total clearance of APIs (Cl ⁇ ).
  • AUC F • D/Cl ⁇ , where F is the absolute bioavailability of the API.
  • the present invention provides a process for modulating the bioavailability of an API when administered in its normal and effective dose range as a co-crystal, whereby the AUC is increased, the time to T max is reduced, or C max is increased, as compared to a reference form, which process comprises:
  • Examples of the above embodiments include: co-crystal compositions with a time to T max that is reduced by at least 10% as compared to the reference form, co-crystal compositions with a time to T max that is reduced by at least 20% over the reference form, co-crystal compositions with a time to T max that is reduced by at least 40% over the reference form, co-crystal compositions with a time to T max that is reduced by at least 50% over the reference form, co-crystal compositions with a T max that is reduced by at least 60% over the reference form, co-crystal compositions with a T max that is reduced by at least 70% over the reference form, co-crystal compositions with a T ma that is reduced by at least 80% over the reference form, co-crystal compositions with a T max that is reduced by at least 90% over the reference form, co-crystal compositions with a C ma ⁇ that is increased by at least 20% over the reference form, co-crystal compositions with a C max that is increased by
  • the present invention provides a process for improving the dose response of an API, which process comprises:
  • Dose response is the quantitative relationship between the magnitude of response and the dose inducing the response and may be measured by conventional means known in the art.
  • the curve relating effect (as the dependent variable) to dose (as the independent variable) for an API-cell system is the "dose-response curve".
  • dose-response curve is the measured response to an API plotted against the dose of the API (mg/kg) given.
  • the dose response curve can also be a curve of AUC against the dose of the API given.
  • a co-crystal of the present mvention has an increased dose response curve or a more linear dose response curve than the corresponding reference compound.
  • the present invention provides a process for improving the stability of an API (as compared to a reference form such as its free form or a salt thereof), which process comprises:
  • compositions of the present invention including the API or active pharmaceutical ingredient (API) and formulations comprising the API, are suitably stable for pharmaceutical use.
  • the API or formulations thereof of the present invention are stable such that when stored at 30 degrees C for 2 years, less than 0.2 % of any one degradant is formed.
  • degradant refers herein to product(s) of a single type of chemical reaction. For example, if a hydrolysis event occurs that cleaves a molecule into two products, for the purpose of the present invention, it would be considered a single degradant. More preferably, when stored at 40 degrees C for 2 years, less than 0.2 % of any one degradant is formed.
  • the relative humidity (RH) may be specified as ambient (RH), 75 % (RH), or as any single integer between 1 to 99 %. Difficult to Salt or Unsaltable Compounds
  • the present invention provides a process for making co- crystals of unsaltable or difficult to salt APIs which process comprises:
  • Difficult to salt compounds include bases with a pKa less than 3 or acids with a pKa greater than 10.
  • Zwitter ions are also difficult to salt or unsaltable compounds according to the present invention.
  • the present invention provides a method for decreasing the hygroscopicity of an API, which method comprises:
  • An aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a co-crystal of an API that is less hygroscopic than amorphous or crystalline, free form or salt (including metal salts such as sodium, potassium, lithium, calcium, magnesium) or another reference compound.
  • Hygroscopicity can be assessed by dynamic vapor sorption analysis, in which 5-50 mg of the compound is suspended from a Cahn microbalance.
  • the compound being analyzed should be placed in a non- hygroscopic pan and its weight should be measured relative to an empty pan composed of identical material and having nearly identical size, shape, and weight. Ideally, platinum pans should be used.
  • the pans should be suspended in a chamber through which a gas, such as air or nitrogen, having a controlled and known percent relative humidity (%RH) is flowed until eqilibrium criteria are met.
  • a gas such as air or nitrogen
  • Typical equilibrium criteria include weight changes of less than 0.01 % over 3 minutes at constant humidity and temperature.
  • the relative humidity should be measured for samples dried under dry nitrogen to constant weight ( ⁇ 0.01 % change in 3 minutes) at 40 degrees C unless doing so would de-solvate or otherwise convert the material to an amorphous compound.
  • the hygroscopicity of a dried compound can be assessed by increasing the RH from 5 to 95 % in increments of 5 % RH and then decreasing the RH from 95 to 5 % in 5 % increments to generate a moisture sorption isotherm.
  • the sample weight should be allowed to equilibrate between each change in % RH. If the compound deliquesces or becomes amorphous above 75 % RH, but below 95 % RH, the experiment should be repeated with a fresh sample and the relative humidity range for the cycling should be narrowed to 5-75 % RH or 10-75 % RH, instead of 5-95 %RH.
  • Hygroscopicity can be defined using various parameters. For purposes of the present invention, a non-hygroscopic molecule should not gain or lose more than 1.0 %, or more preferably, 0.5 % weight at 25 degrees C when cycled between 10 and 75 % RH (relative humidity at 25 degrees C).
  • the non-hygroscopic molecule more preferably should not gain or lose more than 1.0 %, or more preferably, 0.5 % weight when cycled between 5 and 95 % RH at 25 degrees C, or more than 0.25 % of its weight between 10 and 75 % RH. Most preferably, a non-hygroscopic molecule will not gain or lose more than 0.25 % of its weight when cycled between 5 and 95 % RH.
  • hygroscopicity can be defined using the parameters of Callaghan et al., "Equilibrium moisture content ofpharmaceutical excipients", in Api Dev. Ind. Pharm., Vol. 8, pp. 335-369 (1982). Callaghan et al. classified the degree of hygroscopicity into four classes.
  • Class 1 Non-hygroscopic Essentially no moisture increases occur at relative humidities below 90 %.
  • Class 2 Slightly hygroscopic Essentially no moisture increases occur at relative humidities below 80%.
  • Class 3 Moderately hygroscopic Moisture content does not increase more than 5 % after storage for 1 week at relative humidities below 60 %.
  • Class 4 Very hygroscopic Moisture content increase may occur at relative humidities as low as 40 to 50 %.
  • hygroscopicity can be defined using the parameters of the European Pharmacopoeia Technical Guide (1999, p. 86) which has defined hygrospocity, based on the static method, after storage at 25 degrees C for 24 hours at 80 % RH:
  • Hygroscopic Increase in mass is less than 15 percent m/m and equal to or greater than 0.2 percent m/m.
  • Co-crystals of the present invention can be set forth as being in Class 1, Class 2, or Class 3, or as being Slightly hygroscopic, Hygroscopic, or Very Hygroscopic. Co- crystals of the present invention can also be set forth based on their ability to reduce hygroscopicity. Thus, preferred co-crystals of the present invention are less hygroscopic than a reference compound.
  • the reference compound can be specified as the API in free form (free acid, free base, hydrate, solvate, etc.) or salt (e.g., especially metal salts such as sodium, potassium, lithium, calcium, or magnesium).
  • co-crystals that do not gain or lose more than 1.0 % weight at 25 degrees C when cycled between 10 and 75 % RH, wherein the reference compound gains or loses more than 1.0 % weight under the same conditions.
  • co-crystals that do not gain or lose more than 0.5 % weight at 25 degrees C when cycled between 10 and 75 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions.
  • co-crystals that do not gain or lose more than 1.0 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 1.0 % weight under the same conditions.
  • co-crystals that do not gain or lose more than 0.5 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions.
  • co-crystals that do not gain or lose more than 0.25 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions.
  • co-crystals that have a hygroscopicity (according to Callaghan et al.) that is at least one class lower than the reference compound or at least two classes lower than the reference compound. Included are a Class 1 co-crystal of a Class 2 reference compound, a Class 2 co-crystal of a Class 3 reference compound, a Class 3 co-crystal of a Class 4 reference compound, a Class 1 co- crystal of a Class 3 reference compound, a Class 1 co-crystal of a Class 4 reference compound, or a Class 2 co-crystal of a Class 4 reference compound.
  • co-crystals that have a hygroscopicity (according to the European Pharmacopoeia Technical Guide) that is at least one class lower than the reference compound or at least two classes lower than the reference compound.
  • a slightly hygroscopic co-crystal of a hygroscopic reference compound a hygroscopic co-crystal of a very hygroscopic reference compound, a very hygroscopic co-crystal of a deliquescent reference compound, a slightly hygroscopic co-crystal of a very hygroscopic reference compound, a slightly hygroscopic co-crystal of a deliquescent reference compound, and a hygroscopic co-crystal of a deliquescent reference compound.
  • the present invention provides a process for crystallizing an amorphous compound, which process comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
  • An amorphous compound includes compounds that do not crystallize using routine methods in the art.
  • the present invention provides a process for reducing the form diversity of an API, which process comprises:
  • the number of forms of a co-crystal is compared to the number of forms of a reference compound (e.g. the free form or a salt of the API) that can be made using routine methods in the art.
  • a reference compound e.g. the free form or a salt of the API
  • the present invention provides a process for modifying the morphology of an API, which process comprises:
  • the co-crystal comprises or consists of a co-crystal former and a pharmaceutical wherein the interaction between the two, e.g., H-bonding, occurs between a functional group of Table III of an API with a corresponding interacting group of Table III.
  • the co-crystal comprises a co-crystal former of Table I or II and an API with a corresponding interacting group of Table III.
  • the co-crystal comprises an API from Table IV and a co-crystal former with a functional group of Table III.
  • the co-crystal is from Table I or II.
  • co-crystals having an H-bond acceptor on the first molecule and an H-bond donor on the second molecule are included in the present invention.
  • Table IV includes the CAS number, chemical name or a PCT or patent reference (each incorporated herein in their entireties). Thus, whether a particular API contains an H-bond donor, acceptor or both is readily apparent.
  • the co-crystal former and API each have only one H- bond donor/acceptor.
  • the molecular weight of the API is less than 2000, 1500, 1000, 750, 500, 350, 200, or 150 Daltons. In another embodiment, the molecular weight of the API is between 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 900-1000, 1000-1200, 1200-1400, 1400-1600, 1600-1800, or 1800-2000. APIs with the above molecular weights may also be specifically excluded from the present invention.
  • the hydrogen bond donor moieties of a co-crystal can include, but are not limited to, any one, any two, any three, any four, or more of the following: amino-pyridine, primary amine, secondary amine, sulfonamide, primary amide, secondary amide, alcohol, and carboxylic acid.
  • the hydrogen bond acceptor moieties of a co-crystal can include, but are not limited to, any one, any two, any three, any four, or more of the following: amino-pyridine, primary amine, secondary amine, sulfonamide, primary amide, secondary amide, alcohol, carboxylic acid, carbonyl, cyano, dimethoxyphenyl, sulfonyl, aromatic nitrogen (6 membered ring), ether, chloride, organochloride, bromide, organobromide, and organoiodide. Hydrogen bonds are known to form many supramolecular structures including, but not limited to, a catemer, a dimer, a trimer, a t ⁇ tramer, or a higher order structure.
  • Tables V-XXI list specific hydrogen bond donor and acceptor moieties and their approximate interaction distances from the electromagnetic donor atom through the hydrogen atom to the electromagnetic acceptor atom.
  • Table V lists functional groups that are known to hydrogen bond with amino-pyridines. Amino-pyridines comprise two distinct sites of hydrogen bond donation/acceptance. Both the aromatic nitrogen atom (Npy) and the amine group (NH 2 ) can participate in hydrogen bonds. The ability of a given functional group to participate in a hydrogen bond as a donor or as an acceptor or both can be determined by inspection by those skilled in the art.
  • Tables V-XXI are taken from an analysis of solid-state structures as reported in the Cambridge Structural Database (CSD). These data include a number of hydrogen bonding interactions between many functional groups and their associated interaction distances.
  • peptides, proteins, nucleic acids or other biological APIs are excluded from the present invention.
  • all non- pharmaceutically acceptable co-crystal formers are excluded from the present invention.
  • organometalic APIs are excluded from the present invention.
  • a co-crystal former comprising any one or more of the functional groups of Table III may be specifically excluded from the present invention.
  • any one or more of the co-crystal formers of Table I or II may be specifically excluded from the present invention. Any APIs currently known in the art may also be specifically excluded from the present invention.
  • the API is not a salt, is not a non-metal salt, or is not a metal salt, e.g., sodium, potassium, lithium, calcium or magnesium.
  • the API is a salt, is a non-metal salt, or is a metal salt, e.g., sodium, potassium, lithium, calcium, magnesium.
  • the API does not contain a halogen. In one embodiment, the API does contain a halogen.
  • any one or more of the APIs of Table IV may be specifically excluded from the present invention.
  • Any APIs currently known in the art may also be specifically excluded from the present invention.
  • nabumetone:2,3-naphthalenediol fluoxetine HCkbenzoic acid, fluoxetine HCLsuccinic acid, acetaminophe piperazine, acetaminophe theophylline, theophylline: salicylic acid, theophylline :p-hydiOxybenzoic acid, theophylline: sorbic acid, theophylline :l-hydroxy-2- naphthoic acid, theophylline :glycolic acid, theophylline:2,5-dihydroxybenzoic acid, theophylline xhloroacetic acid, bis(diphenylhydantoin):9-ethyladenine acetylacetone solvate, bis(diphenylhydantoin)
  • a pharmaceutical composition can be formulated to contain an API in co-crystal form as micronized or nano-sized particles. More specifically, another embodiment couples the processing of a pure API to a co-crystal form with the process of making a controlled particle size for manipulation into a pharmaceutical dosage form. This embodiment combines two processing steps into a single step via techniques such as, but not limited to, grinding, alloying, or sintering (i.e., heating a powder mix). The coupling of these processes overcomes a serious limitation of having to isolate and store the bulk drug that is required for a formulation, which in some cases can be difficult to isolate (e.g., amorphous, chemically or physically unstable).
  • Excipients employed in pharmaceutical compositions of the present invention can be solids, semi-solids, liquids or combinations thereof. Preferably, excipients are solids.
  • Compositions of the invention containing excipients can be prepared by any known technique of pharmacy that comprises admixing an excipient with an API or therapeutic agent.
  • a pharmaceutical composition of the invention contains a desired amount of API per dose unit and, if intended for oral administration, can be in the form, for example, of a tablet, a caplet, a pill, a hard or soft capsule, a lozenge, a cachet, a dispensable powder, granules, a suspension, an elixir, a dispersion, or any other form reasonably adapted for such administration.
  • oral dosage forms that are discrete dose units each containing a predetermined amount of the API, such as tablets or capsules.
  • APIs with an inappropriate pH for transdermal patches can be co-crystallized with an appropriate co-crystal former, thereby adjusting its pH to an appropriate level for use as a transdermal patch.
  • an APIs pH level can be optimized for use in a transdermal patch via co-crystallization with an appropriate co-crystal former.
  • Non-limiting examples follow of excipients that can be used to prepare pharmaceutical compositions of the invention.
  • compositions of the invention optionally comprise one or more pharmaceutically acceptable carriers or diluents as excipients.
  • suitable carriers or diluents illustratively include, but are not limited to, either individually or in combination, lactose, including anhydrous lactose and lactose monohydrate; starches, including directly compressible starch and hydrolyzed starches (e.g., CelutabTM and EmdexTM); mannitol; sorbitol; xylitol; dextrose (e.g., CereloseTM 2000) and dextrose monohydrate; dibasic calcium phosphate dihydrate; sucrose-based diluents; confectioner's sugar; monobasic calcium sulfate monohydrate; calcium sulfate dihydrate; granular calcium lactate trihydrate; dextrates; inositol; hydrolyzed cereal solids; amylose; celluloses including microcrystalline cellulose, food grade sources of alpha- and a
  • Such carriers or diluents constitute in total about 5% to about 99%, preferably about 10% to about 85%, and more preferably about 20% to about 80%, of the total weight of the composition.
  • the carrier, carriers, diluent, or diluents selected preferably exhibit suitable flow properties and, where tablets are desired, compressibility.
  • Lactose, mannitol, dibasic sodium phosphate, and microcrystalline cellulose are preferred diluents. These diluents are chemically compatible with many co-crystals described herein.
  • the use of extragranular microcrystalline cellulose that is, microcrystalline cellulose added to a granulated composition
  • Lactose, especially lactose monohydrate is particularly preferred.
  • Lactose typically provides compositions having suitable release rates of co-crystals, stability, pre- compression flowability, and/or drying properties at a relatively low diluent cost. It provides a high density substrate that aids densification during granulation (where wet granulation is employed) and therefore improves blend flow properties and tablet properties.
  • compositions of the invention optionally comprise one or more pharmaceutically acceptable disintegrants as excipients, particularly for tablet formulations.
  • Suitable disintegrants include, but are not limited to, either individually or in combination, starches, including sodium starch glycolate (e.g., ExplotabTM of PenWest) and pregelatinized corn starches (e.g., NationalTM 1551 of National Starch and Chemical Company, NationalTM 1550, and ColorconTM 1500), clays (e.g., VeegumTM HV of R.T.
  • Vanderbilt celluloses such as purified cellulose, microcrystalline cellulose, methylcellulose, carboxymethylcellulose and sodium carboxymethylcellulose, croscarmellose sodium (e.g., Ac-Di-SolTM of FMC), alginates, crospovidone, and gums such as agar, guar, locust bean, karaya, pectin and fragacanth gums.
  • Disintegrants may be added at any suitable step during the preparation of the composition, particularly prior to granulation or during a lubrication step prior to compression. Such disintegrants, if present, constitute in total about 0.2% to about 30%, preferably about 0.2% to about 10%, and more preferably about 0.2% to about 5%, of the total weight of the composition.
  • Croscarmellose sodium is a preferred disintegrant for tablet or capsule disintegration, and, if present, preferably constitutes about 0.2% to about 10%, more preferably about 0.2% to about 7%, and still more preferably about 0.2% to about 5%, of the total weight of the composition. Croscarmellose sodium confers superior intragranular disintegration capabilities to granulated pharmaceutical compositions of the present invention.
  • Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable binding agents or adhesives as excipients, particularly for tablet formulations. Such binding agents and adhesives preferably impart sufficient cohesion to the powder being tableted to allow for normal processing operations such as sizing, lubrication, compression and packaging, but still allow the tablet to disintegrate and the composition to be absorbed upon ingestion.
  • binding agents may also prevent or inhibit crystallization or recrystallization of a co-crsytal of the present invention once the salt has been dissolved in a solution.
  • Suitable binding agents and adhesives include, but are not limited to, either individually or in combination, acacia; fragacanth; sucrose; gelatin; glucose; starches such as, but not limited to, pregelatinized starches (e.g., NationalTM 1511 and NationalTM 1500); celluloses such as, but not limited to, methylcellulose and carmellose sodium (e.g., TyloseTM); alginic acid and salts of alginic acid; magnesium aluminum silicate; PEG; guar gum; polysaccharide acids; bentonites; povidone, for example povidone K-15, K-30 and K-29/32; polymethacrylates; HPMC; hydroxypropylcellulose (e.g., KlucelTM of Aqualon); and ethylcellulose (e.g., EthocelTM of the Dow Chemical
  • binding agents are polymers comprising amide, ester, ether, alcohol or ketone groups and, as such, are preferably included in pharmaceutical compositions of the present invention.
  • Polyvinylpyrrolidones such as povidone K-30 are especially preferred.
  • Polymeric binding agents can have varying molecular weight, degrees of crosslinking, and grades of polymer.
  • Polymeric binding agents can also be copolymers, such as block co-polymers that contain mixtures of ethylene oxide and propylene oxide units. Variation in these units' ratios in a given polymer affects properties and performance. Examples of block co-polymers with varying compositions of block units are Poloxamer 188 and Poloxamer 237 (BASF Corporation).
  • compositions of the invention optionally comprise one or more pharmaceutically acceptable wetting agents as excipients.
  • Such wetting agents are preferably selected to maintain the co-crystal in close association with water, a condition that is believed to improve bioavailability of the composition.
  • Such wetting agents can also be useful in solubilizing or increasing the solubility of co-crystals.
  • Non-limiting examples of surfactants that can be used as wetting agents in pharmaceutical compositions of the invention include quaternary ammonium compounds, for example benzalkonium chloride, benzethonium chloride and cetylpyridinium chloride, dioctyl sodium sulfosuccinate, polyoxyethylene alkylphenyl ethers, for example nonoxynol 9, nonoxynol 10, and degrees Ctoxynol 9, poloxamers (polyoxyethylene and polyoxypropylene block copolymers), polyoxyethylene fatty acid glycerides and oils, for example polyoxyethylene (8) caprylic/capric mono- and diglycerides (e.g., LabrasolTM of Gattefosse), polyoxyethylene (35) castor oil and polyoxyethylene (40) hydrogenated castor oil; polyoxyethylene alkyl ethers, for example polyoxyethylene (20) cetostearyl ether, polyoxyethylene fatty acid esters, for example polyoxyethylene (40) stearate, poly
  • Sodium lauryl sulfate is a particularly preferred wetting agent.
  • Sodium lauryl sulfate if present, constitutes about 0.25% to about 7%, more preferably about 0.4% to about 4%, and still more preferably about 0.5% to about 2%, of the total weight of the pharmaceutical composition.
  • compositions of the invention optionally comprise one or more pharmaceutically acceptable lubricants (including anti-adherents and/or glidants) as excipients.
  • suitable lubricants include, but are not limited to, either individually or in combination, glyceryl behapate (e.g., CompritolTM 888 of Gattefosse); stearic acid and salts thereof, including magnesium, calcium and sodium stearates; hydrogenated vegetable oils (e.g., Sterotex of Abitec); colloidal silica; talc; waxes; boric acid; sodium benzoate; sodium acetate; sodium fumarate; sodium chloride; DL-leucine; PEG (e.g., CarbowaxTM 4000 and CarbowaxTM 6000 of the Dow Chemical Company); sodium oleate; sodium lauryl sulfate; and magnesium lauryl sulfate.
  • Such lubricants if present, constitute in total about 0. 1% to about 10%, preferably about
  • Magnesium stearate is a preferred lubricant used, for example, to reduce friction between the equipment and granulated mixture during compression of tablet formulations.
  • Suitable anti-adherents include, but are not limited to, talc, cornstarch, DL- leucine, sodium lauryl sulfate and metallic stearates.
  • Talc is a preferred anti-adherent or glidant used, for example, to reduce formulation sticking to equipment surfaces and also to reduce static in the blend.
  • Talc if present, constitutes about 0.1% to about 10%, more preferably about 0.25% to about 5%, and still more preferably about 0.5% to about 2%, of the total weight of the pharmaceutical composition.
  • Glidants can be used to promote powder flow of a solid formulation. Suitable glidants include, but are not limited to, colloidal silicon dioxide, starch, talc, tribasic calcium phosphate, powdered cellulose and magnesium trisilicate. Colloidal silicon dioxide is particularly preferred.
  • compositions of the invention can further comprise, for example, buffering agents.
  • one or more effervescent agents can be used as disintegrants and/or to enhance organoleptic properties of pharmaceutical compositions of the invention.
  • one or more effervescent agents are preferably present in a total amount of about 30% to about 75%, and preferably about 45% to about 70%, for example about 60%, by weight of the pharmaceutical composition.
  • an effervescent agent present in a solid dosage form in an amount less than that effective to promote disintegration of the dosage form, provides improved dispersion of the API in an aqueous medium.
  • the effervescent agent is effective to accelerate dispersion of the API from the dosage form in the gastrointestinal tract, thereby further enhancing absorption and rapid onset of therapeutic effect.
  • an effervescent agent is preferably present in an amount of about 1% to about 20%, more preferably about 2.5% to about 15%, and still more preferably about 5% to about 10%, by weight of the pharmaceutical composition.
  • an “effervescent agent” herein is an agent comprising one or more compounds which, acting together or individually, evolve a gas on contact with water.
  • the gas evolved is generally oxygen or, most commonly, carbon dioxide.
  • Preferred effervescent agents comprise an acid and a base that react in the presence of water to generate carbon dioxide gas.
  • the base comprises an alkali metal or alkaline earth metal carbonate or bicarbonate and the acid comprises an aliphatic carboxylic acid.
  • Non-limiting examples of suitable bases as components of effervescent agents useful in the invention include carbonate salts (e.g., calcium carbonate), bicarbonate salts (e.g., sodium bicarbonate), sesquicarbonate salts, and mixtures thereof. Calcium carbonate is a preferred base.
  • Non-limiting examples of suitable acids as components of effervescent agents and/or solid organic acids useful in the invention include citric acid, tartaric acid (as D-, L-, or D/L-tartaric acid), malic acid (as D-, L-, or DL-malic acid), maleic acid, fumaric acid, adipic acid, succinic acid, acid anhydrides of such acids, acid salts of such acids, and mixtures thereof.
  • Citric acid is a preferred acid.
  • the weight ratio of the acid to the base is about 1 : 100 to about 100:1, more preferably about 1:50 to about 50:1, and still more preferably about 1 : 10 to about 10:1.
  • the ratio of the acid to the base is approximately stoichiometric.
  • Excipients which solubilize APIs typically have both hydrophilic and hydrophobic regions, or are preferably amphiphilic or have amphiphilic regions.
  • One type of amphiphilic or partially-amphiphilic excipient comprises an amphiphilic polymer or is an amphiphilic polymer.
  • a specific amphiphilic polymer is a polyalkylene glycol, which is commonly comprised of ethylene glycol and/or propylene glycol subunits.
  • Such polyalkylene glycols can be esterified at their termini by a carboxylic acid, ester, acid anhyride or other suitable moiety.
  • excipients examples include poloxamers (symmetric block copolymers of ethylene glycol and propylene glycol; e.g., poloxamer 237), polyalkyene glycolated esters of tocopherol (including esters formed from a di- or multi-functional carboxylic acid; e.g., d-alpha-tocopherol polyethylene glycol- 1000 succinate), and macrogolglycerides (formed by alcoholysis of an oil and esterification of a polyalkylene glycol to produce a mixture of mono-, di- and tri-glycerides and mono- and di-esters; e.g., stearoyl macrogol-32 glycerides).
  • poloxamers symmetric block copolymers of ethylene glycol and propylene glycol
  • polyalkyene glycolated esters of tocopherol including esters formed from a di- or multi-functional carboxylic acid; e.g., d-alpha-tocopherol
  • compositions of the present invention can comprise about 10 % to about 50 %, about 25 % to about 50 %, about 30 % to about 45 %, or about 30 % to about 35 % by weight of a co-crystal; about 10 % to about 50 %, about 25 % to about 50 %, about 30 % to about 45 %, or about 30 % to about 35 % by weight of an excipient which inhibits crystallization in aqueous solution, in simulated gastric fluid, or in simulated intestinal fluid; and about 5 % to about 50 %, about 10 % to about 40 %, about 15 % to about 35 %, or about 30 % to about 35 % by weight of a binding agent.
  • the weight ratio of the co-crystal to the excipient which inhibits crystallization to binding agent is about 1 to 1 to 1.
  • Solid dosage forms of the invention can be prepared by any suitable process, not limited to processes described herein.
  • An illustrative process comprises (a) a step of blending an API of the invention with one or more excipients to form a blend, and (b) a step of tableting or encapsulating the blend to form tablets or capsules, respectively.
  • solid dosage forms are prepared by a process comprising (a) a step of blending a co-crystal of the invention with one or more excipients to form a blend, (b) a step of granulating the blend to form a granulate, and (c) a step of tableting or encapsulating the blend to form tablets or capsules respectively.
  • Step (b) can be accomplished by any dry or wet granulation technique known in the art, but is preferably a dry granulation step.
  • a salt of the present invention is advantageously granulated to form particles of about 1 micrometer to about 100 micrometer, about 5 micrometer to about 50 micrometer, or about 10 micrometer to about 25 micrometer.
  • One or more diluents, one or more disintegrants and one or more binding agents are preferably added, for example in the blending step, a wetting agent can optionally be added, for example in the granulating step, and one or more disintegrants are preferably added after granulating but before tableting or encapsulating.
  • a lubricant is preferably added before tableting. Blending and granulating can be performed independently under low or high shear.
  • a process is preferably selected that forms a granulate that is uniform in API content, that readily disintegrates, that flows with sufficient ease so that weight variation can be reliably controlled during capsule filling or tableting, and that is dense enough in bulk so that a batch can be processed in the selected equipment and individual doses fit into the specified capsules or tablet dies.
  • solid dosage forms are prepared by a process that includes a spray drying step, wherein an API is suspended with one or more excipients in one or more sprayable liquids, preferably a non-protic (e.g., non-aqueous or nonalcoholic) sprayable liquid, and then is rapidly spray dried over a current of warm air.
  • a granulate or spray dried powder resulting from any of the above illustrative processes can be compressed or molded to prepare tablets or encapsulated to prepare capsules. Conventional tableting and encapsulation techniques known in the art can be employed. Where coated tablets are desired, conventional coating techniques are suitable.
  • Excipients for tablet compositions of the invention are preferably selected to provide a disintegration time of less than about 30 minutes, preferably about 25 minutes or less, more preferably about 20 minutes or less, and still more preferably about 15 minutes or less, in a standard disintegration assay.
  • compositions can be administered by controlled-, sustained-, or delayed-release means.
  • Controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled release counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Controlled-release formulations include: 1) extended activity of the drug; 2) reduced dosage frequency; 3) increased patient compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects; 6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug activity; and 10) improvement in speed of control of diseases or conditions.
  • Conventional dosage forms generally provide rapid or immediate drug release from the formulation. Depending on the pharmacology and pharmacokinetics of the drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like.
  • controlled- release formulations can be used to control a drug's onset of action, duration of action, plasma levels within the therapeutic window, and peak blood levels.
  • controlled- or extended-release dosage forms or formulations can be used to ensure that the maximum effectiveness of a drug is achieved while minimizing potential adverse effects and safety concerns, which can occur both from under dosing a drug (i.e., going below the minimum therapeutic levels) as well as exceeding the toxicity level for the drug.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions or compounds.
  • a variety of known controlled- or extended-release dosage forms, formulations, and devices can be adapted for use with the co-crystals and compositions of the mvention.
  • Examples include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365,185 Bl; each of which is incorporated herein by reference.
  • dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, liydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), multilayer coatings, microparticles, liposomes, or microspheres or a combination thereof to provide the desired release profile in varying proportions.
  • ion exchange materials can be used to prepare immobilized, adsorbed co-crystals and thus effect controlled delivery of the drug. Examples of specific anion exchangers include, but are not limited to, Duolite® A568 and Duolite® AP143 (Rohm & Haas, Spring House, PA. USA).
  • One embodiment of the invention encompasses a unit dosage form which comprises a pharmaceutically acceptable co-crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof, and one or more pharmaceutically acceptable excipients or diluents, wherein the pharmaceutical composition or dosage form is formulated for controlled-release.
  • Specific dosage forms utilize an osmotic drug delivery system.
  • OROS® osmotic drug delivery system
  • This technology can readily be adapted for the delivery of compounds and compositions of the invention.
  • Various aspects of the technology are disclosed in U.S. Pat. Nos. 6,375,978 Bl; 6,368,626 Bl; 6,342,249 Bl; 6,333,050 B2; 6,287,295 Bl; 6,283,953 Bl; 6,270,787 Bl; 6,245,357 Bl; and 6,132,420; each of which is incorporated herein by reference.
  • OROS® that can be used to administer compounds and compositions of the invention
  • OROS® Push-PullTM Delayed Push-PullTM, Multi- Layer Push-PullTM, and Push-StickTM Systems, all of which are well known. See, e.g., http://www.alza.com.
  • Additional OROS® systems that can be used for the controlled oral delivery of compounds and compositions of the invention include OROS®-CT and L- OROS®. Id.; see also, Delivery Times, vol. II, issue II (Alza Corporation).
  • OROS® oral dosage forms are made by compressing a drag powder (e.g. co-crystal) into a hard tablet, coating the tablet with cellulose derivatives to form a semi-permeable membrane, and then drilling an orifice in the coating (e.g., with a laser).
  • a drag powder e.g. co-crystal
  • Kim, Cherng-ju, Controlled Release Dosage Form Design, 231-238 (Technomic Publishing, Lancaster, Pa.: 2000).
  • the advantage of such dosage forms is that the delivery rate of the drug is not influenced by physiological or experimental conditions. Even a drug with a pH-dependent solubility can be delivered at a constant rate regardless of the pH of the delivery medium.
  • OROS® drug delivery systems cannot be used to effectively deliver drugs with low water solubility. Id. at 234. Because co-crystals of this invention can be far more soluble in water than the API itself, they are well suited for osmotic-based delivery to patients. This invention does, however, encompass the incorporation of conventional crystalline API (e.g. pure API without co-crystal former), and non-salt isomers and isomeric mixtures thereof, into OROS® dosage forms.
  • conventional crystalline API e.g. pure API without co-crystal former
  • a specific dosage form of the invention comprises: a wall defining a cavity, the wall having an exit orifice formed or formable therein and at least a portion of the wall being semipermeable; an expandable layer located within the cavity remote from the exit orifice and in fluid communication with the semipermeable portion of the wall; a dry or substantially dry state drug layer located within the cavity adjacent to the exit orifice and in direct or indirect contacting relationship with the expandable layer; and a flow- promoting layer interposed between the inner surface of the wall and at least the external surface of the drug layer located within the cavity, wherein the drug layer comprises a co- crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof. See U.S. Pat. No. 6,368,626, the entirety of which is incorporated herein by reference.
  • Another specific dosage form of the invention comprises: a wall defining a cavity, the wall having an exit orifice formed or formable therein and at least a portion of the wall being semipermeable; an expandable layer located within the cavity remote from the exit orifice and in fluid communication with the semipermeable portion of the wall; a drug layer located within the cavity adjacent the exit orifice and in direct or indirect contacting relationship with the expandable layer; the drug layer comprising a liquid, active agent formulation absorbed in porous particles, the porous particles being adapted to resist compaction forces sufficient to form a compacted drug layer without significant exudation of the liquid, active agent formulation, the dosage form optionally having a placebo layer between the exit orifice and tlie drug layer, wherein the active agent formulation comprises a co-crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof. See U.S. Pat. No. 6,342,249, the entirety of which is incorporated herein by reference.
  • CrystalMaxTM comprises a sequence of automated, integrated high throughput robotic stations capable of rapid generation, identification and characterization of polymorphs, salts, and co-crystals of APIs and API candidates. Worksheet generation and combinatorial mixture design is carried out using proprietary design software ArchitectTM. Typically, an API or an API candidate is dispensed from an organic solvent into tubes and dried under a stream of nitrogen. Salts and/or co-crystal formers may also be dispensed and dried in the same fashion. Water and organic solvents may be combinatorially dispensed into the tubes using a multi-channel dispenser. Each tube in a 96-tube array is then sealed within 15 seconds of combinatorial dispensing to avoid solvent evaporation.
  • the mixtures are then rendered supersaturated by heating to 70 degrees C for 2 hours followed by a 1 degree C/minute cooling ramp to 5 degrees C.
  • Optical checks are then conducted to detect crystals and/or solid material. Once a solid has been identified in a tube, it is isolated through aspiration and drying. Raman spectra are then obtained on the solids and cluster classification of the spectral patterns is performed using proprietary software (InquireTM).
  • Co-crystals may be obtained by dissolving the separate components in a solvent and adding one to the other. The co-crystal may then precipitate or crystallize as the solvent mixture is evaporated slowly. The co-crystal may also be obtained by dissolving the two components in the same solvent or a mixture of solvents.
  • a co-crystal may be obtained by melting the two components together (i.e., co- melting) and allowing recrystallization to occur.
  • an anti-solvent may be added to facilitate crystallization.
  • a co-crystal may be obtained by melting the higher melting component on a glass slide and allowing it to recrystallize. The second component is then melted and is also allowed to recrystallize. The co-crystal may form as a separated phase/band in between the eutectic bands of the two original components.
  • a co-crystal may be obtained by mixing or grinding two components together in the solid state.
  • a co-crystal may be obtained by co-subliming a mixture of an API and a co- crystal former in the same sample cell as an intimate mixture either by heating, mixing or placing the mixture under vacuum.
  • a co-crystal may also be obtained by co-sublimation using a Kneudsen apparatus where the API and the co-crystal former are contained in separate sample cells, connected to a single cold finger, each of the sample cells is maintained at the same or different temperatures under a vaccum atmosphere in order to co-sublime the two components onto the cold-finger forming the desired co-crystal.
  • the purge gas used was dry nitrogen
  • the reference material was an empty aluminum pan that was crimped
  • the sample purge was 50 mL/minute.
  • DSC analysis of the sample was performed by placing ⁇ 2 mg of sample in an aluminum pan with a crimped pan closure.
  • the starting temperature was typically 20 degrees C with a heating rate of 10 degrees C/minute, and the ending temperature was 300 degrees C. Unless otherwise indicated, all reported transitions are as stated +/- 1.0 degrees C.
  • TGA analysis of samples was performed using a Q500 Thermogravimetric Analyzer (TA Instruments, New Castle, DE, U.S.A.), which uses Advantage for QW- Series, version 1.0.0.78, Thermal Advantage Release 2.0 (2001 TA Instruments-Water LLC).
  • the analysis software used was Universal Analysis 2000 for Windows 95/95/2000/NT, version 3.1E;Build 3.1.0.40 (2001 TA Instruments-Water LLC).
  • the purge gas used was dry nitrogen, the balance purge was 40 mL/minute N 2 , and the sample purge was 60 mL/minute N 2 .
  • TGA of the sample was performed by placing ⁇ 2 mg of sample in a platinum pan.
  • the starting temperature was typically 20 degrees C with a heating rate of 10 degrees C/minute, and the ending temperature was 300 degrees C.
  • a powder X-ray diffraction pattern for the samples was obtained using a D/Max Rapid, Contact (Rigaku/MSC, The Woodlands, TX, U.S.A.), which uses as its control software RINT Rapid Control software, Rigaku Rapid/XRD, version 1.0.0 (1999 Rigaku Co.).
  • RINT Rapid Control software Rigaku Rapid/XRD, version 1.0.0 (1999 Rigaku Co.
  • analysis software used were RINT Rapid display software, version 1.18 (Rigaku MSC), and JADE XRD Pattern Processing, versions 5.0 and 6.0 ((1995- 2002, Materials Data, Inc.).
  • the acquisition parameters were as follows: source was Cu with a K line at 1.5406A; x-y stage was manual; collimator size was 0.3 or 0.8 mm; capillary tube (Charles Supper Company, Natick, MA, U.S.A.) was 0.3 mm ID; reflection mode was used; the power to the X-ray tube was 46 kV; the current to the X-ray tube was 40 mA; the omega-axis was oscillating in a range of 0-5 degrees at a speed of 1 degree/minute; the phi-axis was spinning at an angle of 360 degrees at a speed of 2 degrees/second; 0.3 or 0.8 mm collimator; the collection time was 60 minutes; the temperature was room temperature; and the heater was not used.
  • the sample was presented to the X-ray source in a boron rich glass capillary.
  • the analysis parameters were as follows: the integration 2-theta range was 2-40 or 60 degrees; the integration chi range was 0-360 degrees; the number of chi segments was 1; the step size used was 0.02; the integration utility was cylint; normalization was used; dark counts were 8; omega offset was 180; and chi and phi offsets were 0.
  • the relative intensity of peaks in a diffractogram is not necessarily a limitation of the PXRD pattern because peak intensity can vary from sample to sample, e.g., due to crystalline impurities. Further, the angles of each peak can vary by about +/- 0.1 degrees, preferably +/-0.05. The entire pattern or most of the pattern peaks may also shift by about +/- 0.1 degree due to differences in calibration, settings, and other variations from instrument to instrument and from operator to operator. Procedure for Raman Acquisition. Filtering and Binning
  • the sample was either left in the glass vial in which it was processed or an aliquot of the sample was transferred to a glass slide.
  • the glass vial or slide was positioned in the sample chamber.
  • the measurement was made using an AlmegaTM Dispersive Raman (AlmegaTM Dispersive Raman, Thermo-Nicolet, 5225 Verona Road, Madison, WI 53711- 4495) system fitted with a 785nm laser source.
  • the sample was manually brought into focus using the microscope portion of the apparatus with a lOx power objective (unless otherwise noted), thus directing the laser onto the surface of the sample.
  • the spectrum was acquired using the parameters outlined in Table XXII. (Exposure times and number of exposures may vary; changes to parameters will be indicated for each acquisition.)
  • Each spectrum in a set was filtered using a matched filter of feature size 25 to remove background signals, including glass contributions and sample fluorescence. This is particularly important as large background signal or fluorescence limit the ability to accurately pick and assign peak positions in the subsequent steps of the binning process.
  • Filtered spectra were binned using the peak pick and bin algorithm with the parameters given in Table XXIII.
  • the sorted cluster diagrams for each sample set and the corresponding cluster assignments for each spectral file were used to identify groups of samples with similar spectra, which was used to identify samples for secondary analyses.
  • Single crystal x-ray data were collected on a Bruker SMART-APEX CCD diffractometer (M. J. Zaworotko, Department of Chemistry, University of South Florida). Lattice parameters were determined from least squares analysis. Reflection data was integrated using the program SAINT. The structure was solved by direct methods and refined by full matrix least squares using the program SHELXTL (Sheldrick, G. M. SHELXTL, Release 5.03; Siemans Analytical X-ray Instruments Inc.: Madison, WI).
  • the co-crystals of the present invention can be characterized, e.g., by the TGA or DSC data or by any one, any two, any three, any four, any five, any six, any seven, any eight, any nine, any ten, or any single integer number of PXRD 2-theta angle peaks or Raman shift peaks listed herein or disclosed in a figure, or by single crystal x-ray diffraction data.
  • celecoxib icotinamide co-crystals were prepared. Celecoxib (100 mg, 0.26 mmol) and nicotinamide (32.0 mg, 0.26 mmol) were each dissolved in acetone (2 mL). The two solutions were mixed and the resulting mixture was allowed to evaporate slowly overnight. The precipitated solid was redissolved in acetone a second time and left to evaporate to dryness. The powder was collected and characterized. Detailed characterization of the celecoxib icotinamide co-crystal is listed in Table XXIV. Fig. 1A shows the PXRD diffractogram after subtraction of background noise. Fig. IB shows the raw PXRD data. Fig.
  • FIG. 2 shows a DSC thermogram of the celecoxib :nicotinamide co- crystal.
  • Fig. 3 shows a TGA thermogram of the celecoxib :nicotinamide co-crystal.
  • Fig. 4 shows a Raman spectrum of the celecoxib:nicotinamide co-crystal.
  • Co-crystals of topiramate and 18-crown-6 were prepared. To topiramate (100 mg, 0.29 mmol) dissolved in diethyl ether (5 mL) was added 18-crown-6 (78 mg, 0.29 mmol) in diethyl ether (5 mL). Upon addition of 18-crown-6, the solution became cloudy and was sonicated for 30 seconds. The solution was left standing for 1 hour and a colorless precipitate was observed. The precipitate was collected, washed with diethyl ether and dried to give a 1 :1 co-crystal of topiramate: 18-crown-6 as a colorless solid.
  • Fig. 8 A shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 8B shows the raw PXRD data.
  • Fig. 9 shows a DSC thermogram of the topiramate: 18-crown-6 co-crystal.
  • Co-crystals of olanzapine and nicotinamide were prepared.
  • a 9-block experiment was designed with 12 solvents.
  • a block is a receiving plate, which can be, for example, an industry standard 24 well, 96 well, 384 well, or 1536 well format, or a custom format.
  • 864 crystallization experiments with 10 co-crystal formers and 3 concentrations were carried out using the CrystalMaxTM platform.
  • Form I was obtained from mixtures containing 1:1 and 1:2 molar ratios of olanzapine:nicotinamide in 1,2- dichloroethane.
  • Form II was obtained from mixtures containing a 1 :2 molar ratio of olanzapine and nicotinamide in isopropyl acetate.
  • PXRD and DSC characterization of the olanzapine:nicotinamide co-crystals are listed in Table XXIV.
  • Fig. 10A shows the PXRD diffractogram of form I after subtraction of background noise.
  • Fig. 10B shows the raw PXRD data of form I.
  • Fig. 11 shows a DSC thermogram of the olanzapine :nicotinamide form I co-crystal.
  • Fig. 12 shows the PXRD diffractogram of olanzapinemicotinamide form II after subtraction of background noise.
  • Co-crystals of olanzapine and nicotinamide were prepared.
  • Olanzapine 40 microliters of 25 mg/mL stock solution in tetrahydrofuran
  • nicotinamide 37.6 microliters of 20 mg/mL stock solution in methanol
  • To the solid mixture was added isopropyl acetate (100 microliters) and the vial was sealed with an aluminum cap.
  • the suspension was then heated at 70 degrees C for two hours in order to dissolve all of the solid material.
  • the solution was then cooled to 5 degrees C and maintained at that temperature for 24 hours.
  • Fig. 13 A shows the PXRD diffractogram of form III after subtraction of background noise.
  • Fig. 13B shows the raw PXRD data of form III.
  • Figs. 14A-D show packing diagrams of the olanzapine:nicotinamide form III co-crystal.
  • the olanzapine :nicotinamide (Form III) co-crystal is made up of a ternary system containing olanzapine, nicotinamide, water and isopropyl acetate in the unit cell.
  • the co-crystal crystallizes in the monoclinic space group P2i/c and contains two olanzapine molecules, one nicotinamide molecule, 4 water molecules and one isopropyl acetate molecule in the asymmetric unit.
  • the packing diagram is made up of a two-dimensional hydrogen-bonded network with the water molecules connecting the olanzapine and nicotinamide moieties.
  • the packing diagram is also comprised of alternating olanzapine and nicotinamide layers connected through hydrogen bonding via the water and isopropyl acetate molecules, as shown in Figure 14B.
  • the olanzapine layer propagates along the b axis at c/4 and 3c/4.
  • the nicotinamide layer propagates along the b axis at c/2.
  • the top of Figure 14C illustrates the nicotinamide superstructure.
  • the nicotinamide molecules form dimers which hydrogen bond to chains of 4 water molecules.
  • the water chains terminate with isopropyl acetate molecules on each side.
  • a co-crystal of cw-itraconazole and succinic acid was prepared.
  • succinic acid (16.8 mg, 0.142 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol).
  • THF tetrahydrofuran
  • a clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form.
  • the solid was collected by filtration and washed with cold THF (2 mL).
  • the white solid was air-dried and placed in a glass vial.
  • the crystalline substance was found to be a succinic acid co-crystal of cz ' s-itraconazole.
  • the solid was characterized by PXRD and DSC.
  • Fig. 15 shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 16 shows a DSC thermogram
  • a co-crystal of czAitraconazole and fumaric acid was prepared.
  • To a blend of fumaric acid (8.40 mg, 0.072 mmol) and czAitraconazole (51.8 mg, 0.073 mmol) was added tetrahydrofuran (THF) (1.0 mL).
  • THF tetrahydrofuran
  • To the clear solution was added t-butyl methyl ether (1.0 mL).
  • a white solid formed immediately and was collected by filtration and washed with cold t-butyl methyl ether (2 mL). The white solid was air-dried and placed in a glass vial.
  • the crystalline substance was found to be a fumaric acid co-crystal of czAitraconazole.
  • the solid was characterized by PXRD and DSC.
  • Fig. 17 shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 18 shows a DSC thermogram of the co-crystal.
  • a co-crystal of czAitraconazole and L-tartaric acid was prepared.
  • L-tartaric acid (21.3 mg, 0.142 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol).
  • THF tetrahydrofuran
  • cis- itraconazole 100 mg, 0.142 mmol
  • a clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form.
  • the solid was collected by filtration and washed with cold THF (2 mL).
  • the white solid was air-dried and placed in a glass vial.
  • the crystalline substance was found to be an L- tartaric acid co-crystal of czAitraconazole.
  • the solid was characterized by PXRD and DSC.
  • Fig. 19 shows the PXRD diffractogram after
  • a co-crystal of czAitraconazole and L-malic acid was prepared.
  • L-malic acid (19.1 mg, 0.143 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol).
  • THF tetrahydrofuran
  • a clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form.
  • the solid was collected by filtration and washed with cold THF (2 mL).
  • the white solid was air-dried and placed in a glass vial.
  • the crystalline substance was found to be an L- malic acid co-crystal of czAitraconazole.
  • the solid was characterized by PXRD and DSC.
  • Fig. 21 shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 22 shows a DSC thermogram of the
  • a co-crystal of czAitraconazole hydrochloride and DL-tartaric acid was prepared.
  • a suspension of c/Aitraconazole freebase (20.1 g, 0.0285 mol) in absolute ethanol (100 mL) was added a solution of hydrochloric acid (1.56 g, 0.0428 mol) and DL-tartaric acid (2.99 g, 0.0171mol) in absolute ethanol (100 mL).
  • a clear solution formed with stirring and heating to reflux.
  • the hot solution was gravity filtered and allowed to cool to room temperature (25 degrees C). Upon cooling white crystals formed.
  • the solid was collected by filtration and washed with cold absolute ethanol (15 mL).
  • the white solid was dried in a vacuum oven overnight at 80 degrees C.
  • the crystalline substance was found to be a DL-tartaric acid co-crystal of ezAitraconazole hydrochloride.
  • the solid was characterized by PXRD and DSC.
  • Fig. 23 shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 24 shows a DSC thermogram of the co-crystal.
  • Co-crystals of modafinil and malonic acid were prepared. Using a 250 mg/ml modafinil-acetic acid solution, malonic acid was dissolved on a hotplate (about 67 degrees C) at a 1 :2 modafinil to malonic acid ratio. The mixture was dried under flowing nitrogen overnight. A powdery white solid was produced. After further drying for 1 day, acetic acid was removed (as determined by TGA) and the crystal structure of the modafinikmalonic acid (Form I) co-crystal, as determined by PXRD, remained the same. The modafinil :malonic acid (Form I) co-crystal was also prepared by grinding the API and co-crystal former together.
  • FIG. 26 shows a DSC thermogram of the modafinil :malonic acid Form I co-crystal.
  • Fig. 27 shows the Raman spectrum of the modafinil :malonic acid Form I co-crystal.
  • Fig. 27 comprises peaks, in order of decreasing intensity, of 1004, 222, 633, 265, 1032, 1183, 814, 1601, 490, 718, 767, 361, 917, 1104, 889, 412, 1225, 1251, 1398, 1442, 1731, 1298, 3065, and 2949 cm "1 .
  • Single crystal data of the modafinikmalonic acid Form I co-crystal were acquired and are reported below.
  • a polymorph of the modafinikmalonic acid Form I co-crystal was prepared in a vial. 11.4 mg of modafinil and 8.9 mg of malonic acid were dissolved in 2 mL of acetone. The solids dissolved at room temperature, and the vial was left open to evaporate the solvent in air. Large parallelogram shaped crystals formed on the walls and bottom of the vial.
  • the PXRD diffractogram of the large crystals showed modafinil :malonic acid co-crystals Form II, a polymorphic form of modafinil :malonic acid Form I.
  • Fig. 28 shows the PXRD diffractogram of the modafinil :malonic acid Form II co-crystal after subtraction of background noise.
  • Co-crystals of modafinil and glycolic acid were prepared. Modafinil (1 mg, 0.0037mmol) and glycolic acid (0.30 mg, 0.0037 mmol) were dissolved in acetone (400 microliters). The solution was allowed to evaporate to dryness and the resulting solid was characterized using PXRD.
  • PXRD data for the modafinil: glycolic acid co-crystal is listed in Table XXIV.
  • Fig. 29A shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 29B shows the raw PXRD data.
  • Co-crystals of modafinil and maleic acid were prepared. Using a 250 mg/ml modafinil-acetic acid solution, maleic acid was dissolved on a hotplate (about 67 degrees C) at a 2:1 modafinil to maleic ratio. The mixture was dried under flowing nitrogen overnight. A clear amorphous material remained. Solids began to grow after 2 days stored in a sealed vial at room temperature. The solid was collected and characterized as the modafinil :maleic acid co-crystal using PXRD. Fig. 30A shows the PXRD diffractogram after subtraction of background noise. Fig. 3 OB shows the raw PXRD data.
  • FIG. 32 shows a DSC thermogram of the 5-fluorouracil:urea co-crystal.
  • Fig. 33 shows a TGA thermogram of the 5-fluorouracil:urea co-crystal.
  • Fig. 34 shows a Raman spectrum of the 5-fluorouracil:urea co-crystal. Single crystal data of the 5-fluorouracil:urea co- crystal were acquired and are reported below.
  • Co-crystals of hydrochlorothiazide and nicotinic acid were prepared. Hydrochlorothiazide (12.2 mg, 0.041 mmol) and nicotinic acid (5 mg, 0.041 mmol) were dissolved in methanol (1 mL). The solution was then cooled to 5 degrees C and maintained at that temperature for 12 hours. A white solid precipitated and was collected and characterized as the hydrochlorothiazide:nicotinic acid co-crystal using PXRD.
  • Fig. 35A shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 35B shows the raw PXRD data.
  • Co-crystals of hydrochlorothiazide and 18-crown-6 were prepared. Hydrochlorothiazide (100 mg, 0.33 mmol) was dissolved in diethyl ether (15 mL) and was added to a solution of 18-crown-6 (87.2 mg, 0.33 mmol) in diethyl ether (15 mL). A white precipitate immediately began to form and was collected and characterized as the hydrochlorothiazide: 18-crown-6 co-crystal using PXRD.
  • Fig. 36A shows the PXRD diffractogram after subtraction of background noise.
  • Fig. 36B shows the raw PXRD data.
  • Crystal packing The co-crystals contain bilayered sheets in which water molecules act as a hydrogen bonded bridge between the network bipyridine moieties and the acetaminophen. Bipyridine guests are sustained by ⁇ - ⁇ stacking interactions between two network bipyridines. The layers stack via ⁇ - ⁇ interactions between the phenyl groups of the acetaminophen moieties.
  • Crystal packing The co-crystal is sustained by hydrogen bonding of adjacent phentoin molecules between the carbonyl and the amine closest to the tetrahedral carbon, and by hydrogen bonding between pyridone carbonyl functionalities and the amine not involved in phenytoin-phenytoin interactions.
  • the pyridone carbonyl also hydrogen bonds with adjacent pyridone molecules forming a one-dimensional network.
  • Infrared Spectroscopy (Nicolet Avatar 320 FTIR), characteristic peaks for the co-crystal were identified as: 2° amine found at 3311cm "1 , carbonyl (ketone) found at 1711cm "1 , olephin peak found at 1390cm "1 .
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), a 29.09% weight loss starting at 192.80 degrees C, 48.72% weight loss starting at 238.27 degrees C, and 18.38% loss starting at 260.17 degrees C followed by complete decomposition.
  • the co-crystal contains the carboxylic acid-pyridine heterodimer that crystallizes in the Pbcn space group.
  • the structure is an inclusion compound containing disordered solvent in the channels.
  • ⁇ - ⁇ stacking of the bipyridine and phenyl groups of the aspirin and hydrophobic interactions contribute to the overall packing interactions.
  • Infrared Spectroscopy (Nicolet Avatar 320 FTIR), characteristic (-COOH) peak at 1679 cm “1 was shifted up and less intense at 1694cm " , where as the lactone peak is shifted down slightly from 1750cm “1 to 1744cm “1 .
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), weight loss of 9% starting at 22.62 degrees C, 49.06% weight loss starting at 102.97 degrees C followed by complete decomposition starting at 209.37 degrees C.
  • Crystal packing The co-crystal contains ibuprofembipyridine heterodimers, sustained by two hydrogen bonded carboxylic acidpyridine supramolecular synthons, arranged in a herringbone motif that packs in the space group P-1.
  • the heterodimer is an extended version of the homodimer and packs to form a two-dimensional network sustained by ⁇ - ⁇ stacking of the bipyridine and phenyl groups of the ibuprofen and hydrophobic interactions from the ibuprofen tails.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), 13.28% weight loss between room temperature and 100.02 degrees C immediately followed by complete decomposition.
  • the co-crystal contains flurbiprofe bipyridine heterodimers, sustained by two hydrogen bonded carboxylic acidpyridine supramolecular synthon, arranged in a herringbone motif that packs in the space group P2j/n.
  • the heterodimer is an extended version of the homodimer and packs to form a two-dimensional network sustained by ⁇ - ⁇ stacking and hydrophobic interactions of the bipyridine and phenyl groups of the flurbiprofen.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), 30.93% weight loss starting at 31.13 degrees C and a 46.26% weight loss starting at 168.74 degrees C followed by complete decomposition.
  • the co-crystal contains flurbiprofen: 1 ,2-bis (4-pyridyl) ethylene heterodimers, sustained by two hydrogen bonded carboxylic acid-pyridine supramolecular synthons, arranged in a herringbone motif that packs in the space group P2]/n.
  • the heterodimer from 1,2-bis (4-pyridyl) ethylene further extends the homodimer relative to example 21 and packs to form a two-dimensional network sustained by ⁇ - ⁇ stacking and hydrophobic interactions of the bipyridine and phenyl groups of the flurbiprofen.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), 91.79% weight loss starting at 133.18 degrees C followed by complete decomposition.
  • Crystal packing The co-crystals contain hydrogen bonded carboxamide homodimers that crystallize in the space group C2/c.
  • the 1° amines of the homodimer are bifurcated to the carbonyl of the/7-phthalaldehyde forming a chain with an adjacent homodimer.
  • the chains pack in a crinkled tape motif sustained by ⁇ - ⁇ interactions between phenyl rings of the carbamazepine.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), 17.66% weight loss starting at 30.33 degrees C then a 17.57% weight loss starting at 100.14 degrees C followed by complete decomposition.
  • PXRD derived from the single crystal data, experimental (calculated): 8.5 (8.7); 10.6 (10.8); 11.9 (12.1); 14.4 (14.7) 15.1 (15.2); 18.0 (18.1); 18.5 (18.2); 19.8 (18.7); 23.7 (24.0); 24.2 (24.2); 26.4 (26.7); 27.6 (27.9); 27.8 (28.2); 28.7 (29.1); 29.3
  • Crystal packing The co-crystals contain hydrogen bonded carboxamide homodimers.
  • the 1° amines are bifurcated to the carbonyl of the nicotinamide on each side of the dimer.
  • the 1° amines of each nicotinamide are hydrogen bonded to the carbonyl of the adjoining dimer.
  • the dimers form chains with ⁇ - ⁇ interactions from the phenyl groups of the carbamazepine.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA), 57.94% weight loss starting at 205.43 degrees C followed by complete decomposition.
  • PXRD Showed analogous peaks to the simulated PXRD derived from the single crystal data.
  • carbamazepine saccharin co-crystals were also prepared via another method.
  • a 12-block experiment was designed with 12 solvents.
  • a block is a receiving plate, which can be an industry standard 96 well, 384 well, or 1536 well format, or a custom format.
  • 1152 crystallization experiments were carried out using the CrystalMaxTM platform.
  • the carbamazepine: saccharin co-crystal was obtained from a mixture of isopropyl acetate and heptane.
  • the resulting co-crystal was characterized by PXRD and DSC and these data are shown in Figures 49 and 50, respectively.
  • the co-crystal prepared from a mixture of isopropyl acetate and heptane may contain impurities such as carbamazepine in free form due to incomplete purification.
  • Crystal packing The co-crystals contain hydrogen bonded carboxamide homodimers.
  • the 2° amines of the saccharin are hydrogen bonded to the carbonyl of the carbamazepine on each side forming a tetramer.
  • the crystal has a space group of P-1 with ⁇ - ⁇ interactions between the phenyl groups of the carbamazepine and the saccharin phenyl groups.
  • PXRD derived from the single crystal data, experimental (calculated): 6.9 (7.0); 12.2 (12.2); 13.6 (13.8); 14.0 (14.1); 14.1 (14.4); 15.3 (15.6); 15.9 (15.9); 18.1 (18.2); 18.7 (18.8); 20.2 (20.3); 21.3 (21.5); 23.7 (23.9); 26.3 (26.4); 28.3 (28.3).
  • Crystal packing Each hydrogen on the carbamazepine 1° amine is hydrogen bonded to a carbonyl group of a different 2,6-pyridinedicarboxylic acid moiety.
  • the carbonyl of the carbamazepine carboxamide is hydrogen bonded to two hydroxide groups of one 2,6-pyridinedicarboxylic acid moiety.
  • Crystal packing The co-crystals are sustained by hydrogen bonded carboxylic acid homodimers between the two 5-nitroisophthalic acid moieties and hydrogen bonded carboxy-amide heterodimers between the carbamazepine and 5-nitroisophthalic acid moiety. There is solvent hydrogen bonded to an additional N-H donor from the carbamazepine moiety.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA). 32.02% weight loss starting at 202 degrees C, a 12.12% weight loss starting at 224 degrees C and a 17.94% weight loss starting at 285 degrees C followed by complete decomposition.
  • PXRD Showed analogous peaks to the simulated PXRD derived from the single crystal data.
  • PXRD analysis experimental (calculated): 10.138 (10.283), 15.291 (15.607), 17.438 (17.791), 21.166 (21.685), 31.407 (31.738), 32.650 (32.729).
  • Crystal packing The co-crystals form a single 3D network of four tetrahedron, linked by square planes similar to the PtS topology. The crystals are sustained by hydrogen bonding.
  • Crystal packing The co-crystals contain hydrogen bonded carboxamide homodimers. Each 1° amine on the carbamazepine is bifurcated to a carbonyl group of a benzoquinone moiety. The dimers form infinite chains.
  • Crystal packing The co-crystals are sustained by hydrogen bonded carboxylic acid homodimers between carbamazepine and trimesic acid moieties and hydrogen bonded carboxylic acid-amine heterodimers between two trimesic acid moieties arranged in a stacked ladder formation.
  • Thermogravimetric Analysis (TA Instruments 2950 Hi-Resolution TGA). 62.83%o weight loss starting at 253 degrees C and a 30.20% weight loss starting at 278 degrees C followed by complete decomposition.
  • PXRD analysis experimental 10.736, 12.087, 16.857, 24.857, 27.857.
  • DSC Two endothermic transitions at about 117 and 119 degrees C and a sharp endotherm at about 130 degrees C
  • TGA Decomposition beginning at about 150 degrees
  • TGA Decomposition above 200 degrees C with a 25% weight loss between about 190-
  • TGA Rapid decomposition beginning at about 135 degrees C and leveling off slightly after 200 degrees C
  • TGA 29.09 percent weight loss starting at about 193 degrees C, 48.72 percent weight loss starting at about 238 degrees C, 18.38 percent weight loss starting at about 260 degrees C
  • TGA 9 percent weight loss starting at about 23 degrees C, 49.06 percent weight loss starting at about 103 degrees C, decomposition starting at about 209 degrees C
  • PXRD 16.8, 17.1, 18.1, 19.0, 20.0, 21.3, 22.7, 25.0, 26.0, 26.1, 28.2, 29.1
  • TGA 30.93 percent weight loss starting at about 31 degrees C, 46.26 percent weight loss starting at about 169 degrees C
  • TGA 17.66 percent weight loss starting at about 30 degrees C, 17.57 percent weight loss starting at about 100 degrees C
  • TGA Decomposition beginning at about 150 degrees
  • TGA 3.342 percent weight loss starting at about 67 degrees C, 55.09 percent weight loss starting at about 119 degrees C
  • TGA 32.02 percent weight loss starting at about 202 degrees C, 12.12 percent weight loss starting at about 224 degrees C, 17.94 percent weight loss starting at about 285 degrees C
  • TGA 9 percent weight loss starting at about 189 degrees C, 52 percent weight loss starting at about 251 degrees C, 31 percent weight loss starting at about 374 degrees C
  • TGA 20.62 percent weight loss starting at about 168 degrees C, 78 percent weight loss starting at about 223 degrees C
  • TGA 62.83 percent weight loss starting at about 253 degrees C, 30.20 percent weight loss starting at about 278 degrees C
  • a co-crystal with a modulated dissolution profile has been prepared.
  • Celecoxib nicotinamide co-crystals were prepared via methods shown in Example 1. (See Fig. 57)
  • a co-crystal with a modulated dissolution profile has been prepared, cis- Itraconazole: succinic acid, cw-itraconazolei-tartaric acid and czAitraconazole:L-malic acid co-crystals were prepared via methods shown in Examples 5, 7 and 8. (See Fig. 58)
  • a co-crystal of an unsaltable or difficult to salt API has been prepared.
  • Celecoxib nicotinamide co-crystals were prepared via methods shown in Example 1.
  • Example 34
  • a co-crystal with an improved hygroscopicity profile has been prepared.
  • Celecoxib nicotinamide co-crystals were prepared via methods shown in Example 1. (See Fig. 59)
  • a co-crystal with reduced form diversity as compared to the API has been prepared.
  • Co-crystals of carbamazepine and saccharin have been prepared via method shown in Example 25.
  • the formulation of a modafinil :malonic acid form I co-crystal was completed using lactose.
  • Two mixtures, one of modafinil and lactose, and the second of modafinil :malonic acid co-crystal and lactose, were ground together in a mortar an pestle.
  • the mixtures targeted a 1 :1 weight ratio of modafinil to lactose.
  • 901.2 mg of modafinil and 901.6 mg of lactose were ground together.
  • 1221.6 mg of co-crystal and 871.4 mg of lactose were ground together.
  • the resulting powders were analyzed by PXRD and DSC.
  • the PXRD patterns and DSC thermograms of the mixtures showed virtually no change upon comparison with both individual components.
  • the DSC of the co-crystal mixture showed only the co-crystal melting peak at 113.6 degrees C with a heat of fusion of 75.9 J/g. This heat of fusion is 59.5 % of that found for the co-crystal alone (127.5 J/g). This result is consistent with a 58.4 % weight ratio of co-crystal in the mixture.
  • the DSC of the modafinil and lactose mixture had a melting point of 165.7 degrees C. This is slightly lower then the measured melting point of modafinil (168.7 degrees C).
  • the heat of fusion of the mixture (59.3 J/g) is 46.9 % that of the modafinil alone (126.6 J/g), which is consistent with the estimated value of 50 %.
  • Modafinil and the modafinikmalonic acid form I co-crystal were passed through a 38 micrometer sieve.
  • Gelatin capsules Size 0, B&B Pharmaceuticals, Lot # 15-01202
  • Dissolution studies were performed in a Vankel VK 7000 Benchsaver Dissolution Testing Apparatus with the VK750D heater/circulator set at 37 degrees C. At 0 minutes, the capsules were dropped into vessels containing 900 mL 0.01 M HCl and stirred by paddles.
  • Absorbance readings were taken using a Cary 50 Spectrophotometer (wavelength set at 260nm) at the following time points: 0, 5, 10, 15, 20, 25, 30, 40, 50, and 60 minutes. The absorbance values were compared to those of standards and the modafinil concentrations of the solutions were calculated.
  • Modafinil and the modafinil :malonic acid form I co-crystal were mixed with equivalent amounts of lactose (Spectrum, Lot QV0460) for approximately 5 minutes.
  • Gelatin capsules Size 0, B&B Pharmaceuticals, Lot # 15-01202 were filled with 400.2 mg modafinil and lactose (approximately 200 mg modafinil), or 561.0 mg modafinil :malonic acid form I co-crystal and lactose (approximately 200 mg modafinil).
  • HPMC capsules (Size 0, Shionogi, Lot # A312A6) were filled with 399.9 mg modafinil and lactose, 560.9 mg modafinikmalonic acid co-crystal and lactose, 199.9 mg modafinil, or 280.5 mg modafinil :malonic acid form I co-crystal.
  • the dissolution study was carried out as described above.
  • the modafinikmalonic acid form I co-crystal (from Example 10) was administered to dogs in a pharmacokinetic study.
  • Particles of modafinil :malonic acid co- crystal with a median particle size of about 16 micrometers were administered in the study.
  • micronized modafinil with a median particle size of about 2 micrometers was also administered in the study.
  • the AUC of the modafinil :malonic acid co-crystal was determined to be 40 to 60 percent higher than that of the pure modafinil.
  • Such a higher bioavailability illustrates the modulation of an important pharmacokinetic parameter due to an embodiment of the present invention.
  • a compilation of important pharmacokinetic parameters measured during the animal study are included in Table XXV.
  • the increased half-life and bioavailability of modafinil in the malonic acid form I co-crystal may be due to the presence of malonic acid. It is believed that the malonic acid may be inhibiting one or more pathways responsible for the metabolism or elimination of modafinil. It is noted that modafinil and malonic acid share a similar structure: each including two carbonyl or sulfonyl groups separated by a -CH 2 - and each molecule is terminated with a group that is capable of participation in a hydrogen bond with an enzyme. Such a mechanism may take place with other APIs or co-crystal formers of similar structure.
  • the stability of the modafinikmalonic acid form I co-crystal was measured at various temperatures and relative humidities over a four week period. No degradation was found to occur at 20 or 0 degrees C. At 60 degrees C, about 0.14 percent degradation per day was determined based on a simple exponential model. At 80 degrees C, about 8 percent degradation per day was determined. TABLE I

Abstract

A pharmaceutical composition comprising a co-crystal of an API and a co-crystal former; wherein the API has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphinic acid, phosphonic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, 0-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, pyridine and the co-crystal former has at least one functional group selected from amine, amide, pyridine, imidazole, indole, pyrrolidine, carbonyl, carboxyl, hydroxyl, phenol, sulfone, sulfonyl, mercapto and methyl thio, such that the API and co-crystal former are capable of co-crystallizing from a solution phase under crystallization conditions.

Description

PHARMACEUTICAL CO-CRYSTAL COMPOSITIONS
Cross-Reference to Related Applications
This application is a continuation-in-part of United States Patent Application 10/660,202, filed September 1 1, 2003 (which claims the benefit of US Provisional Patent Application No. 60/451,213 filed on February 28, 2003; U.S. Provisional Patent Application No. 60/463,962, filed on April 18, 2003; and U.S. Provisional Application No. 60/487,064, filed on July 11, 2003 each of which incorporated herein by reference in its entirety for all purposes.
This application is also a continuation-in-part of PCT US03/27772, filed on September 4, 2003 which is a continuation-in-part of U.S. Patent Application No. 10/378,956, filed March 1, 2003, which claims the benefit of U.S. Provisional Application No. 60/360,768, filed March 1, 2002; said PCT US03/27772 also claims the benefit of US Provisional Patent Application No. 60/451,213 filed on February 28, 2003; U.S. Provisional Patent Application No. 60/463,962, filed on April 18, 2003; and U.S. Provisional Application No. 60/487,064, filed on July 11, 2003 each of which are hereby incorporated by reference in its entirety for all purposes.
Said 10/660,202 and PCT US03/27772 are also continuations-in-part of U.S. Patent Application No. 10/637,829, filed August 8, 2003, which is a divisional of U.S. Patent Application No. 10/295,995, filed November 18, 2002, which is a continuation of U.S. Patent Application No.10/232,589, filed September 3, 2002, which claims the benefit of US Provisional Patent Application No. 60/406,974, filed August 30, 2002 and US Provisional Patent Application No.60/380,288, filed May 15, 2002 and US Provisional Patent Application No. 60/356,764, filed February 15, 2002 each of which are hereby incorporated by reference in its entirety for all purposes.
Said 10/660,202 and PCT US03/27772 are also continuations-in-part of US Patent Application No. 10/449,307, filed May 30, 2003 which claims the benefit of US Provisional Patent Application No. 60/463,962 filed April 18, 2003 and US Provisional Patent Application No. 60/444,315, filed January 31, 2003 and US Provisional Patent Application No. 60/439,282 filed January 10, 2003 and US Provisional Patent Application No. 60/384,152, filed May 31, 2002 each of which are hereby incorporated by reference in its entirety for all purposes.
Said 10/660,202 and PCT US03/27772 are also continuations-in-part of US Patent Application No. 10/601,092, filed June 20, 2003, which claims the benefit of US Provisional Patent Application No. 60/451,213, filed February 28, 2003 each of which are hereby incorporated by reference in its entirety for all purposes.
This application is also a continuation-in-part of U.S. Patent Application No. 10/637,829, filed August 8, 2003, which is a divisional of U.S. Patent Application No. 10/295,995, filed November 18, 2002, which is a continuation of U.S. Patent Application No.10/232,589, filed September 3, 2002, which claims, the benefit of US Provisional Patent Application No. 60/406,974, filed August 30, 2002 and US Provisional Patent Application No.60/380,288, filed May 15, 2002 and US Provisional Patent Application No. 60/356,764, filed February 15, 2002 each of which are hereby incorporated by reference in its entirety for all purposes.
This application is also a continuation-in-part of US Patent Application No. 10/449,307, filed May 30, 2003 which claims the benefit of US Provisional Patent Application , No. 60/463,962 filed April 18, 2003 and US Provisional Patent Application No. 60/444,315, filed January 31, 2003 and US Provisional Patent Application No. 60/439,282 filed January 10, 2003 and US Provisional Patent Application No. 60/384,152, filed May 31, 2002 each of which are hereby incorporated by reference in its entirety for all purposes.
This application is also a continuation-in-part of US Patent Application No. 10/601,092, filed June 20, 2003, which claims the benefit of TJS Provisional Patent Application No. 60/451,213, filed February 28, 2003 each of which are hereby incorporated by reference in its entirety for all purposes.
This application claims benefit of United States Provisional Patent Application 60/508,208, filed October 2, 2003 and United States Provisional Patent Application 60/542,752, filed February 6, 2004 (Entitled: "Modafinil Compositions"; having Docket TPIP044A+; Magali B. Hickey, Matthew Peterson, Om Almarsson, and Mark Oliveira) each of which are hereby incorporated by reference in its entirety for all purposes.
This application is also a continuation-in-part of PCT/US03/41273, filed December 24, 2003, which is a continuation in part of PCT/03/19584, filed June 20, 2003, which claims the benefit of U.S. Provisional Application No. 60/390,881, filed on June 21, 2002, U.S. Provisional Application No. 60/426,275, filed on November 14, 2002; U.S. Provisional Application No. 60/427,086 filed on November 15, 2002; U.S. Provisional Application No. 60/429,515 filed on November 26, 2002; U.S. Provisional Application No. 60/437,516 filed on December 30, 2002; and U.S. Provisional Application No. 60/456,027 filed on March 18, 2003 each which are hereby incorporated by reference in its entirety for all purposes.
This application is also a continuation-in-part of United States Patent Application 10/601,092, filed June 20, 2003 which claims the benefit of U.S. Provisional Application No. 60/390,881, filed on June 21, 2002, U.S. Provisional Application No. 60/426,275, filed on November 14, 2002; U.S. Provisional Application No. 60/427,086 filed on November 15, 2002; U.S. Provisional Application No. 60/429,515 filed on November 26, 2002; U.S. Provisional Application No. 60/437,516 filed on December 30, 2002; and U.S. Provisional Application No. 60/456,027 filed on March 18, 2003 each of which are hereby incorporated by reference in its entirety for all purposes.
FIELD OF THE INVENTION
The present mvention relates to co-crystal API-containing compositions, pharmaceutical compositions comprising such APIs, and methods for preparing the same.
BACKGROUND OF THE INVENTION
Active pharmaceutical ingredients (API or APIs (plural)) in pharmaceutical compositions can be prepared in a variety of different forms. Such APIs can be prepared so as to have a variety of different chemical forms including chemical derivatives or salts. Such APIs can also be prepared to have different physical forms. For example, the APIs may be amorphous, may have different crystalline polymorphs, or may exist in different solvation or hydration states. By varying the form of an API, it is possible to vary the physical properties thereof. For example, crystalline polymorphs typically have different solubilities from one another, such that a more thermodynamically stable polymorph is less soluble than a less thermodynamically stable polymorph. Pharmaceutical polymorphs can also differ in properties such as shelf-life, bioavailability, morphology, vapour pressure, density, colour, and compressibility. Accordingly, variation of the crystalline state of an API is one of many ways in which to modulate the physical properties thereof.
It would be advantageous to have new forms of these APIs that have improved properties, in particular, as oral formulations. Specifically, it is desirable to identify improved forms of APIs that exhibit significantly improved properties including increased aqueous solubility and stability. Further, it is desirable to improve the processability, or preparation of pharmaceutical formulations. For example, needle-like crystal forms or habits of APIs can cause aggregation, even in compositions where the API is mixed with other substances, such that a non- uniform mixture is obtained. It is also desirable to increase or decrease the dissolution rate of API-containing pharmaceutical compositions in water, increase or decrease the bioavailability of orally-administered compositions, and provide a more rapid or more delayed onset to therapeutic effect. It is also desirable to have a form of the API which, when administered to a subject, reaches a peak plasma level faster or slower, has a longer lasting therapeutic plasma concentration, and higher or lower overall exposure when compared to equivalent amounts of the API in its presently-known form. The improved properties discussed above can be altered in a way which is most beneficial to a specific API for a specific therapeutic effect. SUMMARY OF THE INVENTION
It has now been found that new co-crystalline forms of APIs can be obtained which improve the properties of APIs as compared to such APIs in a non-co-crystalline slate (free acid, free base, zwitter ions, salts, etc.).
Accordingly, in a first aspect, the present invention provides a co-crystal pharmaceutical composition comprising an API compound and a co-crystal former, such that the API and co- crystal former are capable of co-crystallizing from a solid or solution phase under crystallization conditions.
Another aspect of the present invention provides a process for the production of a pharmaceutical composition, which process comprises:
(1) providing an API which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(2) providing a co-crystal former which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(3) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions;
(4) isolating co-crystals formed thereby; and
(5) incorporating the co-crystals into a pharmaceutical composition.
A further aspect of the present invention provides a process for the production of a pharmaceutical composition, which comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution an API compound with a co-crystal former, under crystallization conditions, so as to form a solid phase; (2) isolating co-crystals comprising the API and the co-crystal former; and
(3) incorporating the co-crystals into a pharmaceutical composition.
In a further aspect, the present invention provides a process for the production of a pharmaceutical composition, which comprises:
(1) providing (f) an API or a plurality of different APIs, and (ii) a co-crystal former or a plurality of different co-crystal formers, wherein at least one of the APIs and the co-crystal formers is provided as a plurality thereof;
(2) isolating co-crystals comprising the API and the co-crystal former; and
(3) incorporating the co-crystals into a pharmaceutical composition.
Solubility Modulation
In a further aspect, the present invention provides a process for modulating the solubility of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Dissolution Modulation
In a further aspect, the present invention provides a process for modulating the dissolution of an API, whereby the aqueous dissolution rate or the dissolution rate in simulated gastric fluid or in simulated intestinal fluid, or in a solvent or plurality of solvents is increased or decreased, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former. In one embodiment, the dissolution of the API is increased.
Bioavailability Modulation
In a further aspect, the present invention provides a process for modulating the bioavailability of an API, whereby the AUC is increased, the time to Tmax is reduced, the length of time the concentration of the API is above ' _ Tmax is increased, or Cmax is increased, which process comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Dose Response Modulation
In a further aspect the present invention provides a process for improving the linearity of a dose response of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution an API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Increased Stability
In a still further aspect the present invention provides a process for improving the stability of a pharmaceutical salt, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the pharmaceutical salt with a co-crystal former under crystallization conditions, so as to form a co- crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Difficult to Salt or Unsaltable Compounds
In a still further aspect the present invention provides a process for making co-crystals of difficult to salt or unsaltable APIs, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Decreasing Hygroscopicity
In a still further aspect the present invention provides a method for decreasing the hygroscopicity of an API, which method comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Crystallizing Amorphous Compounds
In a still further embodiment aspect the present invention provides a process for crystallizing an amorphous compound, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Decreasing Form Diversity
In a still further embodiment aspect the present invention provides a process for reducing the form diversity of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Morphology Modulation
In a still further embodiment aspect the present invention provides a process for modifying the morphology of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
In a further aspect, the present invention provides a co-crystal composition comprising a co-crystal, wherein said co-crystal comprises an API compound and a co-crystal former. In further embodiments the co-crystal has an improved property as compared to the free form (including a free acid, free base, zwitter ion, hydrate, solvate, etc.) or a salt (which includes salt hydrates and solvates). In further embodiments, the improved property is selected from the group consisting of: increased solubility, increased dissolution, increased bioavailability, increased dose response, decreased hygroscopicity, a crystalline form of a normally amorphous compound, a crystalline form of a difficult to salt or unsaltable compound, decreased form diversity, more desired morphology, or other property described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
Figs. I A-B PXRD diffractograms of a co-crystal comprising celecoxib and nicotinamide, with the background removed and as collected, respectively.
Fig. 2 DSC thermogram for a co-crystal comprising celecoxib and nicotinamide.
Fig. 3 TGA thermogram for a co-crystal comprising celecoxib and nicotinamide.
Fig. 4 Raman spectrum for a co-crystal comprising celecoxib and nicotinamide.
Figs. 5A-B PXRD diffractograms of a co-crystal comprising celecoxib and 18-crown-6, with the background removed and as collected, respectively.
Fig. 6 DSC thermogram for a co-crystal comprising celecoxib and 18-crown-6.
Fig. 7 TGA thermogram for a co-crystal comprising celecoxib and 18-crown-6.
Figs. 8A-B PXRD diffractograms of a co-crystal comprising topiramate and 18-crown-6, with the background removed and as collected, respectively.
Fig. 9 DSC thermogram for a co-crystal comprising topiramate and 18-crown-6.
Figs. 10A-B PXRD diffractograms of a co-crystal comprising olanzapine and nicotinamide (Form
I), with the background removed and as collected, respectively.
Fig. 11 DSC thermogram for a co-crystal comprising olanzapine and nicotinamide (Form I).
Fig. 12 PXRD diffractogram of a co-crystal comprising olanzapine and nicotinamide (Form II).
Figs. 13A-B PXRD diffractograms of a co-crystal comprising olanzapine and nicotinamide (Form
III), with the background removed and as collected, respectively.
Figs. 14A-D Packing diagrams and crystal structure of a co-crystal comprising olanzapine and nicotinamide (Form III).
Fig. 15 PXRD diffractogram of a co-crystal comprising cώ-itraconazole and succinic acid.
Fig. 16 DSC thermogram for a co-crystal comprising cώ-itraconazole and succinic acid.
Fig. 17 PXRD diffractogram of a co-crystal comprising c/s-itraconazole and fumaric acid.
Fig. 18 DSC thermogram for a co-crystal comprising CM-itraconazole and fumaric acid.
Fig. 19 PXRD diffractogram of a co-crystal comprising czAitraconazole and L-tartaric acid.
Fig. 20 DSC thermogram for a co-crystal comprising cώ-itraconazole and L-tartaric acid.
Fig. 21 PXRD diffractogram of a co-crystal comprising cw-itraconazole and L-malic acid.
Fig. 22 DSC thermogram for a co-crystal comprising c/'s-itraconazole and L-malic acid.
Fig. 23 PXRD diffractogram of a co-crystal comprising cw-itraconazoleHCl and DL-tartaric acid. Fig 24 DSC thermogram for a co-crystal comprising cώ-itraconazoleHCl and DL-tartaric acid.
Fig. 25 PXRD diffractogram of a co-crystal comprising modafinil and malonic acid (Form I).
Fig. 26 DSC thermogram for a co-crystal comprising modafinil and malonic acid (Form I).
Fig. 27 Raman spectrum for a co-crystal comprising modafinil and malonic acid (Form I).
Fig. 28 PXRD diffractogram of a co-crystal comprising modafinil and malonic acid (Form II).
Figs. 29A-B PXRD diffractograms of a co-crystal comprising modafinil and glycolic acid, with the background removed and as collected, respectively.
Figs. 30A-B PXRD diffractograms of a co-crystal comprising modafinil and maleic acid, with the background removed and as collected, respectively.
Figs. 31 A-B PXRD diffractograms of a co-crystal comprising 5-fluorouracil and urea, with the background removed and as collected, respectively.
Fig. 32 DSC thermogram for a co-crystal comprising 5-ftuorouracil and urea.
Fig. 33 TGA thermogram for a co-crystal comprising 5-fluorouracil and urea.
Fig. 34 Raman spectrum for a co-crystal comprising 5-fluorouracil and urea.
Figs. 35 A-B PXRD diffractograms of a co-crystal comprising hydrochlorothiazide and nicotinic acid, with the background removed and as collected, respectively.
Figs. 36A-B PXRD diffractograms of a co-crystal comprising hydrochlorothiazide and 18-crown-
6, with the background removed and as collected, respectively.
Figs. 37A-B PXRD diffractograms of a co-crystal comprising hydrochlorothiazide and piperazine, with the background removed and as collected, respectively.
Figs. 38A-B An acetaminophen 1-D polymeric chain and a co-crystal of acetaminophen and 4,4'- bipyridine, respectively.
Figs. 39A-B Pure phenytoin and a co-crystal with phenytoin and pyridone, respectively.
Figs. 4 OA-D Pure aspirin and the corresponding crystal structure are shown in Figures 40A and
40B, respectively. Figures 40C and 40D show the supramolecular entity containing the synthon and corresponding co-crystal of aspirin and 4,4'-bipyridine, respectively.
Figs. 41A-D Pure ibuprofen and the corresponding crystal structure are shown in Figures 41A and 4 IB, respectively. Figures 41C and 41D show the supramolecular entity containing the synthon and corresponding co-crystal of ibuprofen and 4,4'-bipyridine, respectively.
Figs. 2A-D Pure flurbiprofen and the corresponding crystal structure are shown in Figures 42A and 2B, respectively. Figures 42C and 42D show the supramolecular synthon and corresponding co-crystal of flurbiprofen and 4,4'-bipyridine, respectively.
Figs. 43A-B The supramolecular entity containing the synthon and the corresponding co-crystal structure of flurbiprofen and trans- l,2-bis(4-pyridyl)ethylene, respectively.
Figs. 44A-B The crystal structure of pure carbamazepine and the co-crystal structure of carbamazepine and -phthalaldehyde, respectively.
Fig. 45 A packing diagram of the co-crystal structure of carbamazepine and nicotinamide.
Fig. 46 PXRD diffractogram of a co-crystal comprising carbamazepine and nicotinamide.
Fig. 47 DSC thermogram for a co-crystal comprising carbamazepine and nicotinamide.
Fig. 48 A packing diagram of the co-crystal structure of carbamazepine and saccharin.
Fig. 49 PXRD diffractogram of a co-crystal comprising carbamazepine and saccharin.
Fig. 50 DSC thermogram for a co-crystal comprising carbamazepine and saccharin.
Figs. 51 A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 2,6-pyridinedicarboxylic acid, respectively.
Figs. 52A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 5-nitroisophthalic acid, respectively.
Figs. 53 A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and 1,3,5,7-adamantanetetracarboxylic acid, respectively.
Figs. 54A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepme and benzoquinone, respectively.
Figs. 55A-B The crystal structure of carbamazepine and the co-crystal structure of carbamazepine and trimesic acid, respectively.
Fig. 56 PXRD diffractogram of a co-crystal comprising carbamazepine and trimesic acid.
Fig. 57 Dissolution profile for a co-crystal of celecoxib :nicotinamide vs. celecoxib free acid.
Fig. 58 Dissolution profile for co-crystals of itraconazole: succinic acid, itraconazle:tartaric acid and itraconazole:malic acid vs. itraconazole free base.
Fig. 59 Hygroscopicity profile for a co-crystal of celecoxib:nicotinamide vs. celecoxib sodium.
Fig. 60 Hydrogen-bonding motifs observed in co-crystals.
Fig. 61 Dissolution profile of several formulations of modafinil free form and modafinikmalonic acid (Form I).
DETAILED DESCRIPTION OF THE INVENTION
The term "co-crystal" as used herein means a crystalline material comprised of two or more unique solids at room temperature, each containing distinctive physical characteristics, such as structure, melting point and heats of fusion, with the exception that, if specifically stated, the API may be a liquid at room temperature. The co-crystals of the present invention comprise a co-crystal former H-bonded to an API. The co- crystal former may be H-bonded directly to the API or may be H-bonded to an additional molecule which is bound to the API. The additional molecule may be H-bonded to the API or bound ionically or covalently to the API. The additional molecule could also be a different API. Solvates of API compounds that do not further comprise a co-crystal former are not co-crystals according to the present invention. The co-crystals may however, include one or more solvate molecules in the crystalline lattice. That is, solvates of co-crystals, or a co-crystal further comprising a solvent or compound that is a liquid at room temperature, is included in the present invention, but crystalline material comprised of only one solid and one or more liquids (at room temperature) are not included in the present invention, with the previously noted exception of specifically stated liquid APIs. The co-crystals may also be a co-crystal between a co-crystal former and a salt of an API, but the API and the co-crystal former of the present invention are constructed or bonded together through hydrogen bonds. Other modes of molecular recognition may also be present including, pi-stacking, guest-host complexation and van der Waals interactions. Of the interactions listed above, hydrogen-bonding is the dominant interaction in the formation of the co-crystal, (and a required interaction according to the present invention) whereby a non-covalent bond is formed between a hydrogen bond donor of one of the moieties and a hydrogen bond acceptor of the other. Hydrogen bonding can result in several different intermolecular configurations. For example, hydrogen bonds can result in the formation of dimers, linear chains, or cyclic structures. These configurations can further include extended (two-dimensional) hydrogen bond networks and isolated triads (Fig. 60). An alternative embodiment provides for a co-crystal wherein the co-crystal former is a second API. In another embodiment, the co-crystal former is not an API. In another embodiment the co-crystal comprises two co-crystal formers. For purposes of the present invention, the chemical and physical properties of an API in the form of a co-crystal may be compared to a reference compound that is the same API in a different form. The reference compound may be specified as a free form, or more specifically, a free acid, free base, or zwitterion; a salt, or more specifically for example, an inorganic base addition salt such as sodium, potassium, lithium, calcium, magnesium, ammonium, aluminum salts or organic base addition salts, or an inorganic acid addition salts such as HBr, HCl, sulfuric, nitric, or phosphoric acid addition salts or an organic acid addition salt such as acetic, propionic, pyruvic, malanic, succinic, malic, maleic, fumaric, tartaric, citric, benzoic, methanesulfonic, ethanesulforic, stearic or lactic acid addition salt; an anhydrate or hydrate of a free form or salt, or more specifically, for example, a hemihydrate, monohydrate, dihydrate, trihydrate, quadrahydrate, pentahydrate, sesquihydrate; or a solvate of a free form or salt. For example, the reference compound for an API in salt form co-crystallized with a co-crystal former can be the API salt form. Similarly, the reference compound for a free acid API co-crystallized with a co-crystal former can be the free acid API. The reference compound may also be specified as crystalline or amorphous.
According to the present invention, the co-crystals can include an acid addition salt or base addition salt of an API. Acid addition salts include, but are not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid, and organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, madelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2- hydroxyethanesulfonic acid, benzenesulfonic acid,/?-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4- methylbicyclo[2.2.2]oct-2-ene-l -carboxylic acid, glucoheptonic acid, 4,4'- methylenebis(3-hydroxy-2-ene-l -carboxylic acid), 3-phenylpropionic acid, trirnefhylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutaric acid, hydroxynaphthoic acid, salicylic acid, stearic acid, and muconic acid. Base addition salts include, but are not limited to, inorganic bases such as sodium, potassium, lithium, ammonium, calcium and magnesium salts, and organic bases such as primary, secondary and tertiary amines (e.g. isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, and N-ethylpiperidine).
The ratio of API to co-crystal former may be stoichiometric or non-stoichiometric according to the present invention. For example, 1 :1, 1.5:1, 1 :1.5, 2:1 and 1 :2 ratios of API:co-crystal former are acceptable.
It has surprisingly been found that when an API and a selected co-crystal former are allowed to form co-crystals, the resulting co-crystals give rise to improved properties of the API, as compared to the API in a free form (including free acids, free bases, and zwitterions, hydrates, solvates, etc.), or an acid or base salt thereof particularly with respect to: solubility, dissolution, bioavailability, stability, Cmax, Tmax, processability, longer lasting therapeutic plasma concentration, hygroscopicity, crystallization of amorphous compounds, decrease in form diversity (including polymorphism and crystal habit), change in morphology or crystal habit, etc. For example, a co-crystal form of an API is particularly advantageous where the original API is insoluble or sparingly soluble in water. Additionally, the co-crystal properties conferred upon the API are also useful because the bioavailability of the API can be improved and the plasma concentration and/or serum concentration of the API can be improved. This is particularly advantageous for orally-administrable formulations. Moreover, the dose response of the API can be improved, for example by increasing the maximum attainable response and/or increasing the potency of the API by increasing the biological activity per dosing equivalent.
Accordingly, in a first aspect, the present invention provides a pharmaceutical composition comprising a co-crystal of an API and a co-crystal former, such that the API and co-crystal former are capable of co-crystallizing from a solution phase under crystallization conditions or from the solid-state, for example, through grinding, heating, or through vapor transfer (e.g., co-sublimation). In another aspect, the API has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine and a co-crystal former which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine, or a functional group in a Table herein, such that the API and co-crystal former are capable of co-crystallizing from a solution phase under crystallization conditions.
The co-crystals of the present invention are formed where the API and co-crystal former are bonded together through hydrogen bonds. Other non-covalent interactions, including pi-stacking and van der Waals interactions, may also be present.
In one embodiment, the co-crystal former is selected from the co-crystal formers of Table I and Table II. In other embodiments, the co-crystal former of Table I is specified as a Class 1, Class 2, or Class 3 co-crystal former (see column labeled "class" Table I). In another embodiment, the difference in pKa value of the co-crystal former and the API is less than 2. In other embodiments, the difference in pKa values of the co- crystal former and API is less than 3, less than 4, less than 5, between 2 and 3, between 3 and 4, or between 4 and 5. Table I lists multiple pKa values for co-crystal formers having multiple functionalities. It is readily apparent to one skilled in the art the particular functional group corresponding to a particular pKa value.
In another embodiment the particular functional group of a co-crystal former interacting with the API is specified (see for example Table I, columns labeled "Functionality" and "Molecular Structure" and the column of Table II labeled "Co- Crystal Former Functional Group"). In a further embodiment the functional group of the API interacting with the co-crystal former functional group is specified (see, for example, Tables II and III).
In another embodiment, the co-crystal comprises more than one co-crystal former. For example, two, three, four, five, or more co-crystal formers can be incorporated in a co-crystal with an API. Co-crystals which comprise two or more co-crystal formers and an API are bound together via hydrogen bonds. In one embodiment, incorporated co- crystal formers are hydrogen bonded to the API molecules. In another embodiment, co- crystal formers are hydrogen bonded to either the API molecules or the incorporated co- crystal formers.
In a further embodiment, several co-crystal formers can be contained in a single compartment, or kit, for ease in screening an API for potential co-crystal species. The co-crystal kit can comprise 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or more of the co-crystal formers in Tables I and II. The co-crystal formers are in solid form or in solution and in an array of individual reaction vials such that individual co-crystal formers can be tested with one or more APIs by one or more crystallization methods or multiple co-crystal formers can be easily tested against one or more compounds by one or more crystallization methods. The crystallization methods include, but are not limited to, melt recrystallization, grinding, milling, standing, co-crystal formation from solution by evaporation, thermally driven crystallization from solution, co-crystal formation from solution by addition of anti-solvent, co-crystal formation from solution by vapor- diffusion, co-crystal formation from solution by drown-out, co-crystal formation from solution by any combination of the above mentioned techniques, co-crystal formation by co-sublimation, co-crystal formation by sublimation using a Knudsen cell apparatus, co- crystal formation by standing the desired components of the co-crystal in the presence of solvent vapor, co-crystal formation by slurry conversion of the desired components of the co-crystal in a solvent or mixtures of solvents, or co-crystal formation by any combination of the above techniques in the presence of additives, nucleates, crystallization enhancers, precipitants, chemical stabilizers, or anti-oxidants. The co- crystallization kits can be used alone or as part of larger crystallization experiments. For example, kits can be constructed as single co-crystal former single well kits, single co- crystal former multi-well kits, multi-co-crystal former single well kits, or multi-co-crystal former multi-well kits. High-throughput crystallization (e.g., the CrystalMax™ platform) can be used to construct and customize co-crystal former kits. Multi-well plates (e.g., 96 wells, 384 wells, 1536 wells, etc.), for example, can be used to store or employ an array of co-crystal formers.
In a further embodiment, the API is selected from an API of Table IV or elsewhere herein. For pharmaceuticals listed in Table IV, co-crystals can comprise such APIs in free form (i.e. free acid, free base, zwitter ion), salts, solvates, hydrates, or the like. For APIs in Table IV listed as salts, solvates, hydrates, and the like, the API can either be of the form listed in Table IV or its corresponding free form, or of another form that is not listed. Table IV includes the CAS number, chemical name, or a PCT or patent reference (each incorporated herein in their entireties). In further embodiments, the functional group of the particular API interacting with the co-crystal former is specified. A specific functional group of a co-crystal former, a specific co-crystal former, or a specified functional group or a specific co-crystal former interacting with the particular API may also be specified. It is noted that for Table II, the co-crystal former, and optionally the specific functionality, and each of the listed corresponding interacting groups are included as individual species of the present invention. Thus, each specific combination of a co-crystal former and one of the interacting groups in the same row may be specified as a species of the present invention. The same is true for other combinations as discussed in the Tables and elsewhere herein.
In another embodiment of the present invention, the co-crystal comprises an API wherein the API forms a dimeric primary amide structure via hydrogen bonds with an R2 2 (8) motif. In such a structure, the NH2 moiety can also participate in a hydrogen bond with a donor or an acceptor moiety from, for example, a co-crystal former or an additional (third) molecule, and the C=O moiety can participate in a hydrogen bond with a donor moiety from the co-crystal former or the additional molecule. In a further embodiment, the dimeric primary amide structure further comprises one, two, three, or four hydrogen bond donors. In a further embodiment, the dimeric primary amide structure further comprises one or two hydrogen bond acceptors. In a further embodiment, the dimeric primary amide structure further comprises a combination of hydrogen bond donors and acceptors. For example, the dimeric primary amide structure can further comprise one hydrogen bond donor and one hydrogen bond acceptor, one hydrogen bond donor and two hydrogen bond acceptors, two hydrogen bond donors and one hydrogen bond acceptor, two hydrogen bond donors and two hydrogen bond acceptors, or three hydrogen bond donors and one hydrogen bond acceptor. Two non- limiting examples of APIs which form a dimeric primary amide co-crystal structure include modafinil and carbamazepine. Some examples of APIs which include a primary amide functional group include, but are not limited to, arotinolol, atenolol, carpipramine, cefotetan, cefsulodin, docapromine, darifenacin, exalamide, fidarestat, frovatriptan, silodosin, levetiracetam, MEN- 10700, mizoribine, oxiracetam, piracetam, protirelin, TRH, ribavirm, valrecemide, temozolomide, tiazofurin, antiPARP-2, levovirin, N- benzyloxycarbonyl glycinamide, and UCB-34714.
In each process according to the invention, there is a need to contact the API with the co-crystal former. This may involve grinding or milling the two solids together or melting one or both components and allowing them to recrystallize. The use of a granulating liquid may improve or may impede co-crystal formation. Non-limiting examples of tools useful for the formation of co-crystals may include, for example, an extruder or a mortar and pestle. Further, contacting the API with the co-crystal former may also involve either solubilizing the API and adding the co-crystal former, or solubilizing the co-crystal former and adding the API. Crystallization conditions are applied to the API and co-crystal former. This may entail altering a property of the solution, such as pH or temperature and may require concentration of the solute, usually by removal of the solvent, typically by drying the solution. Solvent removal results in the concentration of both API and co-crystal former increasing over time so as to facilitate crystallization. For example, evaporation, cooling, co-sublimation, or the addition of an antisolvent may be used to crystallize co-crystals. In another embodiment, a slurry comprising an API and a co-crystal former is used to form co-crystals. Once the solid phase comprising any crystals is formed, this may be tested as described herein.
The manufacture of co-crystals on a large and/or commercial scale may be successfully completed using one or more of the processes and techniques described herein. For example, crystallization of co-crystals from a solvent and grinding or milling are conceivable non-limiting processes.
In another embodiment, the use of an excess (more than 1 molar equivalent for a 1 : 1 co-crystal) of a co-crystal former has been shown to drive the formation of stoichiometric co-crystals. For example, co-crystals with stoichiometries of 1:1, 2:1, or 1 :2 can be produced by adding co-crystal former in an amount that is 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 50, 75, 100 times or more than the stoichiometric amount for a given co- crystal. Such an excessive use of a co-crystal former to form a co-crystal can be employed in solution or when grinding an API and a co-crystal former to drive co-crystal formation.
In another embodiment, the present invention provides for the use of an ionic liquid as a medium for the formation of a co-crystal, and can also be used to crystallize other forms in addition to co-crystals (e.g., salts, solvates, free acid, free base, zwitterions, etc.). This medium is useful, for example, where the above methods do not work or are difficult or impossible to control. Several non-limiting examples of ionic liquids useful in co-crystal formation are: l-butyl-3-methylimidazolium lactate, 1-ethyl- 3-methylimidazolium lactate, and 1-butylpyridinium hexafluorophosphate. The co-crystals obtained as a result of one or more of the above processes or techniques may be readily incorporated into a pharmaceutical composition by conventional means. Pharmaceutical compositions in general are discussed in further detail below and may further comprise a pharmaceutically-acceptable diluent, excipient or carrier.
In a further aspect, the present invention provides a process for the production of a pharmaceutical composition, which process comprises:
(1) providing an API which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine or of Table II or III;
(2) providing a co-crystal former which has at least one functional group selected from ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O- heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine or of Table I, II, or III; (3) grinding, heating or contacting in solution the API with the co-crystal former under crystallization conditions;
(4) isolating co-crystals formed thereby; and
(5) incorporating the co-crystals into a pharmaceutical composition.
In a still further aspect the present invention provides a process for the production of a pharmaceutical composition, which comprises:
(1) grinding, heating or contacting in solution an API with a co-crystal former, under crystallization conditions, so as to form a solid phase;
(2) isolating co-crystals comprising the API and the co-crystal former; and
(3) incorporating the co-crystals into a pharmaceutical composition.
Assaying the solid phase for the presence of co-crystals of the API and the co- crystal former may be carried out by conventional methods known in the art. For example, it is convenient and routine to use powder X-ray diffraction techniques to assess the presence of co-crystals. This may be affected by comparing the spectra of the API, the crystal former and putative co-crystals in order to establish whether or not true co- crystals had been formed. Other techniques, used in an analogous fashion, include differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), solid state NMR spectroscopy, and Raman spectroscopy. Single crystal X-ray diffraction is especially useful in identifying co-crystal structures.
In a further aspect, the present invention therefore provides a process of screening for co-crystal compounds, which comprises:
(1) providing (i) an API compound, and (ii) a co-crystal former; and
(2) screening for co-crystals of APIs with co-crystal formers by subjecting each combination of API and co-crystal former to a step comprising:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions so as to form a solid phase; and
(b) isolating co-crystals comprising the API and the co-crystal former. An alternative embodiment is drawn to a process of screening for co-crystal compounds, which comprises:
(1) providing (i) an API or a plurality of different APIs, and (if) a co-crystal former or a plurality of different co-crystal formers, wherein at least one of the API and the co-crystal former is provided as a plurality thereof; and
(2) screening for co-crystals of APIs with co-crystal formers by subjecting each combination of API and co-crystal former to a step comprising
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions so as to form a solid phase; and
(b) isolating co-crystals comprising the API and the co-crystal former.
Some of the APIs and co-crystal formers of the present invention have one or more chiral centers and may exist in a variety of stereoisomeric configurations. As a consequence of these chiral centers, several APIs and co-crystal formers of the present invention occur as racemates, mixtures of enantiomers and as individual enantiomers, as well as diastereomers and mixtures of diastereomers. All such racemates, enantiomers, and diastereomers are within the scope of the present invention including, for example, cis- and tr< s-isomers, R- and S-enantiomers, and (D)- and (L)-isomers. Co-crystals of the present invention can include isomeric forms of either the API or the co-crystal former or both. Isomeric forms of APIs and co-crystal formers include, but are not limited to, stereoisomers such as enantiomers and diastereomers. In one embodiment, a co-crystal can comprise a racemic API and/or co-crystal former. In another embodiment, a co-crystal can comprise an enantiomerically pure API and/or co-crystal former. In another embodiment, a co-crystal can comprise an API or a co-crystal former with an enantiomeric excess of about 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent, 96 percent, 97 percent, 98 percent, 99 percent, greater than 99 percent, or any intermediate value. Several non- limiting examples of stereoisomeric APIs include modafinil, cw-itraconazole, ibuprofen, and flurbiprofen. Several non-limiting examples of stereoisomeric co-crystal formers include tartaric acid and malic acid.
Co-crystals comprising enantiomerically pure components (e.g., API or co-crystal former) can give rise to chemical and/or physical properties which are modulated with respect to those of the corresponding co-crystal comprising a racemic component. For example, the modafinil :malonic acid co-crystal from Example 10 comprises racemic modafinil. Enantiomerically pure R-modafinil:malonic acid can conceivably be synthesized via the same or another method of the present invention and is therefore included in the scope of the invention. Likewise, enantiomerically pure S- modafϊnikmalonic acid can conceivably be synthesized via a method of the present invention and is therefore included in the scope of the invention. A co-crystal comprising an enantiomerically pure component can give rise to a modulation of, for example, activity, bioavailability, or solubility, with respect to the corresponding co-crystal comprising a racemic component. As an example, the co-crystal R-modafinil:malonic acid can have modulated properties as compared to the racemic modafinikmalonic acid co-crystal.
As used herein and unless otherwise noted, the term "racemic co-crystal" refers to a co-crystal which is comprised of an equimolar mixture of two enantiomers of the API, the co-crystal former, or both. For example, a co-crystal comprising a stereoisomeric API and a non-stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the API enantiomers. Similarly, a co-crystal comprising a non-stereoisomeric API and a stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the co-crystal former enantiomers. In addition, a co-crystal comprising a stereoisomeric API and a stereoisomeric co-crystal former is a "racemic co-crystal" when there is present an equimolar mixture of the API enantiomers and of the co-crystal former enantiomers.
As used herein and unless otherwise noted, the term "enantiomerically pure co- crystal" refers to a co-crystal which is comprised of a stereoisomeric API or a stereoisomeric co-crystal former or both where the enantiomeric excess of the stereoisomeric species is greater than or equal to about 90 percent ee.
In another embodiment, the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the bioavailability is modulated with respect to the racemic co-crystal. In another embodiment, the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the activity is modulated with respect to the racemic co-crystal. In another embodiment, the present invention includes a pharmaceutical composition comprising a co-crystal with an enantiomerically pure API or co-crystal former wherein the solubility is modulated with respect to the racemic co-crystal.
As used herein, the term "enantiomerically pure" includes a composition which is substantially enantiomerically pure and includes, for example, a composition with greater than or equal to about 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent enantiomeric excess.
Solubility Modulation
In a further aspect, the present invention provides a process for modulating the solubility of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
In one embodiment, the solubility of the API is modulated such that the aqueous solubility is increased. Solubility of APIs may be measured by any conventional means such as chromatography (e.g., HPLC) or spectroscopic determination of the amount of API in a saturated solution of the API, such as UV-spectroscopy, IR-spectroscopy, Raman spectroscopy, quantitative mass spectroscopy, or gas chromatography.
In another aspect of the invention, the API may have low aqueous solubility. Typically, low aqueous solubility in the present application refers to a compound having a solubility in water which is less than or equal to 10 mg/mL, when measured at 37 degrees C, and preferably less than or equal to 5 mg/mL or 1 mg/mL. Low aqueous solubility can further be specifically defined as less than or equal to 900, 800, 700, 600, 500, 400, 300, 200 150 100, 90, 80, 70, 60, 50, 40, 30, 20 micrograms/mL, or further 10, 5 or 1 micrograms/mL, or further 900, 800, 700, 600, 500, 400, 300, 200 150, 100 90, 80, 70, 60, 50, 40, 30, 20, or 10 ng/mL, or less than 10 ng/mL when measured at 37 degrees C. Aqueous solubility can also be specified as less than 500, 400, 300, 200, 150, 100, 75, 50 or 25 mg/mL. As embodiments of the present invention, solubility can be increased 2, 3, 4, 5, 7, 10, 15, 20, 25, 50, 75, 100, 200, 300, 500, 750, 1000, 5000, or 10,000 times by making a co-crystal of the reference form (e.g., crystalline or amorphous free acid, free base or zwitter ion, hydrate or solvate), or a salt thereof. Further aqueous solubility can be measured in simulated gastric fluid (SGF) or simulated intestinal fluid (SIF) rather than water. SGF (non-diluted) of the present invention is made by combining 1 g/L Triton X-100 and 2 g/L NaCl in water and adjusting the pH with 20 mM HCl to obtain a solution with a final pH=1.7 (SIF is 0.68% monobasic potassium phosphate, 1% pancreatin, and sodium hydroxide where the pH of the final solution is 7.5). The pH of the solvent used may also be specified as 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, or 14 or any pH in between successive values.
Examples of embodiments includes: co-crystal compositions with an aqueous solubility, at 37 degrees C and a pH of 7.0, that is increased at least 5 fold over the reference form, co-crystal compositions with a solubility in SGF that is increased at least 5 fold over the reference form, co-crystal compositions with a solubility in SIF that is increased at least 5 fold over the reference form.
Dissolution Modulation
In another aspect of the present invention, the dissolution profile of the API is modulated whereby the aqueous dissolution rate or the dissolution rate in simulated gastric fluid or in simulated intestinal fluid, or in a solvent or plurality of solvents is increased. Dissolution rate is the rate at which API solids dissolve in a dissolution medium. For APIs whose absorption rates are faster than the dissolution rates (e.g., steroids), the rate-limiting step in the absorption process is often the dissolution rate. Because of a limited residence time at the absorption site, APIs that are not dissolved before they are removed from intestinal absorption site are considered useless. Therefore, the rate of dissolution has a major impact on the performance of APIs that are poorly soluble. Because of this factor, the dissolution rate of APIs in solid dosage forms is an important, routine, quality control parameter used in the API manufacturing process.
Dissolution rate = K S (C3-C) where K is dissolution rate constant, S is the surface area, Cs is the apparent solubility, and C is the concentration of API in the dissolution medium. For rapid API absorption, C3-C is approximately equal to Cs. The dissolution rate of APIs may be measured by conventional means known in the art.
The increase in the dissolution rate of a co-crystal, as compared to the reference form (e.g., free form or salt), may be specified, such as by 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100%, or by 2, 3, 4, 5 ,6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500, 1000, 10,000, or 100,000 fold greater than the reference form (e.g., free form or salt form) in the same solution. Conditions under which the dissolution rate is measured is the same as discussed above The increase in dissolution may be further specified by the time the composition remains supersaturated before reaching equilibrium solubility.
Examples of above embodiments include: co-crystal compositions with a dissolution rate in aqueous solution, at 37 degrees C and a pH of 7.0, that is increased at least 5 fold over the reference form, co-crystal compositions with a dissolution rate in SGF that is increased at least 5 fold over the reference form, co-crystal compositions with a dissolution rate in SIF that is increased at least 5 fold over the reference form.
Bioavailability Modulation
The methods of the present invention are used to make a pharmaceutical API formulation with greater solubility, dissolution, and bioavailability. Bioavailability can be improved via an increase in AUC, reduced time to Tma , (the time to reach peak blood serum levels), or increased Cmax,. The present invention can result in higher plasma concentrations of API when compared to the neutral form or salt alone (reference form). AUC is the area under the plot of plasma concentration of API (not logarithm of the concentration) against time after API administration. The area is conveniently determined by the "trapezoidal rule": The data points are connected by straight line segments, perpendiculars are erected from the abscissa to each data point, and the sum of the areas of the triangles and trapezoids so constructed is computed. When the last measured concentration (C„, at time tn) is not zero, the AUC from tn to infinite time is estimated by C„/keI.
The AUC is of particular use in estimating bioavailability of APIs, and in estimating total clearance of APIs (Clχ). Following single intravenous doses, AUC = D/Clτ, for single compartment systems obeying first-order elimination kinetics, where D is the dose; alternatively, AUC = C0 keι, where keι is the API elimination rate constant. With routes other than the intravenous, for such systems, AUC = F D/Clχ, where F is the absolute bioavailability of the API.
Thus, in a further aspect, the present invention provides a process for modulating the bioavailability of an API when administered in its normal and effective dose range as a co-crystal, whereby the AUC is increased, the time to Tmax is reduced, or Cmax is increased, as compared to a reference form, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Examples of the above embodiments include: co-crystal compositions with a time to Tmax that is reduced by at least 10% as compared to the reference form, co-crystal compositions with a time to Tmax that is reduced by at least 20% over the reference form, co-crystal compositions with a time to Tmax that is reduced by at least 40% over the reference form, co-crystal compositions with a time to Tmax that is reduced by at least 50% over the reference form, co-crystal compositions with a Tmax that is reduced by at least 60% over the reference form, co-crystal compositions with a Tmax that is reduced by at least 70% over the reference form, co-crystal compositions with a Tma that is reduced by at least 80% over the reference form, co-crystal compositions with a Tmax that is reduced by at least 90% over the reference form, co-crystal compositions with a Cmaχ that is increased by at least 20% over the reference form, co-crystal compositions with a Cmax that is increased by at least 30% over the reference form, co-crystal compositions with a Cmax that is increased by at least 40% over the reference form, co-crystal compositions with a Cmax that is increased by at least 50% over the reference form, co-crystal compositions with a Cmax that is increased by at least 60% over the reference form, co- crystal compositions with a Cmax that is increased by at least 70% over the reference form, co-crystal compositions with a Cmaκ that is increased by at least 80% over the reference form, co-crystal compositions with a Cmax that is increased by at least 2 fold, 3 fold, 5 fold, 7.5 fold, 10 fold, 25 fold, 50 fold or 100 fold, co-crystal compositions with an AUC that is increased by at least 10% over the reference form, co-crystal compositions with an AUC that is increased by at least 20% over the reference form, co- crystal compositions with an AUC that is increased by at least 30% over the reference form, co-crystal compositions with an AUC that is increased by at least 40% over the reference form, co-crystal compositions with an AUC that is increased by at least 50% over the reference form, co-crystal compositions with an AUC that is increased by at least 60% over the reference form, co-crystal compositions with an AUC that is increased by at least 70% over the reference form, co-crystal compositions with an AUC that is increased by at least 80% over the reference form or co-crystal compositions with an AUC that is increased by at least 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, or 10 fold. Other examples include wherein the reference form is crystalline, wherein the reference form is amorphous, wherein the reference form is an anhydrous crystalline sodium salt, or wherein the reference form is an anhydrous crystalline HCl salt.
Dose Response Modulation
In a further aspect the present invention provides a process for improving the dose response of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution an API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
Dose response is the quantitative relationship between the magnitude of response and the dose inducing the response and may be measured by conventional means known in the art. The curve relating effect (as the dependent variable) to dose (as the independent variable) for an API-cell system is the "dose-response curve". Typically, the dose-response curve is the measured response to an API plotted against the dose of the API (mg/kg) given. The dose response curve can also be a curve of AUC against the dose of the API given.
In an embodiment of the present invention, a co-crystal of the present mvention has an increased dose response curve or a more linear dose response curve than the corresponding reference compound.
Increased Stability
In a still further aspect the present invention provides a process for improving the stability of an API (as compared to a reference form such as its free form or a salt thereof), which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the pharmaceutical salt with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
In a preferred embodiment, the compositions of the present invention, including the API or active pharmaceutical ingredient (API) and formulations comprising the API, are suitably stable for pharmaceutical use. Preferably, the API or formulations thereof of the present invention are stable such that when stored at 30 degrees C for 2 years, less than 0.2 % of any one degradant is formed. The term degradant refers herein to product(s) of a single type of chemical reaction. For example, if a hydrolysis event occurs that cleaves a molecule into two products, for the purpose of the present invention, it would be considered a single degradant. More preferably, when stored at 40 degrees C for 2 years, less than 0.2 % of any one degradant is formed. Alternatively, when stored at 30 degrees C for 3 months, less than 0.2% or 0.15 %, or 0.1 % of any one degradant is formed, or when stored at 40 degrees C for 3 months, less than 0.2 % or 0.15 %, or 0.1 % of any one degradant is formed. Further alternatively, when stored at 60 degrees C for 4 weeks, less than 0.2 % or 0.15 %, or 0.1 % of any one degradant is formed. The relative humidity (RH) may be specified as ambient (RH), 75 % (RH), or as any single integer between 1 to 99 %. Difficult to Salt or Unsaltable Compounds
In a still further aspect the present invention provides a process for making co- crystals of unsaltable or difficult to salt APIs which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution an API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former. Difficult to salt compounds include bases with a pKa less than 3 or acids with a pKa greater than 10. Zwitter ions are also difficult to salt or unsaltable compounds according to the present invention.
Decreasing Hygroscopicity
In a still further aspect, the present invention provides a method for decreasing the hygroscopicity of an API, which method comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
An aspect of the present invention provides a pharmaceutical composition comprising a co-crystal of an API that is less hygroscopic than amorphous or crystalline, free form or salt (including metal salts such as sodium, potassium, lithium, calcium, magnesium) or another reference compound. Hygroscopicity can be assessed by dynamic vapor sorption analysis, in which 5-50 mg of the compound is suspended from a Cahn microbalance. The compound being analyzed should be placed in a non- hygroscopic pan and its weight should be measured relative to an empty pan composed of identical material and having nearly identical size, shape, and weight. Ideally, platinum pans should be used. The pans should be suspended in a chamber through which a gas, such as air or nitrogen, having a controlled and known percent relative humidity (%RH) is flowed until eqilibrium criteria are met. Typical equilibrium criteria include weight changes of less than 0.01 % over 3 minutes at constant humidity and temperature. The relative humidity should be measured for samples dried under dry nitrogen to constant weight (<0.01 % change in 3 minutes) at 40 degrees C unless doing so would de-solvate or otherwise convert the material to an amorphous compound. In one aspect, the hygroscopicity of a dried compound can be assessed by increasing the RH from 5 to 95 % in increments of 5 % RH and then decreasing the RH from 95 to 5 % in 5 % increments to generate a moisture sorption isotherm. The sample weight should be allowed to equilibrate between each change in % RH. If the compound deliquesces or becomes amorphous above 75 % RH, but below 95 % RH, the experiment should be repeated with a fresh sample and the relative humidity range for the cycling should be narrowed to 5-75 % RH or 10-75 % RH, instead of 5-95 %RH. If the sample cannot be dried prior to testing due to lack of form stability, than the sample should be studied using two complete humidity cycles of either 10-75 % RH or 5-95 % RH, and the results of the second cycle should be used if there is significant weight loss at the end of the first cycle. Hygroscopicity can be defined using various parameters. For purposes of the present invention, a non-hygroscopic molecule should not gain or lose more than 1.0 %, or more preferably, 0.5 % weight at 25 degrees C when cycled between 10 and 75 % RH (relative humidity at 25 degrees C). The non-hygroscopic molecule more preferably should not gain or lose more than 1.0 %, or more preferably, 0.5 % weight when cycled between 5 and 95 % RH at 25 degrees C, or more than 0.25 % of its weight between 10 and 75 % RH. Most preferably, a non-hygroscopic molecule will not gain or lose more than 0.25 % of its weight when cycled between 5 and 95 % RH.
Alternatively, for purposes of the present invention, hygroscopicity can be defined using the parameters of Callaghan et al., "Equilibrium moisture content ofpharmaceutical excipients", in Api Dev. Ind. Pharm., Vol. 8, pp. 335-369 (1982). Callaghan et al. classified the degree of hygroscopicity into four classes.
Class 1 : Non-hygroscopic Essentially no moisture increases occur at relative humidities below 90 %.
Class 2: Slightly hygroscopic Essentially no moisture increases occur at relative humidities below 80%. Class 3: Moderately hygroscopic Moisture content does not increase more than 5 % after storage for 1 week at relative humidities below 60 %.
Class 4: Very hygroscopic Moisture content increase may occur at relative humidities as low as 40 to 50 %.
Alternatively, for purposes of the present invention, hygroscopicity can be defined using the parameters of the European Pharmacopoeia Technical Guide (1999, p. 86) which has defined hygrospocity, based on the static method, after storage at 25 degrees C for 24 hours at 80 % RH:
Slightly hygroscopic: Increase in mass is less than 2 percent m/m and equal to or greater than 0.2 percent m/m.
Hygroscopic: Increase in mass is less than 15 percent m/m and equal to or greater than 0.2 percent m/m.
Very Hygroscopic: Increase in mass is equal to or greater than 15 percent m/m. Deliquescent: Sufficient water is absorbed to form a liquid.
Co-crystals of the present invention can be set forth as being in Class 1, Class 2, or Class 3, or as being Slightly hygroscopic, Hygroscopic, or Very Hygroscopic. Co- crystals of the present invention can also be set forth based on their ability to reduce hygroscopicity. Thus, preferred co-crystals of the present invention are less hygroscopic than a reference compound. The reference compound can be specified as the API in free form (free acid, free base, hydrate, solvate, etc.) or salt (e.g., especially metal salts such as sodium, potassium, lithium, calcium, or magnesium). Further included in the present invention are co-crystals that do not gain or lose more than 1.0 % weight at 25 degrees C when cycled between 10 and 75 % RH, wherein the reference compound gains or loses more than 1.0 % weight under the same conditions. Further included in the present invention are co-crystals that do not gain or lose more than 0.5 % weight at 25 degrees C when cycled between 10 and 75 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions. Further included in the present invention are co-crystals that do not gain or lose more than 1.0 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 1.0 % weight under the same conditions. Further included in the present invention are co-crystals that do not gain or lose more than 0.5 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions. Further included in the present invention are co-crystals that do not gain or lose more than 0.25 % weight at 25 degrees C when cycled between 5 and 95 % RH, wherein the reference compound gains or loses more than 0.5 % or more than 1.0 % weight under the same conditions.
Further included in the present invention are co-crystals that have a hygroscopicity (according to Callaghan et al.) that is at least one class lower than the reference compound or at least two classes lower than the reference compound. Included are a Class 1 co-crystal of a Class 2 reference compound, a Class 2 co-crystal of a Class 3 reference compound, a Class 3 co-crystal of a Class 4 reference compound, a Class 1 co- crystal of a Class 3 reference compound, a Class 1 co-crystal of a Class 4 reference compound, or a Class 2 co-crystal of a Class 4 reference compound.
Further included in the present invention are co-crystals that have a hygroscopicity (according to the European Pharmacopoeia Technical Guide) that is at least one class lower than the reference compound or at least two classes lower than the reference compound. Non-limiting examples include; a slightly hygroscopic co-crystal of a hygroscopic reference compound, a hygroscopic co-crystal of a very hygroscopic reference compound, a very hygroscopic co-crystal of a deliquescent reference compound, a slightly hygroscopic co-crystal of a very hygroscopic reference compound, a slightly hygroscopic co-crystal of a deliquescent reference compound, and a hygroscopic co-crystal of a deliquescent reference compound.
Crystallizing Amorphous Compounds
In a further aspect, the present invention provides a process for crystallizing an amorphous compound, which process comprises: (1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former. An amorphous compound includes compounds that do not crystallize using routine methods in the art.
Decreasing Form Diversity
In a still further embodiment aspect the present invention provides a process for reducing the form diversity of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
For purposes of the present invention, the number of forms of a co-crystal is compared to the number of forms of a reference compound (e.g. the free form or a salt of the API) that can be made using routine methods in the art.
Morphology Modulation
In a still further aspect the present invention provides a process for modifying the morphology of an API, which process comprises:
(1) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal former under crystallization conditions, so as to form a co-crystal of the API and the co-crystal former; and
(2) isolating co-crystals comprising the API and the co-crystal former.
In an embodiment the co-crystal comprises or consists of a co-crystal former and a pharmaceutical wherein the interaction between the two, e.g., H-bonding, occurs between a functional group of Table III of an API with a corresponding interacting group of Table III. In a further embodiment, the co-crystal comprises a co-crystal former of Table I or II and an API with a corresponding interacting group of Table III. In a further embodiment the co-crystal comprises an API from Table IV and a co-crystal former with a functional group of Table III. In a further embodiment, the co-crystal is from Table I or II. In an aspect of the invention, only co-crystals having an H-bond acceptor on the first molecule and an H-bond donor on the second molecule, where the first and second molecules are either co-crystal former and API respectively or API and co-crystal former respectively, are included in the present invention. Table IV includes the CAS number, chemical name or a PCT or patent reference (each incorporated herein in their entireties). Thus, whether a particular API contains an H-bond donor, acceptor or both is readily apparent.
In another embodiment, the co-crystal former and API each have only one H- bond donor/acceptor. In another aspect, the molecular weight of the API is less than 2000, 1500, 1000, 750, 500, 350, 200, or 150 Daltons. In another embodiment, the molecular weight of the API is between 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 900-1000, 1000-1200, 1200-1400, 1400-1600, 1600-1800, or 1800-2000. APIs with the above molecular weights may also be specifically excluded from the present invention.
The hydrogen bond donor moieties of a co-crystal can include, but are not limited to, any one, any two, any three, any four, or more of the following: amino-pyridine, primary amine, secondary amine, sulfonamide, primary amide, secondary amide, alcohol, and carboxylic acid. The hydrogen bond acceptor moieties of a co-crystal can include, but are not limited to, any one, any two, any three, any four, or more of the following: amino-pyridine, primary amine, secondary amine, sulfonamide, primary amide, secondary amide, alcohol, carboxylic acid, carbonyl, cyano, dimethoxyphenyl, sulfonyl, aromatic nitrogen (6 membered ring), ether, chloride, organochloride, bromide, organobromide, and organoiodide. Hydrogen bonds are known to form many supramolecular structures including, but not limited to, a catemer, a dimer, a trimer, a tβtramer, or a higher order structure. Tables V-XXI list specific hydrogen bond donor and acceptor moieties and their approximate interaction distances from the electromagnetic donor atom through the hydrogen atom to the electromagnetic acceptor atom. For example, Table V lists functional groups that are known to hydrogen bond with amino-pyridines. Amino-pyridines comprise two distinct sites of hydrogen bond donation/acceptance. Both the aromatic nitrogen atom (Npy) and the amine group (NH2) can participate in hydrogen bonds. The ability of a given functional group to participate in a hydrogen bond as a donor or as an acceptor or both can be determined by inspection by those skilled in the art.
The data included in Tables V-XXI are taken from an analysis of solid-state structures as reported in the Cambridge Structural Database (CSD). These data include a number of hydrogen bonding interactions between many functional groups and their associated interaction distances.
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Table XVIII- Hydrogen bonding functional groups with organochlorides and associated interaction distances
Figure imgf000040_0002
Figure imgf000040_0003
Figure imgf000040_0004
Figure imgf000041_0001
In another embodiment, peptides, proteins, nucleic acids or other biological APIs are excluded from the present invention. In another embodiment, all non- pharmaceutically acceptable co-crystal formers are excluded from the present invention. In another embodiment, organometalic APIs are excluded from the present invention. In another embodiment, a co-crystal former comprising any one or more of the functional groups of Table III may be specifically excluded from the present invention. In another embodiment, any one or more of the co-crystal formers of Table I or II may be specifically excluded from the present invention. Any APIs currently known in the art may also be specifically excluded from the present invention. For example, carbamazepine, itraconazole, nabumetone, fluoxetine, acetaminophen and theophylline can each be specifically excluded from the present invention. In another embodiment, the API is not a salt, is not a non-metal salt, or is not a metal salt, e.g., sodium, potassium, lithium, calcium or magnesium. In another embodiment, the API is a salt, is a non-metal salt, or is a metal salt, e.g., sodium, potassium, lithium, calcium, magnesium. In one embodiment, the API does not contain a halogen. In one embodiment, the API does contain a halogen.
In another embodiment, any one or more of the APIs of Table IV may be specifically excluded from the present invention. Any APIs currently known in the art may also be specifically excluded from the present invention. For example, nabumetone:2,3-naphthalenediol, fluoxetine HCkbenzoic acid, fluoxetine HCLsuccinic acid, acetaminophe piperazine, acetaminophe theophylline, theophylline: salicylic acid, theophylline :p-hydiOxybenzoic acid, theophylline: sorbic acid, theophylline :l-hydroxy-2- naphthoic acid, theophylline :glycolic acid, theophylline:2,5-dihydroxybenzoic acid, theophylline xhloroacetic acid, bis(diphenylhydantoin):9-ethyladenine acetylacetone solvate, bis(diphenylhydantoin):9-ethyladenine 2,4-pentanedione solvate, 5,5- diphenylbarbituric acid:9-ethyladenine, bis(diphenylhydantoin):9-ethyladenine, 4- aminobenzoic acid:4-aminobenzonitrile, sulfadimidine: salicylic acid, 8- hydroxyquinolinium 4-nitrobenzoate:4-nitrobenzoic acid, sulfaproxyline affeine, retro- inverso-isopropyl (2R,3 S)-4-cyclohexyl-2-hydroxy-3 -(N-((2R)-2- morpholinocarbonylmethyl-3 -( 1 -naphthyl)propionyl)-L-histidylamino)butyrate xinnamic acid monohydrate, benzoic acid sonicotinamide, 3-(2-N',N'-(dimethylhydrazino)-4- thiazolylmethylthio)-N"-sulfamoylpropionamidine:maleic acid, diglycine hydrochloride (C H5NO2:C2H6NO2 +Cr), octadecanoic acid:3-pyridinecarboxamide, cw-N-(3-methyl-l- (2-( 1 ,2,3 ,4-tetrahydro)naphthyl)-piperidin-4-yl)-N-phenylpropanamide hydrochloride: oxalic acid, trans-N-(3 -methyl- 1 -(2-(l ,2,3, 4-tetrahydro)naphthyl)- piperidin-4-ylium)-N-phenylpropanamide oxalate: oxalic acid dihydrate, bis(l-(3-((4-(2- isopropoxyphenyl)- 1 -piperazinyl)methyl)benzoyl)piperidine) succinate : succinic acid, bis(p-cyanophenyl)imidazolylmethane:succinic acid, cz's- l-((4-(l- imidazolylmethyl)cyclohexyl)methyl)imidazole:succinic acid, (+)-2-(5,6-dimethoxy- l,2,3,4-tetrahydro-l-naphthyl)imidazoline:(+)-dibenzoyl-D-tartaric acid, raclopride:tartaric acid, 2,6-diamino-9-ethylpurine:5,5-diethylbarbituric acid, 5,5- diethylbarbituric acid:bis(2-aminopyridine), 5,5-diethylbarbituric acid:acetamide, 5,5- diethylbarbituric acid:KI3, 5,5-diethylbarbituric acid:urea, bis(barbital):hexamethylphosphoramide, 5,5-diethylbarbituric acid:imidazole, barbitahl- methylimidazole, 5,5-diethylbarbituric acid:N-methyl-2-pyridone, 2,4-diamino-5-(3,4,5- trimethoxybenzyl)-pyrimidine:5,5-diethylbarbituric acid, bis(barbital): caffeine, bis arbital) : 1 -methylimidazole, bis(beta-cyclodextrin) :bis(barbital) hydrate, tetrakis(beta-cyclodextrin):tetrakis(barbital), 9-ethyladenine:5,5-diethylbarbituric acid, barbitaLN' -(p-cyanophenyl)-N-(p-iodophenyl)melamine, barbital :2-amino-4-(m- bromophenylamino)-6-chloro-l,3,5-triazine, 5,5-diethylbarbituric acid:N,N'- diphenyl elamine, 5,5-diethylbarbituric acid:N,N'-bis(p-chlorophenyl)melamine, N,N'- bis(p-bromophenyl)melamine:5,5-diethylbarbituric acid, 5,5-diethylbarbituric acid:N,N'- bis(p-iodophenyl)melamine, 5,5-diethylbarbituric acid:N,N'-bis(p-tolyl)melamine, 5,5- diethylbarbituric acid:N,N'-bis(rn-tolyl)melamine, 5,5-diethylbarbituric acid:N,N'-bis(m- chlorophenyl)melamine, N,N'-Bis(7«-methylphenyl)melamine:barbital, N,N'-bis(/??- chlorophenyl)melamine:barbital tetrahydrofuran solvate, 5,5-diethylbarbituric acid:N,N'- bis(/ert-butyl)melamine, 5,5-diethylbarbituric acid:N,N'-di(tert-butyl)melamine, 6,6'- diquinolyl ether:5,5-diethylbarbituric acid, 5-tert-butyl-2,4,6- triaminopyrimidine:diethylbarbituric acid, N,N'-bis(4- carboxymethylρhenyl)melamine:barbital ethanol solvate, N,N'-bis(4-tert- butylphenyl)melamine:barbital, tris(5, 17-N,N'-bis(4-amino-6-(butylamino)- 1 ,3 ,5-triazin- 2-yl)diamino-l 1 ,23-dinitro-25,26,27,28- tetrapropoxycalix(4)arene):hexakis(diethylbarbituric acid) toluene solvate, N,N'-bis( - fluorophenyl)melamine:barbital, N,N' -bis(m-bromophenyl)melamine:barbital acetone solvate, N,N'-bis(m-iodophenyl)melamine:barbital acetonitrile solvate, N,N'-bis( - trifluoromethylphenyl)melamine:barbital acetonitrile solvate, aminopyrine:barbital, N,N'-bis(4-fluorophenyl)melamine:barbital, N,N'-bis(4- trifluoromethylphenyl)melamine:barbital, 2,4-diamino-5-(3,4,5- trimethoxybenzyl)pyrimidine:barbital, hydroxybutyrate:hydroxyvalerate, 2- aminopyrimidine: succinic acid, l,3-bis(((6-methylpyrid-2- yl)amino)carbonyl)benzene:glutaric acid, 5-tert-butyl-2,4,6- triaminopyrimidine:diethylbarbituric acid, bis(dithiobiuret-S,S')nickel(II):diuracil, platinum 3,3'-dihydroxymethyl-2,2'-bipyridine dichloride:AgF3CSO3, 4,4'- bipyridyl:isophthalic acid, 4,4'-bipyridyl:l,4-naphthalenedicarboxylic acid, 4,4'- bipyridyl:l,3,5-cyclohexane-tricarboxylic acid, 4,4'-bipyridyl:tricaballylic acid, urotropin:azelaic acid, insulin:C8-HI (octanoyl-Ne-LysB29-human insulin), isonicotinamide: cinnamic acid, isonicotinamide:3-hydroxybenzoic acid, isonicotinamide : 3 -N,N-dimethylaminobenzoic acid, isonicotinamide : 3 ,5 - bis(trifluoromethyl)-benzoic acid, isonicotinamide:d,l-mandelic acid, isonicotinamidexhloroacetic acid, isonicotinamide:fumaric acid monoethyl ester, isonicotinamide: 12-bromododecanoic acid, isonicotinamide:fumaric acid, isonicotinamide: succinic acid, isonicotinamide :4-ketopimelic acid, isonicotinamide :thiodiglycolic acid, 1,3,5-cyclohexane-tricarboxylic acid:hexamethyltetramine, 1,3,5-cyclohexane-tricarboxylic acid:4,7-phenanthroline, 4,7- phenanthroline: oxalic acid, 4,7-phenanthroline:terephthalic acid, 4,7-phenanthroline: 1 ,5-cyclohexane-tricarboxylic acid, 4,7-pl enantlιroline: 1 ,4-naphthalenedicarboxylic acid, pyrazine:methanoic acid, pyrazine:ethanoic acid, pyrazine:propanoic acid, pyrazine:butanoic acid, pyrazine:pentanoic acid, pyrazine:hexanoic acid, pyrazine:heptanoic acid, pyrazine:octanoic acid, pyrazinemonanoic acid, pyrazine:decanoic acid, diammine-(deoxy-quanyl-quanyl-N ,N )-platinum:tris(glycine) hydrate, 2-aminopyrimidine:p-phenylenediacetic acid, bis(2-aminopyrimidin-l- ium)fumarate:fiιmaric acid, 2-aminopyrimidine:indole-3 -acetic acid, 2- aminopyrimidine:N-methylpyrrole-2-carboxylic acid, 2-aminopyrimidine:thiophen-2- carboxylic acid, 2-aminopyrimidine:(+)-camphoric acid, 2,4,6-Trinitrobenzoic acid:2- aminopyrimidine, 2-aminopyrimidine:4-aminobenzoic acid, 2- aminopyrimidine:bis(phenoxyacetic acid), 2-aminopyrimidine:(2,4- dichlorophenoxy)acetic acid, 2-aminopyrimidine:(3,4-dichlorophenoxy)acetic acid, 2- aminopyrimidine:indole-2-carboxylic acid, 2-aminopyrimidine:terephthalic acid, 2- aminopyrimidine:bis(2-nitrobenzoic acid), 2-aminopyrimidine:bis(2-aminobenzoic acid), 2-aminopyrimidine:3-aminobenzoic acid, 2-hexeneoic acid:isonicotinamide, 4- nitrobenzoic acid:isonicotinamide, 3,5-dinitrobenzoic acid:isonicotinamide:4- methylbenzoic acid, 2-amino-5-nitropyrimidine:2-amino-3-nitropyridine, 3,5- dinitrobenzoic acid:4-chlorobenzamide, 3-dimethylaminobenzoic acid:4- chlorobenzamide, fumaric acid:4-chlorobenzamide, oxine:4-nitrobenzoic acid, oxine:3,5- dinitrobenzoic acid, oxine:3,5-dinitrosalicylic acid, 3-[2-(N',N'-dimethylhydrazino)-4- thiazolylmethylthio] -N2-sulfamoylpropionamidine :maleic acid, 5 -fluorouracil : 9- ethylhypoxanthine, 5-fluorouracil:cytosine dihydrate, 5-fluorouracil :theophylline monohydrate, stearic acid:nicotinamide, c ,y-l-{[4-(l- imidazolylmethyl)cyclohexyl]methyl}imidazole:succinic acid, CGS 18320B: succinic acid, sulfaproxyline: caffeine, 4-aminobenzoic acid:4-aminobenzonitrile, 3,5- dinitrobenzoic acid:isonicotinamide:3-methylbenzoic acid, 3,5-dinitrobenzoic acid:isonicotinamide:4-(dimethylamino)benzoic acid, 3,5-dinitrobenzoic acid:isonicotinamide:4-hydroxy-3-methoxycinnamic acid, isonicotinamide: oxalic acid, isonicotinamide:malonic acid, isonicotinamide: succinic acid, isonicotinamide :glutaric acid, isonicotinamide :adipic acid, benzoic acid:isonicotinamide, mazapertine:succinate, betaine:dichloronitrophenol, betainepyridine:dichloronitrophenol, betainepyridine :pentachlorophenol, 4- {2-[ 1 -(2-hydroxyethyl)-4-pyridylidene] - ethylidene}-cyclo-hexa-2,5-dien-l-one:methyl 2,4-dihydroxybenzoate, 4-{2-[l-(2- hydroxyethyl)-4-pyridylidene]-ethylidene} -cyclo-hexa-2,5-dien- 1 -one:2,4- dihydroxypropiophenone, 4- {2- [ 1 -(2-hydroxyethyl)-4-pyridylidene] -ethylidene } -cyclo- hexa-2,5-dien-l-one:2,4-dihydroxyacetophenone5 squaric acid:4,4'-dipyridylacetylene, squaric acid:l,2-bis(4-pyridyl)ethylene, chloranilic acid:l,4-bis[(4- pyridyl)ethynyl] benzene, 4,4'-bipyridine:phthalic acid, 4,4'-dipyridylacetylene:phthalic acid, bis(pentamethylcyclopentadienyl)iron:bromanilic acid, bis(pentamethylcyclopentadienyl)iron:chloranilic acid, bis(pentamethylcyclopentadienyl)iron:cyananilic acid, pyrazinotetrathiafulvalene: chloranilic acid, phenoLpentafluorophenol, co-crystals of cis- itraconazole, and co-crystals of topiramate are specifically excluded from the present invention.
In another embodiment, a pharmaceutical composition can be formulated to contain an API in co-crystal form as micronized or nano-sized particles. More specifically, another embodiment couples the processing of a pure API to a co-crystal form with the process of making a controlled particle size for manipulation into a pharmaceutical dosage form. This embodiment combines two processing steps into a single step via techniques such as, but not limited to, grinding, alloying, or sintering (i.e., heating a powder mix). The coupling of these processes overcomes a serious limitation of having to isolate and store the bulk drug that is required for a formulation, which in some cases can be difficult to isolate (e.g., amorphous, chemically or physically unstable).
Excipients employed in pharmaceutical compositions of the present invention can be solids, semi-solids, liquids or combinations thereof. Preferably, excipients are solids. Compositions of the invention containing excipients can be prepared by any known technique of pharmacy that comprises admixing an excipient with an API or therapeutic agent. A pharmaceutical composition of the invention contains a desired amount of API per dose unit and, if intended for oral administration, can be in the form, for example, of a tablet, a caplet, a pill, a hard or soft capsule, a lozenge, a cachet, a dispensable powder, granules, a suspension, an elixir, a dispersion, or any other form reasonably adapted for such administration. If intended for parenteral administration, it can be in the form, for example, of a suspension or transdermal patch. If intended for rectal administration, it can be in the form, for example, of a suppository. Presently preferred are oral dosage forms that are discrete dose units each containing a predetermined amount of the API, such as tablets or capsules.
In another embodiment, APIs with an inappropriate pH for transdermal patches can be co-crystallized with an appropriate co-crystal former, thereby adjusting its pH to an appropriate level for use as a transdermal patch. In another embodiment, an APIs pH level can be optimized for use in a transdermal patch via co-crystallization with an appropriate co-crystal former.
Non-limiting examples follow of excipients that can be used to prepare pharmaceutical compositions of the invention.
Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable carriers or diluents as excipients. Suitable carriers or diluents illustratively include, but are not limited to, either individually or in combination, lactose, including anhydrous lactose and lactose monohydrate; starches, including directly compressible starch and hydrolyzed starches (e.g., Celutab™ and Emdex™); mannitol; sorbitol; xylitol; dextrose (e.g., Cerelose™ 2000) and dextrose monohydrate; dibasic calcium phosphate dihydrate; sucrose-based diluents; confectioner's sugar; monobasic calcium sulfate monohydrate; calcium sulfate dihydrate; granular calcium lactate trihydrate; dextrates; inositol; hydrolyzed cereal solids; amylose; celluloses including microcrystalline cellulose, food grade sources of alpha- and amorphous cellulose (e.g., RexcelJ), powdered cellulose, hydroxypropylcellulose (HPC) and hydroxypropylmethylcellulose (HPMC); calcium carbonate; glycine; bentonite; block co-polymers; polyvinylpyrrolidone; and the like. Such carriers or diluents, if present, constitute in total about 5% to about 99%, preferably about 10% to about 85%, and more preferably about 20% to about 80%, of the total weight of the composition. The carrier, carriers, diluent, or diluents selected preferably exhibit suitable flow properties and, where tablets are desired, compressibility.
Lactose, mannitol, dibasic sodium phosphate, and microcrystalline cellulose (particularly Avicel PH microcrystalline cellulose such as Avicel PH 101), either individually or in combination, are preferred diluents. These diluents are chemically compatible with many co-crystals described herein. The use of extragranular microcrystalline cellulose (that is, microcrystalline cellulose added to a granulated composition) can be used to improve hardness (for tablets) and/or disintegration time. Lactose, especially lactose monohydrate, is particularly preferred. Lactose typically provides compositions having suitable release rates of co-crystals, stability, pre- compression flowability, and/or drying properties at a relatively low diluent cost. It provides a high density substrate that aids densification during granulation (where wet granulation is employed) and therefore improves blend flow properties and tablet properties.
Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable disintegrants as excipients, particularly for tablet formulations. Suitable disintegrants include, but are not limited to, either individually or in combination, starches, including sodium starch glycolate (e.g., Explotab™ of PenWest) and pregelatinized corn starches (e.g., National™ 1551 of National Starch and Chemical Company, National™ 1550, and Colorcon™ 1500), clays (e.g., Veegum™ HV of R.T. Vanderbilt), celluloses such as purified cellulose, microcrystalline cellulose, methylcellulose, carboxymethylcellulose and sodium carboxymethylcellulose, croscarmellose sodium (e.g., Ac-Di-Sol™ of FMC), alginates, crospovidone, and gums such as agar, guar, locust bean, karaya, pectin and fragacanth gums.
Disintegrants may be added at any suitable step during the preparation of the composition, particularly prior to granulation or during a lubrication step prior to compression. Such disintegrants, if present, constitute in total about 0.2% to about 30%, preferably about 0.2% to about 10%, and more preferably about 0.2% to about 5%, of the total weight of the composition.
Croscarmellose sodium is a preferred disintegrant for tablet or capsule disintegration, and, if present, preferably constitutes about 0.2% to about 10%, more preferably about 0.2% to about 7%, and still more preferably about 0.2% to about 5%, of the total weight of the composition. Croscarmellose sodium confers superior intragranular disintegration capabilities to granulated pharmaceutical compositions of the present invention. Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable binding agents or adhesives as excipients, particularly for tablet formulations. Such binding agents and adhesives preferably impart sufficient cohesion to the powder being tableted to allow for normal processing operations such as sizing, lubrication, compression and packaging, but still allow the tablet to disintegrate and the composition to be absorbed upon ingestion. Such binding agents may also prevent or inhibit crystallization or recrystallization of a co-crsytal of the present invention once the salt has been dissolved in a solution. Suitable binding agents and adhesives include, but are not limited to, either individually or in combination, acacia; fragacanth; sucrose; gelatin; glucose; starches such as, but not limited to, pregelatinized starches (e.g., National™ 1511 and National™ 1500); celluloses such as, but not limited to, methylcellulose and carmellose sodium (e.g., Tylose™); alginic acid and salts of alginic acid; magnesium aluminum silicate; PEG; guar gum; polysaccharide acids; bentonites; povidone, for example povidone K-15, K-30 and K-29/32; polymethacrylates; HPMC; hydroxypropylcellulose (e.g., Klucel™ of Aqualon); and ethylcellulose (e.g., Ethocel™ of the Dow Chemical Company). Such binding agents and/or adhesives, if present, constitute in total about 0.5% to about 25%, preferably about 0.75% to about 15%, and more preferably about 1% to about 10%, of the total weight of the pharmaceutical composition.
Many of the binding agents are polymers comprising amide, ester, ether, alcohol or ketone groups and, as such, are preferably included in pharmaceutical compositions of the present invention. Polyvinylpyrrolidones such as povidone K-30 are especially preferred. Polymeric binding agents can have varying molecular weight, degrees of crosslinking, and grades of polymer. Polymeric binding agents can also be copolymers, such as block co-polymers that contain mixtures of ethylene oxide and propylene oxide units. Variation in these units' ratios in a given polymer affects properties and performance. Examples of block co-polymers with varying compositions of block units are Poloxamer 188 and Poloxamer 237 (BASF Corporation).
Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable wetting agents as excipients. Such wetting agents are preferably selected to maintain the co-crystal in close association with water, a condition that is believed to improve bioavailability of the composition. Such wetting agents can also be useful in solubilizing or increasing the solubility of co-crystals.
Non-limiting examples of surfactants that can be used as wetting agents in pharmaceutical compositions of the invention include quaternary ammonium compounds, for example benzalkonium chloride, benzethonium chloride and cetylpyridinium chloride, dioctyl sodium sulfosuccinate, polyoxyethylene alkylphenyl ethers, for example nonoxynol 9, nonoxynol 10, and degrees Ctoxynol 9, poloxamers (polyoxyethylene and polyoxypropylene block copolymers), polyoxyethylene fatty acid glycerides and oils, for example polyoxyethylene (8) caprylic/capric mono- and diglycerides (e.g., Labrasol™ of Gattefosse), polyoxyethylene (35) castor oil and polyoxyethylene (40) hydrogenated castor oil; polyoxyethylene alkyl ethers, for example polyoxyethylene (20) cetostearyl ether, polyoxyethylene fatty acid esters, for example polyoxyethylene (40) stearate, polyoxyethylene sorbitan esters, for example polysorbate 20 and polysorbate 80 (e.g., Tween 80 of ICI), propylene glycol fatty acid esters, for example propylene glycol laurate (e.g., Lauroglycol™ of Gattefosse), sodium lauryl sulfate, fatty acids and salts thereof, for example oleic acid, sodium oleate and triethanolamine oleate, glyceryl fatty acid esters, for example glyceryl monostearate, sorbitan esters, for example sorbitan monolaurate, sorbitan monooleate, sorbitan monopalmitate and sorbitan monostearate, tyloxapol, and mixtures thereof. Such wetting agents, if present, constitute in total about 0.25% to about 15%, preferably about 0.4% to about 10%, and more preferably about 0.5% to about 5%, of the total weight of the pharmaceutical composition.
Wetting agents that are anionic surfactants are preferred. Sodium lauryl sulfate is a particularly preferred wetting agent. Sodium lauryl sulfate, if present, constitutes about 0.25% to about 7%, more preferably about 0.4% to about 4%, and still more preferably about 0.5% to about 2%, of the total weight of the pharmaceutical composition.
Pharmaceutical compositions of the invention optionally comprise one or more pharmaceutically acceptable lubricants (including anti-adherents and/or glidants) as excipients. Suitable lubricants include, but are not limited to, either individually or in combination, glyceryl behapate (e.g., Compritol™ 888 of Gattefosse); stearic acid and salts thereof, including magnesium, calcium and sodium stearates; hydrogenated vegetable oils (e.g., Sterotex of Abitec); colloidal silica; talc; waxes; boric acid; sodium benzoate; sodium acetate; sodium fumarate; sodium chloride; DL-leucine; PEG (e.g., Carbowax™ 4000 and Carbowax™ 6000 of the Dow Chemical Company); sodium oleate; sodium lauryl sulfate; and magnesium lauryl sulfate. Such lubricants, if present, constitute in total about 0. 1% to about 10%, preferably about 0.2% to about 8%, and more preferably about 0.25% to about 5%, of the total weight of the pharmaceutical composition.
Magnesium stearate is a preferred lubricant used, for example, to reduce friction between the equipment and granulated mixture during compression of tablet formulations.
Suitable anti-adherents include, but are not limited to, talc, cornstarch, DL- leucine, sodium lauryl sulfate and metallic stearates. Talc is a preferred anti-adherent or glidant used, for example, to reduce formulation sticking to equipment surfaces and also to reduce static in the blend. Talc, if present, constitutes about 0.1% to about 10%, more preferably about 0.25% to about 5%, and still more preferably about 0.5% to about 2%, of the total weight of the pharmaceutical composition.
Glidants can be used to promote powder flow of a solid formulation. Suitable glidants include, but are not limited to, colloidal silicon dioxide, starch, talc, tribasic calcium phosphate, powdered cellulose and magnesium trisilicate. Colloidal silicon dioxide is particularly preferred.
Other excipients such as colorants, flavors and sweeteners are known in the pharmaceutical art and can be used in pharmaceutical compositions of the present invention. Tablets can be coated, for example with an enteric coating, or uncoated. Compositions of the invention can further comprise, for example, buffering agents. Optionally, one or more effervescent agents can be used as disintegrants and/or to enhance organoleptic properties of pharmaceutical compositions of the invention. When present in pharmaceutical compositions of the invention to promote dosage form disintegration, one or more effervescent agents are preferably present in a total amount of about 30% to about 75%, and preferably about 45% to about 70%, for example about 60%, by weight of the pharmaceutical composition.
According to a particularly preferred embodiment of the invention, an effervescent agent, present in a solid dosage form in an amount less than that effective to promote disintegration of the dosage form, provides improved dispersion of the API in an aqueous medium. Without being bound by theory, it is believed that the effervescent agent is effective to accelerate dispersion of the API from the dosage form in the gastrointestinal tract, thereby further enhancing absorption and rapid onset of therapeutic effect. When present in a pharmaceutical composition of the invention to promote intragastrointestinal dispersion but not to enhance disintegration, an effervescent agent is preferably present in an amount of about 1% to about 20%, more preferably about 2.5% to about 15%, and still more preferably about 5% to about 10%, by weight of the pharmaceutical composition.
An "effervescent agent" herein is an agent comprising one or more compounds which, acting together or individually, evolve a gas on contact with water. The gas evolved is generally oxygen or, most commonly, carbon dioxide. Preferred effervescent agents comprise an acid and a base that react in the presence of water to generate carbon dioxide gas. Preferably, the base comprises an alkali metal or alkaline earth metal carbonate or bicarbonate and the acid comprises an aliphatic carboxylic acid.
Non-limiting examples of suitable bases as components of effervescent agents useful in the invention include carbonate salts (e.g., calcium carbonate), bicarbonate salts (e.g., sodium bicarbonate), sesquicarbonate salts, and mixtures thereof. Calcium carbonate is a preferred base.
Non-limiting examples of suitable acids as components of effervescent agents and/or solid organic acids useful in the invention include citric acid, tartaric acid (as D-, L-, or D/L-tartaric acid), malic acid (as D-, L-, or DL-malic acid), maleic acid, fumaric acid, adipic acid, succinic acid, acid anhydrides of such acids, acid salts of such acids, and mixtures thereof. Citric acid is a preferred acid.
In a preferred embodiment of the invention, where the effervescent agent comprises an acid and a base, the weight ratio of the acid to the base is about 1 : 100 to about 100:1, more preferably about 1:50 to about 50:1, and still more preferably about 1 : 10 to about 10:1. In a further preferred embodiment of the invention, where the effervescent agent comprises an acid and a base, the ratio of the acid to the base is approximately stoichiometric. Excipients which solubilize APIs typically have both hydrophilic and hydrophobic regions, or are preferably amphiphilic or have amphiphilic regions. One type of amphiphilic or partially-amphiphilic excipient comprises an amphiphilic polymer or is an amphiphilic polymer. A specific amphiphilic polymer is a polyalkylene glycol, which is commonly comprised of ethylene glycol and/or propylene glycol subunits. Such polyalkylene glycols can be esterified at their termini by a carboxylic acid, ester, acid anhyride or other suitable moiety. Examples of such excipients include poloxamers (symmetric block copolymers of ethylene glycol and propylene glycol; e.g., poloxamer 237), polyalkyene glycolated esters of tocopherol (including esters formed from a di- or multi-functional carboxylic acid; e.g., d-alpha-tocopherol polyethylene glycol- 1000 succinate), and macrogolglycerides (formed by alcoholysis of an oil and esterification of a polyalkylene glycol to produce a mixture of mono-, di- and tri-glycerides and mono- and di-esters; e.g., stearoyl macrogol-32 glycerides). Such pharmaceutical compositions are advantageously administered orally.
Pharmaceutical compositions of the present invention can comprise about 10 % to about 50 %, about 25 % to about 50 %, about 30 % to about 45 %, or about 30 % to about 35 % by weight of a co-crystal; about 10 % to about 50 %, about 25 % to about 50 %, about 30 % to about 45 %, or about 30 % to about 35 % by weight of an excipient which inhibits crystallization in aqueous solution, in simulated gastric fluid, or in simulated intestinal fluid; and about 5 % to about 50 %, about 10 % to about 40 %, about 15 % to about 35 %, or about 30 % to about 35 % by weight of a binding agent. In one example, the weight ratio of the co-crystal to the excipient which inhibits crystallization to binding agent is about 1 to 1 to 1.
Solid dosage forms of the invention can be prepared by any suitable process, not limited to processes described herein.
An illustrative process comprises (a) a step of blending an API of the invention with one or more excipients to form a blend, and (b) a step of tableting or encapsulating the blend to form tablets or capsules, respectively.
In a preferred process, solid dosage forms are prepared by a process comprising (a) a step of blending a co-crystal of the invention with one or more excipients to form a blend, (b) a step of granulating the blend to form a granulate, and (c) a step of tableting or encapsulating the blend to form tablets or capsules respectively. Step (b) can be accomplished by any dry or wet granulation technique known in the art, but is preferably a dry granulation step. A salt of the present invention is advantageously granulated to form particles of about 1 micrometer to about 100 micrometer, about 5 micrometer to about 50 micrometer, or about 10 micrometer to about 25 micrometer. One or more diluents, one or more disintegrants and one or more binding agents are preferably added, for example in the blending step, a wetting agent can optionally be added, for example in the granulating step, and one or more disintegrants are preferably added after granulating but before tableting or encapsulating. A lubricant is preferably added before tableting. Blending and granulating can be performed independently under low or high shear. A process is preferably selected that forms a granulate that is uniform in API content, that readily disintegrates, that flows with sufficient ease so that weight variation can be reliably controlled during capsule filling or tableting, and that is dense enough in bulk so that a batch can be processed in the selected equipment and individual doses fit into the specified capsules or tablet dies.
In an alternative embodiment, solid dosage forms are prepared by a process that includes a spray drying step, wherein an API is suspended with one or more excipients in one or more sprayable liquids, preferably a non-protic (e.g., non-aqueous or nonalcoholic) sprayable liquid, and then is rapidly spray dried over a current of warm air. A granulate or spray dried powder resulting from any of the above illustrative processes can be compressed or molded to prepare tablets or encapsulated to prepare capsules. Conventional tableting and encapsulation techniques known in the art can be employed. Where coated tablets are desired, conventional coating techniques are suitable. Excipients for tablet compositions of the invention are preferably selected to provide a disintegration time of less than about 30 minutes, preferably about 25 minutes or less, more preferably about 20 minutes or less, and still more preferably about 15 minutes or less, in a standard disintegration assay.
Pharmaceutically acceptable co-crystals can be administered by controlled-, sustained-, or delayed-release means. Controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled release counterparts. Ideally, the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled-release formulations include: 1) extended activity of the drug; 2) reduced dosage frequency; 3) increased patient compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects; 6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug activity; and 10) improvement in speed of control of diseases or conditions. (Kim, Cherng-ju, Controlled Release Dosage Form Design, 2 Technomic Publishing, Lancaster, Pa.: 2000).
Conventional dosage forms generally provide rapid or immediate drug release from the formulation. Depending on the pharmacology and pharmacokinetics of the drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like. Advantageously, controlled- release formulations can be used to control a drug's onset of action, duration of action, plasma levels within the therapeutic window, and peak blood levels. In particular, controlled- or extended-release dosage forms or formulations can be used to ensure that the maximum effectiveness of a drug is achieved while minimizing potential adverse effects and safety concerns, which can occur both from under dosing a drug (i.e., going below the minimum therapeutic levels) as well as exceeding the toxicity level for the drug.
Most controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions or compounds. A variety of known controlled- or extended-release dosage forms, formulations, and devices can be adapted for use with the co-crystals and compositions of the mvention. Examples include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365,185 Bl; each of which is incorporated herein by reference. These dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, liydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), multilayer coatings, microparticles, liposomes, or microspheres or a combination thereof to provide the desired release profile in varying proportions. Additionally, ion exchange materials can be used to prepare immobilized, adsorbed co-crystals and thus effect controlled delivery of the drug. Examples of specific anion exchangers include, but are not limited to, Duolite® A568 and Duolite® AP143 (Rohm & Haas, Spring House, PA. USA).
One embodiment of the invention encompasses a unit dosage form which comprises a pharmaceutically acceptable co-crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof, and one or more pharmaceutically acceptable excipients or diluents, wherein the pharmaceutical composition or dosage form is formulated for controlled-release. Specific dosage forms utilize an osmotic drug delivery system.
A particular and well-known osmotic drug delivery system is referred to as OROS® (Alza Corporation, Mountain View, Calif. USA). This technology can readily be adapted for the delivery of compounds and compositions of the invention. Various aspects of the technology are disclosed in U.S. Pat. Nos. 6,375,978 Bl; 6,368,626 Bl; 6,342,249 Bl; 6,333,050 B2; 6,287,295 Bl; 6,283,953 Bl; 6,270,787 Bl; 6,245,357 Bl; and 6,132,420; each of which is incorporated herein by reference. Specific adaptations of OROS® that can be used to administer compounds and compositions of the invention include, but are not limited to, the OROS® Push-Pull™, Delayed Push-Pull™, Multi- Layer Push-Pull™, and Push-Stick™ Systems, all of which are well known. See, e.g., http://www.alza.com. Additional OROS® systems that can be used for the controlled oral delivery of compounds and compositions of the invention include OROS®-CT and L- OROS®. Id.; see also, Delivery Times, vol. II, issue II (Alza Corporation).
Conventional OROS® oral dosage forms are made by compressing a drag powder (e.g. co-crystal) into a hard tablet, coating the tablet with cellulose derivatives to form a semi-permeable membrane, and then drilling an orifice in the coating (e.g., with a laser). Kim, Cherng-ju, Controlled Release Dosage Form Design, 231-238 (Technomic Publishing, Lancaster, Pa.: 2000). The advantage of such dosage forms is that the delivery rate of the drug is not influenced by physiological or experimental conditions. Even a drug with a pH-dependent solubility can be delivered at a constant rate regardless of the pH of the delivery medium. But because these advantages are provided by a buildup of osmotic pressure within the dosage form after administration, conventional OROS® drug delivery systems cannot be used to effectively deliver drugs with low water solubility. Id. at 234. Because co-crystals of this invention can be far more soluble in water than the API itself, they are well suited for osmotic-based delivery to patients. This invention does, however, encompass the incorporation of conventional crystalline API (e.g. pure API without co-crystal former), and non-salt isomers and isomeric mixtures thereof, into OROS® dosage forms.
A specific dosage form of the invention comprises: a wall defining a cavity, the wall having an exit orifice formed or formable therein and at least a portion of the wall being semipermeable; an expandable layer located within the cavity remote from the exit orifice and in fluid communication with the semipermeable portion of the wall; a dry or substantially dry state drug layer located within the cavity adjacent to the exit orifice and in direct or indirect contacting relationship with the expandable layer; and a flow- promoting layer interposed between the inner surface of the wall and at least the external surface of the drug layer located within the cavity, wherein the drug layer comprises a co- crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof. See U.S. Pat. No. 6,368,626, the entirety of which is incorporated herein by reference.
Another specific dosage form of the invention comprises: a wall defining a cavity, the wall having an exit orifice formed or formable therein and at least a portion of the wall being semipermeable; an expandable layer located within the cavity remote from the exit orifice and in fluid communication with the semipermeable portion of the wall; a drug layer located within the cavity adjacent the exit orifice and in direct or indirect contacting relationship with the expandable layer; the drug layer comprising a liquid, active agent formulation absorbed in porous particles, the porous particles being adapted to resist compaction forces sufficient to form a compacted drug layer without significant exudation of the liquid, active agent formulation, the dosage form optionally having a placebo layer between the exit orifice and tlie drug layer, wherein the active agent formulation comprises a co-crystal, or a solvate, hydrate, dehydrate, anhydrous, or amorphous form thereof. See U.S. Pat. No. 6,342,249, the entirety of which is incorporated herein by reference.
The invention will now be described in further detail, by way of example, with reference to the accompanying drawings.
EXEMPLIFICATION
General Methods for the Preparation of Co-Crystals
a) High Throughput crystallization using the CrystalMax™ platform
CrystalMax™ comprises a sequence of automated, integrated high throughput robotic stations capable of rapid generation, identification and characterization of polymorphs, salts, and co-crystals of APIs and API candidates. Worksheet generation and combinatorial mixture design is carried out using proprietary design software Architect™. Typically, an API or an API candidate is dispensed from an organic solvent into tubes and dried under a stream of nitrogen. Salts and/or co-crystal formers may also be dispensed and dried in the same fashion. Water and organic solvents may be combinatorially dispensed into the tubes using a multi-channel dispenser. Each tube in a 96-tube array is then sealed within 15 seconds of combinatorial dispensing to avoid solvent evaporation. The mixtures are then rendered supersaturated by heating to 70 degrees C for 2 hours followed by a 1 degree C/minute cooling ramp to 5 degrees C. Optical checks are then conducted to detect crystals and/or solid material. Once a solid has been identified in a tube, it is isolated through aspiration and drying. Raman spectra are then obtained on the solids and cluster classification of the spectral patterns is performed using proprietary software (Inquire™).
b) Crystallization from solution
Co-crystals may be obtained by dissolving the separate components in a solvent and adding one to the other. The co-crystal may then precipitate or crystallize as the solvent mixture is evaporated slowly. The co-crystal may also be obtained by dissolving the two components in the same solvent or a mixture of solvents.
c) Crystallization from the melt (Co-melting)
A co-crystal may be obtained by melting the two components together (i.e., co- melting) and allowing recrystallization to occur. In some cases, an anti-solvent may be added to facilitate crystallization.
d) Thermal microscopy
A co-crystal may be obtained by melting the higher melting component on a glass slide and allowing it to recrystallize. The second component is then melted and is also allowed to recrystallize. The co-crystal may form as a separated phase/band in between the eutectic bands of the two original components.
e) Mixing and/or grinding
A co-crystal may be obtained by mixing or grinding two components together in the solid state.
f) Co-sublimation
A co-crystal may be obtained by co-subliming a mixture of an API and a co- crystal former in the same sample cell as an intimate mixture either by heating, mixing or placing the mixture under vacuum. A co-crystal may also be obtained by co-sublimation using a Kneudsen apparatus where the API and the co-crystal former are contained in separate sample cells, connected to a single cold finger, each of the sample cells is maintained at the same or different temperatures under a vaccum atmosphere in order to co-sublime the two components onto the cold-finger forming the desired co-crystal.
Analytical Methods
Procedure for DSC analysis
DSC analysis of the samples was performed using a Q1000 Differential Scanning
Calorimeter (TA Instruments, New Castle, DE, U.S.A.), which uses Advantage for QW-
Series, version 1.0.0.78, Thermal Advantage Release 2.0 (2001 TA Instruments- Water
LLC). In addition, the analysis software used was Universal Analysis 2000 for Windows
95/95/2000/NT, version 3.1E;Build 3.1.0.40 (2001 TA Instruments-Water LLC).
For the DSC analysis, the purge gas used was dry nitrogen, the reference material was an empty aluminum pan that was crimped, and the sample purge was 50 mL/minute.
DSC analysis of the sample was performed by placing < 2 mg of sample in an aluminum pan with a crimped pan closure. The starting temperature was typically 20 degrees C with a heating rate of 10 degrees C/minute, and the ending temperature was 300 degrees C. Unless otherwise indicated, all reported transitions are as stated +/- 1.0 degrees C.
Procedure for TGA analysis
TGA analysis of samples was performed using a Q500 Thermogravimetric Analyzer (TA Instruments, New Castle, DE, U.S.A.), which uses Advantage for QW- Series, version 1.0.0.78, Thermal Advantage Release 2.0 (2001 TA Instruments-Water LLC). In addition, the analysis software used was Universal Analysis 2000 for Windows 95/95/2000/NT, version 3.1E;Build 3.1.0.40 (2001 TA Instruments-Water LLC).
For all of the TGA experiments, the purge gas used was dry nitrogen, the balance purge was 40 mL/minute N2, and the sample purge was 60 mL/minute N2.
TGA of the sample was performed by placing < 2 mg of sample in a platinum pan. The starting temperature was typically 20 degrees C with a heating rate of 10 degrees C/minute, and the ending temperature was 300 degrees C. Procedure for PXRD analysis
A powder X-ray diffraction pattern for the samples was obtained using a D/Max Rapid, Contact (Rigaku/MSC, The Woodlands, TX, U.S.A.), which uses as its control software RINT Rapid Control software, Rigaku Rapid/XRD, version 1.0.0 (1999 Rigaku Co.). In addition, the analysis software used were RINT Rapid display software, version 1.18 (Rigaku MSC), and JADE XRD Pattern Processing, versions 5.0 and 6.0 ((1995- 2002, Materials Data, Inc.).
For the PXRD analysis, the acquisition parameters were as follows: source was Cu with a K line at 1.5406A; x-y stage was manual; collimator size was 0.3 or 0.8 mm; capillary tube (Charles Supper Company, Natick, MA, U.S.A.) was 0.3 mm ID; reflection mode was used; the power to the X-ray tube was 46 kV; the current to the X-ray tube was 40 mA; the omega-axis was oscillating in a range of 0-5 degrees at a speed of 1 degree/minute; the phi-axis was spinning at an angle of 360 degrees at a speed of 2 degrees/second; 0.3 or 0.8 mm collimator; the collection time was 60 minutes; the temperature was room temperature; and the heater was not used. The sample was presented to the X-ray source in a boron rich glass capillary.
In addition, the analysis parameters were as follows: the integration 2-theta range was 2-40 or 60 degrees; the integration chi range was 0-360 degrees; the number of chi segments was 1; the step size used was 0.02; the integration utility was cylint; normalization was used; dark counts were 8; omega offset was 180; and chi and phi offsets were 0.
The relative intensity of peaks in a diffractogram is not necessarily a limitation of the PXRD pattern because peak intensity can vary from sample to sample, e.g., due to crystalline impurities. Further, the angles of each peak can vary by about +/- 0.1 degrees, preferably +/-0.05. The entire pattern or most of the pattern peaks may also shift by about +/- 0.1 degree due to differences in calibration, settings, and other variations from instrument to instrument and from operator to operator. Procedure for Raman Acquisition. Filtering and Binning
Acquisition
The sample was either left in the glass vial in which it was processed or an aliquot of the sample was transferred to a glass slide. The glass vial or slide was positioned in the sample chamber. The measurement was made using an Almega™ Dispersive Raman (Almega™ Dispersive Raman, Thermo-Nicolet, 5225 Verona Road, Madison, WI 53711- 4495) system fitted with a 785nm laser source. The sample was manually brought into focus using the microscope portion of the apparatus with a lOx power objective (unless otherwise noted), thus directing the laser onto the surface of the sample. The spectrum was acquired using the parameters outlined in Table XXII. (Exposure times and number of exposures may vary; changes to parameters will be indicated for each acquisition.)
Filtering and Binning
Each spectrum in a set was filtered using a matched filter of feature size 25 to remove background signals, including glass contributions and sample fluorescence. This is particularly important as large background signal or fluorescence limit the ability to accurately pick and assign peak positions in the subsequent steps of the binning process. Filtered spectra were binned using the peak pick and bin algorithm with the parameters given in Table XXIII. The sorted cluster diagrams for each sample set and the corresponding cluster assignments for each spectral file were used to identify groups of samples with similar spectra, which was used to identify samples for secondary analyses.
Table XXII. Raman Spectral acquisition parameters
Figure imgf000062_0001
Procedure for Single Crystal X-Ray Diffraction
Single crystal x-ray data were collected on a Bruker SMART-APEX CCD diffractometer (M. J. Zaworotko, Department of Chemistry, University of South Florida). Lattice parameters were determined from least squares analysis. Reflection data was integrated using the program SAINT. The structure was solved by direct methods and refined by full matrix least squares using the program SHELXTL (Sheldrick, G. M. SHELXTL, Release 5.03; Siemans Analytical X-ray Instruments Inc.: Madison, WI).
The co-crystals of the present invention can be characterized, e.g., by the TGA or DSC data or by any one, any two, any three, any four, any five, any six, any seven, any eight, any nine, any ten, or any single integer number of PXRD 2-theta angle peaks or Raman shift peaks listed herein or disclosed in a figure, or by single crystal x-ray diffraction data.
Example 1
1 : 1 celecoxib icotinamide co-crystals were prepared. Celecoxib (100 mg, 0.26 mmol) and nicotinamide (32.0 mg, 0.26 mmol) were each dissolved in acetone (2 mL). The two solutions were mixed and the resulting mixture was allowed to evaporate slowly overnight. The precipitated solid was redissolved in acetone a second time and left to evaporate to dryness. The powder was collected and characterized. Detailed characterization of the celecoxib icotinamide co-crystal is listed in Table XXIV. Fig. 1A shows the PXRD diffractogram after subtraction of background noise. Fig. IB shows the raw PXRD data. Fig. 2 shows a DSC thermogram of the celecoxib :nicotinamide co- crystal. Fig. 3 shows a TGA thermogram of the celecoxib :nicotinamide co-crystal. Fig. 4 shows a Raman spectrum of the celecoxib:nicotinamide co-crystal.
Example 2
Co-crystals of celecoxib and 18-crown-6 were prepared. A solution of celecoxib (157.8 mg, 0.4138 mmol) in Et2O (10.0 mL) was added to 18-crown-6 (118.1 mg, 0.447 mmol). The opaque solid dissolves immediately and a white solid subsequently began to crystallize very rapidly. The solid was collected via filtration and was washed with additional diethyl ether (5 L). Detailed characterization of the celecoxib: 18-crown-6 co-crystal is listed in Table XXIV. Fig. 5A shows the PXRD diffractogram after subtraction of background noise. Fig. 5B shows the raw PXRD data. Fig. 6 shows a DSC thermogram of the celecoxib: 18-crown-6 co-crystal. Fig. 7 shows a TGA thermogram of the celecoxib: 18-crown-6 co-crystal.
Example 3
Co-crystals of topiramate and 18-crown-6 were prepared. To topiramate (100 mg, 0.29 mmol) dissolved in diethyl ether (5 mL) was added 18-crown-6 (78 mg, 0.29 mmol) in diethyl ether (5 mL). Upon addition of 18-crown-6, the solution became cloudy and was sonicated for 30 seconds. The solution was left standing for 1 hour and a colorless precipitate was observed. The precipitate was collected, washed with diethyl ether and dried to give a 1 :1 co-crystal of topiramate: 18-crown-6 as a colorless solid. Detailed characterization of the co-crystal is listed in Table XXIV. Fig. 8 A shows the PXRD diffractogram after subtraction of background noise. Fig. 8B shows the raw PXRD data. Fig. 9 shows a DSC thermogram of the topiramate: 18-crown-6 co-crystal.
Example 4
Co-crystals of olanzapine and nicotinamide (Forms I, II and III) were prepared. A 9-block experiment was designed with 12 solvents. (A block is a receiving plate, which can be, for example, an industry standard 24 well, 96 well, 384 well, or 1536 well format, or a custom format.) 864 crystallization experiments with 10 co-crystal formers and 3 concentrations were carried out using the CrystalMax™ platform. Form I was obtained from mixtures containing 1:1 and 1:2 molar ratios of olanzapine:nicotinamide in 1,2- dichloroethane. Form II was obtained from mixtures containing a 1 :2 molar ratio of olanzapine and nicotinamide in isopropyl acetate. PXRD and DSC characterization of the olanzapine:nicotinamide co-crystals are listed in Table XXIV. Fig. 10A shows the PXRD diffractogram of form I after subtraction of background noise. Fig. 10B shows the raw PXRD data of form I. Fig. 11 shows a DSC thermogram of the olanzapine :nicotinamide form I co-crystal. Fig. 12 shows the PXRD diffractogram of olanzapinemicotinamide form II after subtraction of background noise. Co-crystals of olanzapine and nicotinamide (Form III) were prepared. Olanzapine (40 microliters of 25 mg/mL stock solution in tetrahydrofuran) and nicotinamide (37.6 microliters of 20 mg/mL stock solution in methanol) were added to a glass vial and dried under a flow of nitrogen. To the solid mixture was added isopropyl acetate (100 microliters) and the vial was sealed with an aluminum cap. The suspension was then heated at 70 degrees C for two hours in order to dissolve all of the solid material. The solution was then cooled to 5 degrees C and maintained at that temperature for 24 hours. After 24 hours the vial was uncapped and the mixture was concentrated to 50 microliters of total volume. The vial was then resealed with an aluminum cap and was maintained at 5 degrees C for an additional 24 hours. Large, yellow plates were observed and were collected (Form III). The solid was characterized with single crystal x-ray diffraction and powder x-ray diffraction. PXRD characterization of the co-crystal is listed in Table XXIV. Fig. 13 A shows the PXRD diffractogram of form III after subtraction of background noise. Fig. 13B shows the raw PXRD data of form III. Figs. 14A-D show packing diagrams of the olanzapine:nicotinamide form III co-crystal.
Single crystal x-ray analysis reveals that the olanzapine :nicotinamide (Form III) co-crystal is made up of a ternary system containing olanzapine, nicotinamide, water and isopropyl acetate in the unit cell. The co-crystal crystallizes in the monoclinic space group P2i/c and contains two olanzapine molecules, one nicotinamide molecule, 4 water molecules and one isopropyl acetate molecule in the asymmetric unit. The packing diagram is made up of a two-dimensional hydrogen-bonded network with the water molecules connecting the olanzapine and nicotinamide moieties. The packing diagram is also comprised of alternating olanzapine and nicotinamide layers connected through hydrogen bonding via the water and isopropyl acetate molecules, as shown in Figure 14B. The olanzapine layer propagates along the b axis at c/4 and 3c/4. The nicotinamide layer propagates along the b axis at c/2. The top of Figure 14C illustrates the nicotinamide superstructure. The nicotinamide molecules form dimers which hydrogen bond to chains of 4 water molecules. The water chains terminate with isopropyl acetate molecules on each side.
Crystal data: C45H6 N10O7S2, M = 921.18, monoclinic P21/c; a = 14.0961(12) A, b = 12.5984(10) A, c = 27.219(2) A, α = 90°, β = 97.396(2)°, γ = 90°, T = 100(2) K, Z = 4, Dc = 1.276 Mg/m3,U = 4793.6(7) A3, λ = 0.71073 A; 24952 reflections measured, 8457 unique (Rint = 0.0882). Final residuals were R, = 0.0676, wR2 = 0.1461 for I>2σ(I), and Ri = 0.1187, wR2 = 0.1687 for all 8457 data.
Example 5
A co-crystal of cw-itraconazole and succinic acid was prepared. To a solution of succinic acid (16.8 mg, 0.142 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol). A clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form. The solid was collected by filtration and washed with cold THF (2 mL). The white solid was air-dried and placed in a glass vial. The crystalline substance was found to be a succinic acid co-crystal of cz's-itraconazole. The solid was characterized by PXRD and DSC. Fig. 15 shows the PXRD diffractogram after subtraction of background noise. Fig. 16 shows a DSC thermogram of the co-crystal.
Example 6
A co-crystal of czAitraconazole and fumaric acid was prepared. To a blend of fumaric acid (8.40 mg, 0.072 mmol) and czAitraconazole (51.8 mg, 0.073 mmol) was added tetrahydrofuran (THF) (1.0 mL). A clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), no crystals formed. To the clear solution was added t-butyl methyl ether (1.0 mL). A white solid formed immediately and was collected by filtration and washed with cold t-butyl methyl ether (2 mL). The white solid was air-dried and placed in a glass vial. The crystalline substance was found to be a fumaric acid co-crystal of czAitraconazole. The solid was characterized by PXRD and DSC. Fig. 17 shows the PXRD diffractogram after subtraction of background noise. Fig. 18 shows a DSC thermogram of the co-crystal. Example 7
A co-crystal of czAitraconazole and L-tartaric acid was prepared. To a solution of L-tartaric acid (21.3 mg, 0.142 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol). A clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form. The solid was collected by filtration and washed with cold THF (2 mL). The white solid was air-dried and placed in a glass vial. The crystalline substance was found to be an L- tartaric acid co-crystal of czAitraconazole. The solid was characterized by PXRD and DSC. Fig. 19 shows the PXRD diffractogram after subtraction of background noise. Fig. 20 shows a DSC thermogram of the co-crystal.
Example 8
A co-crystal of czAitraconazole and L-malic acid was prepared. To a solution of L-malic acid (19.1 mg, 0.143 mmol) in tetrahydrofuran (THF) (0.50 mL) was added cis- itraconazole (100 mg, 0.142 mmol). A clear solution formed with heating (60 degrees C) and stirring. Upon cooling to room temperature (25 degrees C), crystals began to form. The solid was collected by filtration and washed with cold THF (2 mL). The white solid was air-dried and placed in a glass vial. The crystalline substance was found to be an L- malic acid co-crystal of czAitraconazole. The solid was characterized by PXRD and DSC. Fig. 21 shows the PXRD diffractogram after subtraction of background noise. Fig. 22 shows a DSC thermogram of the co-crystal.
Example 9
A co-crystal of czAitraconazole hydrochloride and DL-tartaric acid was prepared. To a suspension of c/Aitraconazole freebase (20.1 g, 0.0285 mol) in absolute ethanol (100 mL) was added a solution of hydrochloric acid (1.56 g, 0.0428 mol) and DL-tartaric acid (2.99 g, 0.0171mol) in absolute ethanol (100 mL). A clear solution formed with stirring and heating to reflux. The hot solution was gravity filtered and allowed to cool to room temperature (25 degrees C). Upon cooling white crystals formed. The solid was collected by filtration and washed with cold absolute ethanol (15 mL). The white solid was dried in a vacuum oven overnight at 80 degrees C. The crystalline substance was found to be a DL-tartaric acid co-crystal of ezAitraconazole hydrochloride. The solid was characterized by PXRD and DSC. Fig. 23 shows the PXRD diffractogram after subtraction of background noise. Fig. 24 shows a DSC thermogram of the co-crystal.
Example 10
Co-crystals of modafinil and malonic acid were prepared. Using a 250 mg/ml modafinil-acetic acid solution, malonic acid was dissolved on a hotplate (about 67 degrees C) at a 1 :2 modafinil to malonic acid ratio. The mixture was dried under flowing nitrogen overnight. A powdery white solid was produced. After further drying for 1 day, acetic acid was removed (as determined by TGA) and the crystal structure of the modafinikmalonic acid (Form I) co-crystal, as determined by PXRD, remained the same. The modafinil :malonic acid (Form I) co-crystal was also prepared by grinding the API and co-crystal former together. 2.50 g of modafinil was mixed with 1.01 g of malonic acid in a large mortar and pestle (malonic acid added in increments over 7 days with about a 1 : 1.05 ratio made on the first day and increments added over the next seven days which resulted in a 1 :2 modafinil :malonic acid ratio). The mixture was ground for 45 minutes initially and 20 minutes each time more malonic acid was added. On the seventh day the mixture of co-crystal and starting components was heated in a sealed 20 mL vial at 80 degrees C for about 35 minutes to facilitate completion of the co-crystal formation. PXRD analysis of the resultant material was completed and the diffractogram is shown in Fig. 25, after subtraction of background noise. Fig. 26 shows a DSC thermogram of the modafinil :malonic acid Form I co-crystal. Fig. 27 shows the Raman spectrum of the modafinil :malonic acid Form I co-crystal. Fig. 27 comprises peaks, in order of decreasing intensity, of 1004, 222, 633, 265, 1032, 1183, 814, 1601, 490, 718, 767, 361, 917, 1104, 889, 412, 1225, 1251, 1398, 1442, 1731, 1298, 3065, and 2949 cm"1. Single crystal data of the modafinikmalonic acid Form I co-crystal were acquired and are reported below. Crystal data: C18H19NO6S, M = 377.40, monoclinic C2/c; a = 18.728(8) angstroms, b = 5.480(2) angstroms, c = 33.894(13) angstroms, alpha = 90 degrees, beta = 91.864(9) degrees, gamma = 90 degrees, T - 100(2) K, Z = 8, D0 = 1.442 Mg/m3, U = 3477(2) cubic angstroms, λ = 0.71073 angstroms, 6475 reflections measured, 3307 unique (Rj„t = 0.1567). Final residuals were Ri = 0.1598, wR2 = 0.3301 for I>2sigma(I), and Rj = 0.2544, wR2 = 0.3740 for all 3307 data.
A polymorph of the modafinikmalonic acid Form I co-crystal was prepared in a vial. 11.4 mg of modafinil and 8.9 mg of malonic acid were dissolved in 2 mL of acetone. The solids dissolved at room temperature, and the vial was left open to evaporate the solvent in air. Large parallelogram shaped crystals formed on the walls and bottom of the vial. The PXRD diffractogram of the large crystals showed modafinil :malonic acid co-crystals Form II, a polymorphic form of modafinil :malonic acid Form I. Fig. 28 shows the PXRD diffractogram of the modafinil :malonic acid Form II co-crystal after subtraction of background noise.
Example 11
Co-crystals of modafinil and glycolic acid were prepared. Modafinil (1 mg, 0.0037mmol) and glycolic acid (0.30 mg, 0.0037 mmol) were dissolved in acetone (400 microliters). The solution was allowed to evaporate to dryness and the resulting solid was characterized using PXRD. PXRD data for the modafinil: glycolic acid co-crystal is listed in Table XXIV. Fig. 29A shows the PXRD diffractogram after subtraction of background noise. Fig. 29B shows the raw PXRD data.
Example 12
Co-crystals of modafinil and maleic acid were prepared. Using a 250 mg/ml modafinil-acetic acid solution, maleic acid was dissolved on a hotplate (about 67 degrees C) at a 2:1 modafinil to maleic ratio. The mixture was dried under flowing nitrogen overnight. A clear amorphous material remained. Solids began to grow after 2 days stored in a sealed vial at room temperature. The solid was collected and characterized as the modafinil :maleic acid co-crystal using PXRD. Fig. 30A shows the PXRD diffractogram after subtraction of background noise. Fig. 3 OB shows the raw PXRD data.
Example 13
Co-crystals of 5-fluorouracil and urea were prepared. To 5-fluorouracil (Ig, 7.69 mmol) and urea (0.46g, 7.69 mmol) was added methanol (100 mL). The solution was heated at 65 degrees C and sonicated until all the material dissolved. The solution was then cooled to 5 degrees C and maintained at that temperature overnight. After about 3 days a white precipitate was observed and collected. The solid was characterized by DSC, PXRD, Raman spectroscopy, and TGA as the 5-fluorouracil:urea co-crystal. Characterization data are listed in Table XXIV. Fig. 31 A shows the PXRD diffractogram after subtraction of background noise. Fig. 3 IB shows the raw PXRD data. Fig. 32 shows a DSC thermogram of the 5-fluorouracil:urea co-crystal. Fig. 33 shows a TGA thermogram of the 5-fluorouracil:urea co-crystal. Fig. 34 shows a Raman spectrum of the 5-fluorouracil:urea co-crystal. Single crystal data of the 5-fluorouracil:urea co- crystal were acquired and are reported below.
Crystal data: C5H7FN4O3, M = 190.15, monoclinic C2/C, a = 9.461(3) angstroms, b = 10.487(3) angstroms, c - 15.808(4) angstroms, alpha = 90 degrees, beta = 99.891(5), gamma = 90 degrees, T = 100(2) K, Z = 8, Dc = 1.635 Mg/m3, U = 1545.2(7) cubic angstroms, λ = 0.71073 angstroms, 3419 reflections measured, 1633 unique (Rlnt = 0.0330). Final residuals were Ri = 0.0667, wR2 = 0.1505 for I>2sigma(I), and Ri = 0.0872, wR2 = 0.1598 for all 1633 data.
Example 14
Co-crystals of hydrochlorothiazide and nicotinic acid were prepared. Hydrochlorothiazide (12.2 mg, 0.041 mmol) and nicotinic acid (5 mg, 0.041 mmol) were dissolved in methanol (1 mL). The solution was then cooled to 5 degrees C and maintained at that temperature for 12 hours. A white solid precipitated and was collected and characterized as the hydrochlorothiazide:nicotinic acid co-crystal using PXRD. Fig. 35A shows the PXRD diffractogram after subtraction of background noise. Fig. 35B shows the raw PXRD data.
Example 15
Co-crystals of hydrochlorothiazide and 18-crown-6 were prepared. Hydrochlorothiazide (100 mg, 0.33 mmol) was dissolved in diethyl ether (15 mL) and was added to a solution of 18-crown-6 (87.2 mg, 0.33 mmol) in diethyl ether (15 mL). A white precipitate immediately began to form and was collected and characterized as the hydrochlorothiazide: 18-crown-6 co-crystal using PXRD. Fig. 36A shows the PXRD diffractogram after subtraction of background noise. Fig. 36B shows the raw PXRD data.
Example 16
Co-crystals of hydrochlorothiazide and piperazine were prepared. Hydrochlorothiazide (17.3 mg, 0.058 mmol) and piperazine (5 mg, 0.058 mmol) were dissolved in a 1:1 mixture of ethyl acetate and acetonitrile (1 mL). The solution was then cooled to 5 degrees C and maintained at that temperature for 12 hours. A white solid precipitated and was collected and characterized as the hydrochlorothiazide :piperazine co-crystal using PXRD. Fig. 37A shows the PXRD diffractogram after subtraction of background noise. Fig. 37B shows the raw PXRD data.
Example 17
Acetaminophen:4,4'-bipyridine:water (1:1:1 stoichiometry)
50 mg (0.3307 mmol) acetaminophen and 52 mg (0.3329 mmol) 4,4'-bipyridine were dissolved in hot water and allowed to stand. Slow evaporation yielded colorless needles of a 1 : 1 : 1 acetaminophen:4,4 '-bipyridine: water co-crystal, as shown in Figs. 38A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). triclinic, space group PI; a = 7.0534(8), b = 9.5955(12), c = 19.3649(2) A, α = 86.326(2), β = 80.291(2), γ = 88.880(2)°, U= 1308.1(3) A3, T = 200(2) K, Z = 2, μ(Mo-Kα) = 0.090 mm"1,
Dc = 1.294 Mg/m3, λ = 0.71073 A, F(000) = 537, 2θmax = 25.02°; 6289 reflections measured, 4481 unique (Rjnt = 0.0261). Final residuals for 344 parameters were
Ri = 0.0751, wR2 = 0.2082 for I>2σ(I), and i = 0.1119, wR2 = 0.2377 for all 4481 data.
Crystal packing: The co-crystals contain bilayered sheets in which water molecules act as a hydrogen bonded bridge between the network bipyridine moieties and the acetaminophen. Bipyridine guests are sustained by π-π stacking interactions between two network bipyridines. The layers stack via π-π interactions between the phenyl groups of the acetaminophen moieties.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 57.77 degrees C (endotherm); m.p. = 58-60 degrees C (MEL-TEMP); (acetaminophen m.p. = 169 degrees C, 4,4 '-bipyridine m.p. = 111-114 degrees C).
Example 18
PhenytoinPyridone (1:1 stoichiometry)
28 mg (0.1109 mmol) phenytoin and 11 mg (0.1156 mmol) 4-hydroxypyridone were dissolved in 2 mL acetone and 1 mL ethanol with heating and stirring. Slow evaporation yielded colorless needles of a 1:1 phenytoi pyridone co-crystal, as shown in Figs. 39A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C20H17N3O3, M = 347.37, monoclinic P2j/c; a = 16.6583(19), b = 8.8478(10), c = 11.9546(14) A, β = 96.618(2)°, U= 1750.2(3) A3, T = 200(2) K, Z = 4, μ(Mo-Kα) = 0.091 mm"1, D0 = 1.318 Mg/m3, λ = 0.71073 A, F(000) = 728, 2θmax = 56.60°; 10605 reflections measured, 4154 unique (Ri„t = 0.0313). Final residuals for 247 parameters were Ri = 0.0560, wR = 0.1356 for I>2σ(I), and Ri = 0.0816, wR2 = 0.1559 for all 4154 data.
Crystal packing: The co-crystal is sustained by hydrogen bonding of adjacent phentoin molecules between the carbonyl and the amine closest to the tetrahedral carbon, and by hydrogen bonding between pyridone carbonyl functionalities and the amine not involved in phenytoin-phenytoin interactions. The pyridone carbonyl also hydrogen bonds with adjacent pyridone molecules forming a one-dimensional network. Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), characteristic peaks for the co-crystal were identified as: 2° amine found at 3311cm"1, carbonyl (ketone) found at 1711cm"1, olephin peak found at 1390cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 233.39 degrees C (endotherm) and 271.33 degrees C (endotherm); m.p. = 231-233 degrees C (MEL- TEMP); (phenytoin m.p. = 295 degrees C, pyridone m.p. = 148 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), a 29.09% weight loss starting at 192.80 degrees C, 48.72% weight loss starting at 238.27 degrees C, and 18.38% loss starting at 260.17 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu K (λ = 1.540562), 30kV, 15mA). The powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 2Θ and a scan speed of 2.0 minute. PXRD: Showed analogous peaks to the simulated PXRD derived from the single crystal data, experimental (calculated): 5.2 (5.3); 11.1 (11.3); 15.1 (15.2); 16.2 (16.4); 16.7 (17.0); 17.8 (17.9); 19.4 (19.4); 19.8 (19.7); 20.3 (20.1); 21.2 (21.4); 23.3 (23.7); 26.1 (26.4); 26.4 (26.6); 27.3 (27.6); 29.5 (29.9),
Example 19
Aspirin (acetylsalicylic acid): 4,4 '-bipyridine (2:1 stoichiometry)
50 mg (0.2775 mmol) aspirin and 22 mg (0.1388 mmol) 4,4 '-bipyridine were dissolved in 4 mL hexane. 8 mL ether was added to the solution and allowed to stand for one hour, yielding colorless needles of a 2:1 aspirin:4,4' -bipyridine co-crystal, as shown in Figs. 40A-D. Alternatively, aspirin:4,4' -bipyridine (2:1 stoichiometry) can be made by grinding the solid ingredients in a pestle and mortar.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C28H24N2O8, M = 516.49, orthorhombic Pbcn; a = 28.831(3), b = 11.3861(12), c = 8.4144(9) A, U= 2762.2(5) A3, T = 173(2) K, Z = 4, μ(Mo-Kα) = 0.092 mm"1, Dc = 1.242 Mg/m3, λ = 0.71073 A, F(000) = 1080, 2θmax = 25.02°; 12431 reflections measured, 2433 unique (Rint = 0.0419). Final residuals for 202 parameters were Ri = 0.0419, wR2 = 0.1358 for I>2σ(I), and Ri = 0.0541, wR2 = 0.1482 for all 2433 data.
Crystal packing: The co-crystal contains the carboxylic acid-pyridine heterodimer that crystallizes in the Pbcn space group. The structure is an inclusion compound containing disordered solvent in the channels. In addition to the dominant hydrogen bonding interaction of the heterodimer, π-π stacking of the bipyridine and phenyl groups of the aspirin and hydrophobic interactions contribute to the overall packing interactions.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), characteristic (-COOH) peak at 1679 cm"1 was shifted up and less intense at 1694cm" , where as the lactone peak is shifted down slightly from 1750cm"1 to 1744cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 95.14 degrees C (endotherm); m.p. = 91-96 degrees C (MEL-TEMP); (aspirin m.p. = 1345 degrees C, 4,4'-bipyridine m.p. = 111-114 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), weight loss of 9% starting at 22.62 degrees C, 49.06% weight loss starting at 102.97 degrees C followed by complete decomposition starting at 209.37 degrees C.
Example 20
Ibuprofen:4,4' -Bipyridine (2:1 stoichiometry)
50 mg (0.242 mmol) racemic ibuprofen and 18mg (0.0960 mmol) 4,4'- bipyridine were dissolved in 5 mL acetone. Slow evaporation of the solvent yielded colorless needles of a 2:1 ibuprofen:4,4'-bipyridine co-crystal, as shown in Figs. 41 A-D.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C36H4 N2O4, M = 568.73, triclinic, space group P-l; a = 5.759(3), b = 11.683(6), c = 24.705(11) A, α = 93.674(11), β = 90.880(10), γ = 104.045(7)°, U= 1608.3(13) A3, T= 200(2) K, Z = 2, μ(Mo-Kα) = 0.076 mm"1, Dc = 1.174 Mg/m3, λ = 0.71073 A, F(000) = 612, 2θmax = 23.29°; 5208 reflections measured, 3362 unique (Rjnt = 0.0826). Final residuals for 399 parameters were Ri = 0.0964, wR2 = 0.2510 for I>2σ(I), and Ri = 0.1775, wR2 = 0.2987 for all 3362 data. Crystal packing: The co-crystal contains ibuprofembipyridine heterodimers, sustained by two hydrogen bonded carboxylic acidpyridine supramolecular synthons, arranged in a herringbone motif that packs in the space group P-1. The heterodimer is an extended version of the homodimer and packs to form a two-dimensional network sustained by π-π stacking of the bipyridine and phenyl groups of the ibuprofen and hydrophobic interactions from the ibuprofen tails.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). Analysis observed stretching of aromatic C-H at 2899 cm"1; N--H bending and scissoring at 1886 cnx^; C=O stretching at 1679 cm"1; C-H out-of-plane bending for both 4,4 '-bipyridine and ibuprofen at 808 cm" 1 and 628 cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 64.85 degrees C (endotherm) and 118.79 degrees C (endotherm); m.p. = 113-120 degrees C (MEL- TEMP); (ibuprofen m.p. = 75-77 degrees C, 4,4'-bipyridine m.p. = 111-114 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 13.28% weight loss between room temperature and 100.02 degrees C immediately followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30kV, 15mA). The powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 20 and a scan speed of 2.0 minute. PXRD derived from the single crystal data, experimental (calculated): 3.4 (3.6); 6.9 (7.2); 10.4 (10.8); 17.3 (17.5); 19.1 (19.7).
Example 21
Flurbiprofen:4,4'-bipyridine (2:1 stoichiometry)
50 mg (0.2046 mmol) flurbiprofen and 15 mg (0.0960 mmol) 4,4 '-bipyridine were dissolved in 3 mL acetone. Slow evaporation of the solvent yielded colorless needles of a 2:1 flurbiprofen:4,4'-bipyridine co-crystal, as shown in Figs. 42A-D.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C40H34F2N2O4, M = 644.69, monoclinic P2 /n\ a = 5.860(4), b = 47.49(3), c = 5.928(4) A, β = 107.382 (8)°, U= 1574.3(19) A3, T = 200(2) K, Z = 2, μ(Mo-Kα) = 0.096 mm"1, Dc = 1.360 Mg/m3, λ = 0.71073 A, F(000) = 676, 2θraax = 21.69°; 4246 reflections measured, 1634 unique (Rnt = 0.0677). Final residuals for 226 parameters were Rt = 0.0908, wR2 = 0.2065 for I>2σ(I), and Ri = 0.1084, wR2 = 0.2209 for all 1634 data.
Crystal packing: The co-crystal contains flurbiprofe bipyridine heterodimers, sustained by two hydrogen bonded carboxylic acidpyridine supramolecular synthon, arranged in a herringbone motif that packs in the space group P2j/n. The heterodimer is an extended version of the homodimer and packs to form a two-dimensional network sustained by π-π stacking and hydrophobic interactions of the bipyridine and phenyl groups of the flurbiprofen.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), aromatic C-H stretching at 3057 cm"1 and 2981 cm"1; N--H bending and scissoring at 1886 cm"1; C=O stretching at 1690 cm"1; C=C and C=N ring stretching at 1418 cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 162.47 degrees C (endotherm); m.p. = 155-160 degrees C (MEL-TEMP); (flurbiprofen m.p. = 110-111 degrees C, 4,4' -bipyridine m.p. = 111-114 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 30.93% weight loss starting at 31.13 degrees C and a 46.26% weight loss starting at 168.74 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30kV, 15mA), the powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 20 and a scan speed of 2.0 minute. PXRD derived from the single crystal data: experimental (calculated): 16.8 (16.8); 17.1 (17.5); 18.1 (18.4); 19.0 (19.0); 20.0 (20.4); 21.3 (21.7); 22.7 (23.0); 25.0 (25.6); 26.0 (26.1); 26.0 (26.6); 26.1 (27.5); 28.2 (28.7); 29.1 (29.7).
Example 22
Flurbiprofen:trans-l,2-bis (4-pyridyl) ethylene (2:1 stoichiometry)
25 mg (0.1023 mmol) flurbiprofen and 10 mg (0.0548 mmol) trans- 1, 2-bis (4- pyridyl) ethylene were dissolved in 3 mL acetone. Slow evaporation of the solvent yielded colorless needles of a 2:1 flurbiprofen: 1,2-bis (4-pyridyl) ethylene co-crystal, as shown in Figs. 43 A-B.
Crystal data: (Bruker SMART- APEX CCD Diffractometer), C42H36F2N2O4, M = 670.73, monoclinic _?2/»; a = 5.8697(9), b = 47.357(7), c = 6.3587(10) A, β = 109.492(3)°, U= 1666.2(4) A3, T = 200(2) K, Z = 2, μ(Mo-Kα) = 0.093 mm"1, Dc= 1.337 Mg/m3, λ = 0.71073 A, F(000) = 704,
Figure imgf000077_0001
21.69°, 6977 reflections measured, 2383 unique (Rjnt = 0.0383). Final residuals for 238 parameters were Ri = 0.0686, wR2 = 0.1395 for I>2σ(I), and Ri = 0.1403, wR2 = 0.1709 for all 2383 data.
Crystal packing: The co-crystal contains flurbiprofen: 1 ,2-bis (4-pyridyl) ethylene heterodimers, sustained by two hydrogen bonded carboxylic acid-pyridine supramolecular synthons, arranged in a herringbone motif that packs in the space group P2]/n. The heterodimer from 1,2-bis (4-pyridyl) ethylene further extends the homodimer relative to example 21 and packs to form a two-dimensional network sustained by π-π stacking and hydrophobic interactions of the bipyridine and phenyl groups of the flurbiprofen.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), aromatic C-H stretching at 2927 cm"1 and 2850 cm"1; N--H bending and scissoring at 1875 cm"1; C=O stretching at 1707 cm"1; C=C and C=N ring stretching at 1483 cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 100.01 degrees C, 125.59 degrees C and 163.54 degrees C (endotherms); m.p. = 153-158 degrees C (MEL-TEMP); (flurbiprofen m.p. = 110-111 degrees C, trans-1, 2-bis (4-pyridyl) ethylene m.p. = 150-153 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 91.79% weight loss starting at 133.18 degrees C followed by complete decomposition.
I?owder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30kV, 15mA), the powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 2Θ and a scan speed of 2.07minute. PXRD derived from the single crystal data, experimental (calculated): 3.6 (3.7); 17.3 (17.7); 18.1 (18.6); 18.4 (18.6); 19.1 (19.3); 22.3 (22.5); 23.8 (23.9); 25.9 (26.4); 28.1 (28.5). Example 23
Carbamazepine^-Phthalaldehyde (2:1 stoichiometry)
25 mg (0.1058 mmol) carbamazepine and 7 mg (0.0521 mmol) p- phthalaldehyde were dissolved in approximately 3 mL methanol. Slow evaporation of the solvent yielded colorless needles of a 2:1 carbamazepine : -phthalaldehyde co-crystal, as shown in Figs. 44A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C38H30N4O4, M = 606.66, monoclinic C2/c; a = 29.191(16), b = 4.962(3), c = 20.316(11) A, β = 92.105(8)°, U= 2941(3) A3, T= 200(2) K, Z = 4, μ(Mo-Kα) = 0.090 mm"1, Dc = 1.370 Mg/m3, λ = 0.71073 A, F(000) = 1272, 2θmax = 43.66°, 3831 reflections measured, 1559 unique (Rmt = 0.0510). Final residuals for 268 parameters were R] = 0.0332, wR2 = 0.0801 for I>2σ(I), and R} = 0.0403, wR2 = 0.0831 for all 1559 data.
Crystal packing: The co-crystals contain hydrogen bonded carboxamide homodimers that crystallize in the space group C2/c. The 1° amines of the homodimer are bifurcated to the carbonyl of the/7-phthalaldehyde forming a chain with an adjacent homodimer. The chains pack in a crinkled tape motif sustained by π-π interactions between phenyl rings of the carbamazepine.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). The 1° amine unsymmetrical and symmetrical stretching was shifted down to 3418 cm"1; aliphatic aldehyde and 1° amide C=O stretching was shifted up to 1690 cm"1; N-H in-plane bending at 1669 cm'1; C-H aldehyde stretching at 2861 cm"1 and H-C=O bending at 1391 cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 128.46 degrees C (endotherm), m.p. = 121-124 degrees C (MEL-TEMP), (carbamazepine m.p. = 190.2 degrees C, /7-phthalaIdehyde m.p. = 116 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 17.66% weight loss starting at 30.33 degrees C then a 17.57% weight loss starting at 100.14 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30kV, 15mA). The powder data were collected over an angular range of 3° to 40° 2Θ in continuous scan mode using a step size of 0.02° 2Θ and a scan speed of 2.0 minute. PXRD derived from the single crystal data, experimental (calculated): 8.5 (8.7); 10.6 (10.8); 11.9 (12.1); 14.4 (14.7) 15.1 (15.2); 18.0 (18.1); 18.5 (18.2); 19.8 (18.7); 23.7 (24.0); 24.2 (24.2); 26.4 (26.7); 27.6 (27.9); 27.8 (28.2); 28.7 (29.1); 29.3
(29.6); 29.4 (29.8).
Example 24
Carbamazepine :nicotinamide (1 :1 stoichiometry)
25 mg (0.1058 mmol) carbamazepine and 12 mg (0.0982 mmol) nicotinamide were dissolved in 4 mL of DMSO, methanol or ethanol. Slow evaporation of the solvent yielded colorless needles of a 1:1 carbamazepinemicotinamide co-crystal, as shown in Fig. 45.
*
Using a separate method, 25 mg (0.1058 mmol) carbamazepine and 12 mg (0.0982mmol) nicotinamide were ground together with mortar and pestle. The solid was determined to be 1:1 carbamazepine :nicotinamide microcrystals (PXRD).
1:1 carbamazepine:nicotinamide co-crystals were also prepared via another method. A 12-block experiment was designed with 12 solvents. (A block is a receiving plate, which can be an industry standard 96 well, 384 well, or 1536 well format, or a custom format.) 1152 crystallization experiments were carried out using the CrystalMax™ platform. The co-crystal was obtained from samples containing toluene, acetone, or isopropyl acetate. The resulting co-crystal was characterized by PXRD and DSC and these data are shown in Figs. 46 and 47, respectively. The co-crystals prepared from toluene, aceone, or isopropyl acetate may contain impurities such as carbamazepine in free form due to incomplete purification.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C21H18N4O2, M = 358.39, monoclinic P2}/n; a = 5.0961(8), b = 17.595(3), c = 19.647(3) A, β = 90.917(3)°, U= 1761.5(5) A3, T = 200(2) K, Z = 4, μ(Mo-Kα) = 0.090 mm"1, Dc = 1.351 Mg/m3, λ = 0.71073 A, F(000) = 752, 2θmax = 56.60°, 10919 reflections measured, 4041 unique (Rint = 0.0514). Final residuals for 248 parameters were R3 = 0.0732, wR2 = 0.1268 for I>2σ(I), and Rj = 0.1161, wR2 = 0.1430 for all 4041 data. Crystal packing: The co-crystals contain hydrogen bonded carboxamide homodimers. The 1° amines are bifurcated to the carbonyl of the nicotinamide on each side of the dimer. The 1° amines of each nicotinamide are hydrogen bonded to the carbonyl of the adjoining dimer. The dimers form chains with π-π interactions from the phenyl groups of the carbamazepine.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), unsymmetrical and symmetrical stretching shifts down to 3443 cm"1 and 3388 cm" accounting for 1° amines; 1° amide C=O stretching at 1690 cm"1; N-H in-plane bending at 1614 cm"1; C=C stretching shifted down to 1579 cm"1; aromatic H's from 800 cm" to 500 cm" are present.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 74.49 degrees C (endotherm) and 159.05 degrees C (endotherm), m.p. = 153-158 degrees C (MEL- TEMP), (carbamazepine m.p. = 190.2 degrees C, nicotinamide m.p. = 150-160 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 57.94% weight loss starting at 205.43 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30k V, 15mA). The powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 20 and a scan speed of 2.07minute. PXRD: Showed analogous peaks to the simulated PXRD derived from the single crystal data. PXRD analysis experimental (calculated): 6.5 (6.7); 8.8 (9.0); 10.1 (10.3); 13.2 (13.5); 15.6 (15.8); 17.7 (17.9); 17.8 (18.1); 18.3 (18.6); 19.8 (20.1); 20.4 (20.7); 21.6 (N/A); 22.6 (22.8); 22.9 (23.2); 26.4 (26.7); 26.7 (27.0); 28.0 (28.4).
Example 25
Carbamazepine: saccharin (1 :1 stoichiometry)
25 mg (0.1058mmol) carbamazepine and 19 mg (0.1037 mmol) saccharin were dissolved in approximately 4 mL ethanol. Slow evaporation of the solvent yielded colorless needles of a 1:1 carbamazepine: saccharin co-crystal, as shown in Fig. 48. Solubility measurements indicate that this co-crystal of carbamazepine has improved solubility over previously known forms of carbamazepine (e.g., increased molar solubility and longer solubility in aqueous solutions).
1:1 carbamazepine: saccharin co-crystals were also prepared via another method. A 12-block experiment was designed with 12 solvents. (A block is a receiving plate, which can be an industry standard 96 well, 384 well, or 1536 well format, or a custom format.) 1152 crystallization experiments were carried out using the CrystalMax™ platform. The carbamazepine: saccharin co-crystal was obtained from a mixture of isopropyl acetate and heptane. The resulting co-crystal was characterized by PXRD and DSC and these data are shown in Figures 49 and 50, respectively. The co-crystal prepared from a mixture of isopropyl acetate and heptane may contain impurities such as carbamazepine in free form due to incomplete purification.
Crystal data: (Bruker SMART-APEX CCD Diffractometer), C22H17N3O4S, M = 419.45, triclinic P-1; a = 7.5140(11), b = 10.4538(15), c = 12.6826(18) A, α = 83.642(2)°, β = 85.697(2) °, γ = 75.411(2) °, U= 957.0(2) A3, T = 200(2) K, Z = 2, μ(Mb-Kα) = 0.206 mm"1, Dc = 1.456 Mg/m3, λ = 0.71073 A, F(000) = 436, 2θmax = 56.20°; 8426 reflections measured, 4372 unique (Rj„t = 0.0305). Final residuals for 283 parameters were Ri = 0.0458, wR2 = 0.1142 for I>2σ(I), and Ri = 0.0562, wR2 = 0.1204 for all 4372 data.
Crystal packing: The co-crystals contain hydrogen bonded carboxamide homodimers. The 2° amines of the saccharin are hydrogen bonded to the carbonyl of the carbamazepine on each side forming a tetramer. The crystal has a space group of P-1 with π-π interactions between the phenyl groups of the carbamazepine and the saccharin phenyl groups.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR), unsymmetrical and symmetrical stretching shifts up to 3495 cm"1 accounting for 1° amines; C=O aliphatic stretching was shifted up to 1726 cm"1; N-H in-plane bending at 1649 cm"1; C=C stretching shifted down to 1561 cm"1; (O=S=O) sulfonyl peak at 1330 cm"1 C-N aliphatic stretching 1175 cm"1.
Differential Scanning Calorimetry: (TA Instruments 2920 DSC), 75.31 degrees C (endotherm) and 177.32 degrees C (endotherm), m.p. = 148-155 degrees C (MEL- TEMP); (carbamazepine m.p. = 190.2 degrees C, saccharin m.p. = 228.8 degrees C). Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA), 3.342% weight loss starting at 67.03 degrees C and a 55.09% weight loss starting at 118.71 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using Cu Kα (λ = 1.540562), 30kV, 15mA). The powder data were collected over an angular range of 3° to 40° 20 in continuous scan mode using a step size of 0.02° 20 and a scan speed of 2.0 7minute. PXRD derived from the single crystal data, experimental (calculated): 6.9 (7.0); 12.2 (12.2); 13.6 (13.8); 14.0 (14.1); 14.1 (14.4); 15.3 (15.6); 15.9 (15.9); 18.1 (18.2); 18.7 (18.8); 20.2 (20.3); 21.3 (21.5); 23.7 (23.9); 26.3 (26.4); 28.3 (28.3).
Example 26
Carbamazepine:2,6-pyridinedicarboxylic acid (1:1 stoichiometry)
36 mg (0.1524 mmol) carbamazepine and 26 mg (0.1556 mmol) 2,6- pyridinedicarboxylic acid were dissolved in approximately 2 mL ethanol. Slow evaporation of the solvent yielded clear needles of a 1:1 carbamazepine:2,6- pyridinedicarboxylic acid co-crystal, as shown in Figs. 51 A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). C227N3O5, M=403.39, orthorhombic P2(l)2(l)2(l); a=7.2122, b=14.6491, c=17.5864 A,α=90°, β=90°, γ=90°, U=l 858.0(2) A3, T=100 K, Z=4, μ(MO-Kα)=0.104 mm"1, Dc=1.442 Mg/m3, λ=0.71073A , F(000)840, 2θmax=28.3. 16641 reflections measured, 4466 unique (Rint=0.093). Final residuals for 271 parameters were Rι=0.0425 and wR2=0.0944 for I>2σ(I).
Crystal packing: Each hydrogen on the carbamazepine 1° amine is hydrogen bonded to a carbonyl group of a different 2,6-pyridinedicarboxylic acid moiety. The carbonyl of the carbamazepine carboxamide is hydrogen bonded to two hydroxide groups of one 2,6-pyridinedicarboxylic acid moiety.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). 3439 cm"1, (N-H stretch, 1° amine, carbamazepine); 1734 cm"1, (C=O); 1649 cm"1 , (C=C).
Melting Point: 214-216 degrees C (MEL-TEMP), (carbamazepine m.p. = 191-192 degrees C, 2,6-pyridinedicarboxylic acid m.p. = 248-250 degrees C). Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA). 69% weight loss starting at 215 degrees C and a 17% weight loss starting at 392 degrees C followed by complete decomposition.
Example 27
Carbamazepine: 5 -nitroisophthalic acid (1 :1 stoichiometry)
40 mg (0.1693 mmol) carbamazepine and 30 mg (0.1421 mmol) 5- nitroisophthalic acid were dissolved in approximately 3 mL methanol or ethanol. Slow evaporation of the solvent yielded yellow needles of a 1:1 carbamazepine:5- nitroisophthalic acid co-crystal, as shown in Figs. 52A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). monoclinic C2/c; a=34.355(8), b=5.3795(13), c=23.654(6) A,α=90°, β=93.952(6)°, γ=90°, U=4361.2(18)A3, T=200(2) K, Z=4, μ(MO-K )=0.110 mm"1, Dc=1.439 Mg/m3, λ=0.71073A , F(000)1968, 11581 reflections measured, 4459 unique (Rint=0.0611). Final residuals for 311 parameters were Rι=0.0725, wR2=0.1801 for I>2σ(I), and Rι=0.1441, wR2=0.1204 for all 4459 data.
Crystal packing: The co-crystals are sustained by hydrogen bonded carboxylic acid homodimers between the two 5-nitroisophthalic acid moieties and hydrogen bonded carboxy-amide heterodimers between the carbamazepine and 5-nitroisophthalic acid moiety. There is solvent hydrogen bonded to an additional N-H donor from the carbamazepine moiety.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). 3470 cm"1, (N-H stretch, 1° amine, carbamazepine); 3178 cm"1, (C-H stretch, alkene); 1688 cm"1, (C=O); 1602 cm"1, (C=C).
Differential Scanning Calorimetry: (TA Instruments 2920 DSC). 190.51 degrees C (endotherm). m.p. = NA (decomposes at 197-200 degrees C) (MEL-TEMP), (carbamazepine m.p. = 191-192 degrees C, 5-nitroisophthalic acid m.p. = 260-261 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA). 32.02% weight loss starting at 202 degrees C, a 12.12% weight loss starting at 224 degrees C and a 17.94% weight loss starting at 285 degrees C followed by complete decomposition.
Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using CuKα (λ=l.540562), 30kV, 15mA). The powder data were collected over an angular range of 3 to 40 2 in continuous scan mode using a step size of 0.02 2 and a scan speed of 2.0 /min. PXRD: Showed analogous peaks to the simulated PXRD derived from the single crystal data. PXRD analysis experimental (calculated): 10.138 (10.283), 15.291 (15.607), 17.438 (17.791), 21.166 (21.685), 31.407 (31.738), 32.650 (32.729).
Example 28
Carbamazepine: 1,3,5,7-adamantane tetracarboxylic acid (2:1 stoichiometry)
15 mg (0.1524 mmol) carbamazepine and 20 mg (0.1556 mmol) 1,3,5,7- adamantanetetracarboxylic acid were dissolved in approximately 1 mL methanol or 1 mL ethanol. Slow evaporation of the solvent yields clear plates of a 2: 1 carbamazepine: 1,3,5,7-adamantanetetracarboxylic acid co-crystal, as shown in Figs. 53 A- B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). C44H40N40, M=784.80, monoclinicC2/c; a=18.388(4), b=12.682(3), c=16.429(3) A, β=100.491(6)°, U=3767.1(14) A3, T=100(2) K, Z=4, μ(MO-Kα)=0.099 mm"1, Dc=1.384 Mg/m3, λ=0.71073A , F(000)1648, 2θmax=28.20°. 16499 reflections measured, 4481 unique (Ri„t=0.052). Final residuals for 263 parameters were Rι=0.0433 and wR2=0.0913 for I>2σ(I).
Crystal packing: The co-crystals form a single 3D network of four tetrahedron, linked by square planes similar to the PtS topology. The crystals are sustained by hydrogen bonding.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). 3431 cm"1, (N-H stretch, 1° amine, carbamazepine); 3123 cm"1, (C-H stretch, alkene); 1723 cm"1, (C=O); 1649 cm"1, (C=C).
Melting Point: (MEL-TEMP). 258-260 degrees C (carbamazepine m.p. = 191-192 degrees C, adamantanetetracarboxylic acid m.p. = >390 degrees C). Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA). 9% weight loss starting at 189 degrees C, a 52% weight loss starting at 251 degrees C and a 31% weight loss starting at 374 degrees C followed by complete decomposition.
Example 29
Carbamazepine:benzoquinone (1:1 stoichiometry)
25 mg (0.1058 mmol) carbamazepine and 11 mg (0.1018 mmol) benzoquinone was dissolved in 2 mL methanol or THF. Slow evaporation of the solvent produced an average yield of yellow crystals of a 1:1 carbamazepine:benzoquinone co-crystal, as shown in Figs. 54A-B.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). C2ιHι6N2O3, M=344.36, monoclinic P2(l)/c; a=10.3335(18), b=27.611(5), c=4.9960(9) A, β=l 02.275(3)°, U=1392.9(4) A3, T=100(2) K, Z=3, Dc=1.232 Mg/m3, μ(MO-Kα)=0.084 mm"1, λ=0.71073A, F(000)540, 2θmax=28.24°. 8392 reflections measured, 3223 unique (Ri„t=0.1136). Final residuals for 199 parameters were Rι=0.0545 and wR2=0.1358 for I>2σ(I), and R^O.0659 and wR2=0.1427 for all 3223 data.
Crystal packing: The co-crystals contain hydrogen bonded carboxamide homodimers. Each 1° amine on the carbamazepine is bifurcated to a carbonyl group of a benzoquinone moiety. The dimers form infinite chains.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). 3420 cm"1, (N-H stretch, 1° amine, carbamazepine); 2750 cm"1, (aldehyde stretch); 1672 cm"1, (C=O); 1637 cm"1, (C=C, carbamazepine).
Melting Point: 170 degrees C (MEL-TEMP), (carbamazepine m.p. = 191-192 degrees C, benzoquinone m.p. = 115.7 degrees C).
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA). 20.62% weight loss starting at 168 degrees C and a 78% weight loss starting at 223 degrees C followed by complete decomposition. Example 30
Carbamazepine:trimesic acid (1:1 stoichiometry)
36 mg (0.1524 mmol) carbamazepine and 31 mg (0.1475 mmol) trimesic acid were dissolved in a solvent mixture of approximately 2 mL methanol and 2 mL dichloromethane. Slow evaporation of the solvent mixture yielded white starbursts of a 1:1 carbamazepine:trimesic acid co-crystal, as shown in Figs. 55A-B.
1:1 carbamazepine :trimesic acid co-crystals were also prepared via another method. A 9-block experiment was designed with 10 solvents. 864 crystallization experiments with 8 co-crystal formers and 3 concentrations were carried out using the CrystalMax™ platform. The co-crystal was obtained from samples containing methanol. The resulting co-crystal was characterized by PXRD and the diffractogram is shown in Fig. 56.
Crystal data: (Bruker SMART-APEX CCD Diffractometer). C24H18N2O7, M=446.26, monoclinic C2/c; a=32.5312(50), b=5.2697(8), c=24.1594(37) A,α=90°, β=98.191(3)°, γ=90°, U=4099.39(37) A3, T=-173 K, Z=8, μ(MO-Kα)=0.110 mm"1, Dc=1.439 Mg/m3, λ=0.71073A, F(000)1968, 2θmax=26.43°. 11581 reflections measured, 4459 unique
Figure imgf000086_0001
wR2=0.1887 for I>2σ(I), and Rf=0.1441, wR =0.1204 for all 3601 data.
Crystal packing: The co-crystals are sustained by hydrogen bonded carboxylic acid homodimers between carbamazepine and trimesic acid moieties and hydrogen bonded carboxylic acid-amine heterodimers between two trimesic acid moieties arranged in a stacked ladder formation.
Infrared Spectroscopy: (Nicolet Avatar 320 FTIR). 3486 cm"1 (N-H stretch, 1° amine, carbamazepine); 1688 cm"1 (C=O, 1° amide stretch, carbamazepine); 1602 cm"1 (C=C, carbamazepine).
Differential Scanning Calorimetry: (TA Instruments 2920 DSC). 273 degrees C (endotherm). m.p. = NA, decomposes at 278 degrees C (MEL-TEMP), (carbamazepine m.p. = 191-192 degrees C, trimesic acid m.p. = 380 degrees C)
Thermogravimetric Analysis: (TA Instruments 2950 Hi-Resolution TGA). 62.83%o weight loss starting at 253 degrees C and a 30.20% weight loss starting at 278 degrees C followed by complete decomposition. Powder x-ray diffraction: (Rigaku Miniflex Diffractometer using CuKα (λ=l .540562), 30kV, 15mA). The powder data were collected over an angular range of 3 to 40 degrees 2-theta in continuous scan mode using a step size of 0.02 degrees 2-theta and a scan speed of 2.0/min. PXRD analysis experimental: 10.736, 12.087, 16.857, 24.857, 27.857.
Table XXIV. Detailed Characterisation of Co-Crystals
All PXRD peaks are in units of degrees 2-theta
All Raman shifts are in units of cm"1
Celecoxib :Nicotinamide (Example 1)
PXRD: 3.77, 7.56, 9.63, 14.76, 15.21, 16.01, 17.78, 18.68, 19.31, 20.44, 21.19, 22.10
DSC: Two endothermic transitions at about 117 and 119 degrees C and a sharp endotherm at about 130 degrees C
TGA: Decomposition beginning at about 150 degrees C
Raman: 1618, 1599, 1452, 1370, 1163, 1044, 973, 796, 632, 393, 206
Celecoxib: 18-Crown-6 (Example 2)
PXRD: 8.73, 11.89, 12.57, 13.13, 15.01, 16.37, 17.03, 17.75, 18.45, 20.75, 22.37, 23.11,
24.33, 24.97, 26.61, 28.15
DSC: Sharp endotherm at about 190 degrees C
TGA: Decomposition above 200 degrees C with a 25% weight loss between about 190-
210 degrees C
Topiramate: 18-Crown-6 (Example 3)
PXRD: 10.79, 11.07, 12.17, 13.83, 16.13, 18.03, 18.51, 18.79, 19.21, 21.43, 22.25, 24.11
DSC: Sharp endotherm at about 135 degrees C
TGA: Rapid decomposition beginning at about 135 degrees C and leveling off slightly after 200 degrees C
Raman: 2995, 2943, 1472, 1427, 1262, 849, 805, 745, 629, 280, 226
Olanzapine :Nicotinamide (Example 4)
PXRD (Form I): 4.89, 8.65, 12.51, 14.19, 15.59, 17.15, 19.71, 21.05, 23.95, 24.59, 25.53,
26.71
PXRD (Form II): 5.13, 8.65, 11.87, 14.53, 17.53, 18.09, 19.69, 24.19, 26.01 (data as received)
PXRD (Form III): 6.41, 12.85, 14.91, 18.67, 21.85, 24.37
DSC (Form I): Slightly broad endotherm at about 126 degrees C c/s-Itraconazole:Succinic Acid (Example 5)
PXRD: 3.01, 6.01, 8.13, 9.05, 15.87, 16.17, 17.29, 24.47
DSC: Single endothermic transition at about 160 degrees C ± 1.0 degrees C
TGA: Less than 0.1 % volatile components by weight
Figure imgf000088_0001
Phenytoin:Pyridone (Example 18)
PXRD: 5.2, 11.1, 15.1, 16.2, 16.7, 17.8, 19.4, 19.8, 20.3, 21.2, 23.3, 26.1, 26.4, 27.3, 29.5
DSC: Endothermic transitions at about 233 and 271 degrees C
TGA: 29.09 percent weight loss starting at about 193 degrees C, 48.72 percent weight loss starting at about 238 degrees C, 18.38 percent weight loss starting at about 260 degrees C
Aspirin: , '-Bipyridine (Example 19)
DSC: Endothermic transition at about 95 degrees C
TGA: 9 percent weight loss starting at about 23 degrees C, 49.06 percent weight loss starting at about 103 degrees C, decomposition starting at about 209 degrees C
Ibuprofen:4,45 -Bipyridine (Example 20) PXRD: 3.4, 6.9, 10.4, 17.3, 19.1
DSC Endothermic transitions at about 65 and 119 degrees C TGA 13.28 percent weight loss between room temperature and about 100 degrees C
Flurbiprofen:4,4' -Bipyridine (Example 21)
PXRD: 16.8, 17.1, 18.1, 19.0, 20.0, 21.3, 22.7, 25.0, 26.0, 26.1, 28.2, 29.1
DSC: Endothermic transition at about 162 degrees C
TGA: 30.93 percent weight loss starting at about 31 degrees C, 46.26 percent weight loss starting at about 169 degrees C
Flurbiprofen:trans- 1,2-bis (4-pyridyl) ethylene (Example 22)
PXRD: 3.6, 17.3, 18.1, 18.4, 19.1, 22.3, 23.8, 25.9, 28.1
DSC: Endothermic transitions at about 100, 126, and 164 degrees C
TGA: 91.79 percent weight loss starting at about 133 degrees C
Carbamazepine :p-phthalaldehyde (Example 23)
PXRD: 8.5, 10.6, 11.9, 14.4, 15.1, 18.0, 18.5, 19.8, 23.7, 24.2, 26.4, 27.6, 27.8, 28.7,
29.3, 29.4
DSC: Endothermic transition at about 128 degrees C
TGA: 17.66 percent weight loss starting at about 30 degrees C, 17.57 percent weight loss starting at about 100 degrees C
Carbamazepine :Nicotinamide (Example 24)
PXRD: 6.5, 8.8, 10.1, 13.2, 15.6, 17.7, 17.8, 18.3, 19.8, 20.4, 21.6, 22.6, 22.9, 26.4, 26.7,
28.0
DSC: Sharp endotherm at about 157 degrees C
TGA: Decomposition beginning at about 150 degrees C
Carbamazepine: Saccharin (Example 25)
PXRD: 6.9, 12.2, 13.6, 14.0, 14.1, 15.3, 15.9, 18.1, 18.7, 20.2, 21.3, 23.7, 26.3, 28.3
DSC: Endotherms were present at about 75 and 177 degrees C
TGA: 3.342 percent weight loss starting at about 67 degrees C, 55.09 percent weight loss starting at about 119 degrees C
Carbamazepine:2,6-pyridinecarboxylic acid (Example 26)
TGA: 69 percent weight loss starting at about 215 degrees C, 17 percent weight loss starting at about 392 degrees C Carbamazepine:5-nitroisophthalic acid (Example 27)
PXRD: 10.14, 15.29, 17.44, 21.17, 31.41, 32.65
DSC: Endotherm at about 191 degrees C
TGA: 32.02 percent weight loss starting at about 202 degrees C, 12.12 percent weight loss starting at about 224 degrees C, 17.94 percent weight loss starting at about 285 degrees C
Carbamazepine:!, 3, 5,7-adamantane tetracarboxylic acid (Example 28)
TGA: 9 percent weight loss starting at about 189 degrees C, 52 percent weight loss starting at about 251 degrees C, 31 percent weight loss starting at about 374 degrees C
Carbamazepine:Benzoquinone (Example 29)
TGA: 20.62 percent weight loss starting at about 168 degrees C, 78 percent weight loss starting at about 223 degrees C
Carbamazepine:Trimesic acid (Example 30)
PXRD: 10.89, 12.23, 14.83, 16.25, 17.05, 18.13, 18.47, 21.47, 21.95, 24.57, 25.11, 27.99
DSC: Endothermic transition at about 273 degrees C
TGA: 62.83 percent weight loss starting at about 253 degrees C, 30.20 percent weight loss starting at about 278 degrees C
Example 31
A co-crystal with a modulated dissolution profile has been prepared. Celecoxib: nicotinamide co-crystals were prepared via methods shown in Example 1. (See Fig. 57)
Example 32
A co-crystal with a modulated dissolution profile has been prepared, cis- Itraconazole: succinic acid, cw-itraconazolei-tartaric acid and czAitraconazole:L-malic acid co-crystals were prepared via methods shown in Examples 5, 7 and 8. (See Fig. 58)
Example 33
A co-crystal of an unsaltable or difficult to salt API has been prepared. Celecoxib: nicotinamide co-crystals were prepared via methods shown in Example 1. Example 34
A co-crystal with an improved hygroscopicity profile has been prepared. Celecoxib: nicotinamide co-crystals were prepared via methods shown in Example 1. (See Fig. 59)
Example 35
A co-crystal with reduced form diversity as compared to the API has been prepared. Co-crystals of carbamazepine and saccharin have been prepared via method shown in Example 25.
Example 36
The formulation of a modafinil :malonic acid form I co-crystal was completed using lactose. Two mixtures, one of modafinil and lactose, and the second of modafinil :malonic acid co-crystal and lactose, were ground together in a mortar an pestle. The mixtures targeted a 1 :1 weight ratio of modafinil to lactose. In the modafinil and lactose mixture, 901.2 mg of modafinil and 901.6 mg of lactose were ground together. In the modafinil malonic acid co-crystal and lactose mixture, 1221.6 mg of co-crystal and 871.4 mg of lactose were ground together. The resulting powders were analyzed by PXRD and DSC. The PXRD patterns and DSC thermograms of the mixtures showed virtually no change upon comparison with both individual components. The DSC of the co-crystal mixture showed only the co-crystal melting peak at 113.6 degrees C with a heat of fusion of 75.9 J/g. This heat of fusion is 59.5 % of that found for the co-crystal alone (127.5 J/g). This result is consistent with a 58.4 % weight ratio of co-crystal in the mixture. The DSC of the modafinil and lactose mixture had a melting point of 165.7 degrees C. This is slightly lower then the measured melting point of modafinil (168.7 degrees C). The heat of fusion of the mixture (59.3 J/g) is 46.9 % that of the modafinil alone (126.6 J/g), which is consistent with the estimated value of 50 %.
The in vitro dissolution of both the modafinil malonic acid form I co-crystal and pure modafinil were tested in capsules. Both gelatin and hydroxypropylmethyl cellulose (HPMC) capsules were used in the dissolution study. The capsules were formulated with and without lactose. All formulations were ground in a mortar and pestle prior to transfer into a capsule. The dissolution of the capsules was tested in 0.01 M HCl (See Figure 61).
In 0.01M HCl, using sieved and ground materials in gelatin capsules:
Modafinil and the modafinikmalonic acid form I co-crystal were passed through a 38 micrometer sieve. Gelatin capsules (Size 0, B&B Pharmaceuticals, Lot # 15-01202) were filled with 200.0 mg sieved modafinil, 280.4 mg sieved modafinil :malonic acid co- crystal, 200.2 mg ground modafinil, or 280.3 mg ground modafinikmalonic acid co- crystal. Dissolution studies were performed in a Vankel VK 7000 Benchsaver Dissolution Testing Apparatus with the VK750D heater/circulator set at 37 degrees C. At 0 minutes, the capsules were dropped into vessels containing 900 mL 0.01 M HCl and stirred by paddles.
Absorbance readings were taken using a Cary 50 Spectrophotometer (wavelength set at 260nm) at the following time points: 0, 5, 10, 15, 20, 25, 30, 40, 50, and 60 minutes. The absorbance values were compared to those of standards and the modafinil concentrations of the solutions were calculated.
In 0.01M HCl, using ground materials in gelatin or HPMC capsules, with and without lactose:
Modafinil and the modafinil :malonic acid form I co-crystal were mixed with equivalent amounts of lactose (Spectrum, Lot QV0460) for approximately 5 minutes. Gelatin capsules (Size 0, B&B Pharmaceuticals, Lot # 15-01202) were filled with 400.2 mg modafinil and lactose (approximately 200 mg modafinil), or 561.0 mg modafinil :malonic acid form I co-crystal and lactose (approximately 200 mg modafinil). HPMC capsules (Size 0, Shionogi, Lot # A312A6) were filled with 399.9 mg modafinil and lactose, 560.9 mg modafinikmalonic acid co-crystal and lactose, 199.9 mg modafinil, or 280.5 mg modafinil :malonic acid form I co-crystal. The dissolution study was carried out as described above. Example 37
The modafinikmalonic acid form I co-crystal (from Example 10) was administered to dogs in a pharmacokinetic study. Particles of modafinil :malonic acid co- crystal with a median particle size of about 16 micrometers were administered in the study. As a reference, micronized modafinil with a median particle size of about 2 micrometers was also administered in the study. The AUC of the modafinil :malonic acid co-crystal was determined to be 40 to 60 percent higher than that of the pure modafinil. Such a higher bioavailability illustrates the modulation of an important pharmacokinetic parameter due to an embodiment of the present invention. A compilation of important pharmacokinetic parameters measured during the animal study are included in Table XXV. Table XXV- Pharmacokinetic arameters of modafinihmalonic acid co-cr stal and ure modafinil in do s
Figure imgf000093_0001
The increased half-life and bioavailability of modafinil in the malonic acid form I co-crystal may be due to the presence of malonic acid. It is believed that the malonic acid may be inhibiting one or more pathways responsible for the metabolism or elimination of modafinil. It is noted that modafinil and malonic acid share a similar structure: each including two carbonyl or sulfonyl groups separated by a -CH2- and each molecule is terminated with a group that is capable of participation in a hydrogen bond with an enzyme. Such a mechanism may take place with other APIs or co-crystal formers of similar structure.
Example 38
The stability of the modafinikmalonic acid form I co-crystal was measured at various temperatures and relative humidities over a four week period. No degradation was found to occur at 20 or 0 degrees C. At 60 degrees C, about 0.14 percent degradation per day was determined based on a simple exponential model. At 80 degrees C, about 8 percent degradation per day was determined. TABLE I
Figure imgf000094_0001
Figure imgf000094_0002
TABLE I
Figure imgf000095_0001
TABLE I
Figure imgf000096_0001
TABLE I
Figure imgf000097_0001
TABLE I
Figure imgf000098_0001
TABLE I
Figure imgf000099_0001
TABLE I
Figure imgf000100_0001
TABLE I
Figure imgf000101_0001
TABLE I
Figure imgf000102_0001
Figure imgf000102_0002
TABLE I
Figure imgf000103_0001
TABLE I
Figure imgf000104_0001
TABLE I
Figure imgf000105_0001
TABLE I
Figure imgf000106_0001
TABLE I
Figure imgf000107_0001
TABLE I
Figure imgf000108_0001
Figure imgf000108_0003
Figure imgf000108_0002
TABLE II
Figure imgf000109_0001
© oe
Figure imgf000109_0002
TABLE
Figure imgf000110_0001
Figure imgf000110_0002
TABLE II
Figure imgf000111_0001
Figure imgf000111_0002
TABLE
Figure imgf000112_0001
Figure imgf000112_0002
TABLE
Figure imgf000113_0001
Figure imgf000113_0002
TABLE II
Figure imgf000114_0001
Figure imgf000114_0002
TABLfc II
Figure imgf000115_0001
Figure imgf000115_0002
TABLE II
Figure imgf000116_0001
Figure imgf000116_0002
TABLE II
Figure imgf000117_0001
Figure imgf000117_0002
TABLE
Figure imgf000118_0001
Figure imgf000118_0002
TABLE
Figure imgf000119_0001
Figure imgf000119_0002
TABLE
Figure imgf000120_0001
Figure imgf000120_0002
TABLE II
Figure imgf000121_0001
Figure imgf000121_0002
TABLE II
Figure imgf000122_0001
Figure imgf000122_0002
TABLE II
Figure imgf000123_0001
Figure imgf000123_0002
TABLE II
Figure imgf000124_0001
TABLE II
Figure imgf000125_0001
TABLE
Figure imgf000126_0001
TABLE
Figure imgf000127_0001
TABLE II
Figure imgf000128_0001
TABLE II
Figure imgf000129_0001
TABLE II
Figure imgf000130_0001
TABLE II
©
Figure imgf000131_0001
TABLE
Figure imgf000132_0001
TABLE II
Figure imgf000133_0001
TABLE II
Uι Ui
TABLE
Figure imgf000135_0001
TABLE II
Figure imgf000136_0001
TABLE II
ON
Figure imgf000137_0001
TABLE II
Uι -4
Figure imgf000138_0001
TABLE II
00
Figure imgf000139_0002
Figure imgf000139_0001
TABLE
Uι 0
Figure imgf000140_0001
TABLE II
Figure imgf000141_0001
TABLE II
Figure imgf000142_0001
TABLE
4- l»
Figure imgf000143_0001
TABLE II
Figure imgf000144_0001
TABLE
Figure imgf000145_0001
TABLE
Figure imgf000146_0001
TABLE III
Figure imgf000147_0001
TABLE
Figure imgf000148_0001
I ABLE III
Figure imgf000149_0001
lAtSLt III
Figure imgf000150_0001
TABLE
Figure imgf000151_0001
Figure imgf000151_0002
Figure imgf000151_0003
TABLE
Figure imgf000152_0001
TABLE
Figure imgf000153_0001
TABLE
<
Figure imgf000154_0001
TABLE III
Figure imgf000155_0001
TABLE III
CΛ CΛ
Figure imgf000156_0001
Figure imgf000156_0002
TABLE
Figure imgf000157_0001
TABLE III
CΛ -4
Figure imgf000158_0001
Figure imgf000158_0002
TABLE III
00
Figure imgf000159_0001
Figure imgf000159_0002
TABLE
Figure imgf000160_0001
TABLE III
ON ©
Figure imgf000161_0001
TABLE III
Figure imgf000162_0001
TABLE III
ON
Figure imgf000163_0001
TABLE
Figure imgf000164_0001
TABLE III
ON 4-
Figure imgf000165_0001
TABLE III
ON CΛ
Figure imgf000166_0001
TABLE III
ON ON
Figure imgf000167_0001
TABLE III
ON -4
Figure imgf000168_0001
TABLE
Figure imgf000169_0001
TABLE III
Figure imgf000170_0001
TABLE III
-4
©
Figure imgf000171_0001
TABLE
Figure imgf000172_0001
TABLE III
-4
Figure imgf000173_0001
TABLE III
-4
UI
Figure imgf000174_0001
Table IV
Figure imgf000175_0001
Table IV
Figure imgf000176_0001
Table IV
Figure imgf000177_0001
Table IV
-4 -4
Figure imgf000178_0001
Table IV
Figure imgf000179_0001
Table IV
Figure imgf000180_0001
Table IV
Figure imgf000181_0001
Table IV
Figure imgf000182_0001
Table IV
Figure imgf000183_0001
Table IV
Figure imgf000184_0001
Table IV
Figure imgf000185_0001
Table IV
Figure imgf000186_0001
Table IV
Figure imgf000187_0001
Table IV
oo
-4
Figure imgf000188_0001
Table IV
o o
Figure imgf000189_0001
Table IV
Figure imgf000190_0001
Table IV
Figure imgf000191_0001
Table IV
Figure imgf000192_0001
Table IV
Figure imgf000193_0001
Table IV
Figure imgf000194_0001
Table IV
Figure imgf000195_0001
Table IV
Figure imgf000196_0001
Table IV
ON
Figure imgf000197_0001
Table IV
-4
Figure imgf000198_0001
Table IV
Figure imgf000199_0001
Table IV
Figure imgf000200_0001
Table IV
>
© ©
Figure imgf000201_0001
Table IV
>
©
Figure imgf000202_0001
Table IV
>
© >
Figure imgf000203_0001
Table IV
>
©
UI
Figure imgf000204_0001
Table IV
>
©
Figure imgf000205_0001
i ame IV
>
©
Figure imgf000206_0001
Table IV
Figure imgf000207_0001
Table IV
»
©
-4
Figure imgf000208_0001
Table IV
»
©
00
Figure imgf000209_0001
Table IV
»
©
Figure imgf000210_0001
Figure imgf000210_0002
Table IV
Figure imgf000211_0001
Table IV
Figure imgf000212_0001
Table IV
Figure imgf000213_0001
Table IV
Figure imgf000214_0001
Table IV
Figure imgf000215_0001
Table IV
Figure imgf000216_0001
Table IV
Figure imgf000217_0001
Table IV
>
-4
Figure imgf000218_0001
Table IV
Figure imgf000219_0001
Table IV
Figure imgf000220_0001
Table IV
» »
©
Figure imgf000221_0001
Table IV
» »
Figure imgf000222_0001
Table IV
» » »
Figure imgf000223_0001
Table IV
Figure imgf000224_0001
Table IV
) )
4-
Figure imgf000225_0001
Table IV
» »
Figure imgf000226_0001
Table IV
» »
Figure imgf000227_0001
Table IV
» »
-4
Figure imgf000228_0001
Table IV
» »
Figure imgf000229_0001
Table IV
Figure imgf000230_0001
Table IV
»
UI
©
Figure imgf000231_0001
Table IV
Figure imgf000232_0001
Table IV
Figure imgf000233_0001
Table IV
Figure imgf000234_0001
Table IV
» I
Figure imgf000235_0001
Table IV
Figure imgf000236_0001
Table IV
Figure imgf000237_0001
Table IV
»
UI -4
Figure imgf000238_0001
Table IV
» I
Figure imgf000239_0001
Table IV
Figure imgf000240_0001
Table IV
4-
©
Figure imgf000241_0001
Table IV
» -
Figure imgf000242_0001
Table IV
Figure imgf000243_0001
Table IV
» - i
Figure imgf000244_0001
Table IV
Figure imgf000245_0001
Table IV
»
4- CΛ
Figure imgf000246_0001
Table IV
Figure imgf000247_0001
Table IV
»
4- -4
Figure imgf000248_0001
Table IV
» - 0
Figure imgf000249_0001
Table IV
» -
Figure imgf000250_0001
Table IV
Figure imgf000251_0001
Table IV
Figure imgf000252_0001
Table IV
Figure imgf000253_0001
Table IV
Figure imgf000254_0001
Table IV
Figure imgf000255_0001
Table IV
Figure imgf000256_0001
Table IV
Figure imgf000257_0001
Table IV
Figure imgf000258_0001
Table IV
Figure imgf000259_0001
Table IV
Figure imgf000260_0001
Table IV
Figure imgf000261_0001
Table IV
Figure imgf000262_0001
Table IV
» »
Figure imgf000263_0001
Table IV
Figure imgf000264_0001
Table IV
Figure imgf000265_0001
Table IV
Figure imgf000266_0001
Table IV
Figure imgf000267_0001
Table IV
Figure imgf000268_0001
Table IV
Figure imgf000269_0001
Table IV
Figure imgf000270_0001
Table IV
»
-4
©
Figure imgf000271_0001
Table IV
»
-4
Figure imgf000272_0001
Table IV
»
-4 »
Figure imgf000273_0001
Table IV
»
-4
Ul
Figure imgf000274_0001
Table IV
»
-4
Figure imgf000275_0001
Table IV
Figure imgf000276_0001
Table IV
Figure imgf000277_0001
Table IV
Ki -4 -4
Figure imgf000278_0001
Table IV
»
-4
00
Figure imgf000279_0001
Table IV
Figure imgf000280_0001
Table IV
»
00
©
Figure imgf000281_0001
Table IV
Figure imgf000282_0001
Table IV
» »
Figure imgf000283_0001
Table IV
»
90 i
Figure imgf000284_0001
Table IV
Figure imgf000285_0001
Table IV
Figure imgf000286_0001
Table IV
Figure imgf000287_0001
Table IV
»
00 -4
Figure imgf000288_0001
Table IV
Figure imgf000289_0001
Table IV
Figure imgf000290_0001
Table IV
»
©
Figure imgf000291_0001
Table IV
Figure imgf000292_0001
Table IV
» »
Figure imgf000293_0001
Table IV
Figure imgf000294_0001
Table IV
»
Figure imgf000295_0001
Table IV
Figure imgf000296_0001
Table IV
Figure imgf000297_0001
Table IV
»
-4
Figure imgf000298_0001
Table IV
Figure imgf000299_0001
Table IV
Figure imgf000300_0001
Table IV
Ul
© ©
Figure imgf000301_0001
Table IV
Ul
©
Figure imgf000302_0001
Table IV
Ul
© »
Figure imgf000303_0001
Table IV
Ul
©
Ul
Figure imgf000304_0001
Table IV
Ul
©
Figure imgf000305_0001
Table IV
Ul
©
Figure imgf000306_0001
Table IV
Ul
©
Figure imgf000307_0001
Table IV
Ul
©
-4
Figure imgf000308_0001
Table IV
Ul
©
00
Figure imgf000309_0001
Table IV
Ul
©
Figure imgf000310_0001
Table IV
Figure imgf000311_0001
Table IV
Figure imgf000312_0001
Table IV
Figure imgf000313_0001
Table IV
Ul Ul
Figure imgf000314_0001
Table IV
Figure imgf000315_0001
Table IV
Figure imgf000316_0001
Table IV
Ul
Figure imgf000317_0001
Table IV
Ul -4
Figure imgf000318_0001
Table IV
Ul 00
Figure imgf000319_0001
Table IV
Ul
Figure imgf000320_0001
Table IV
Ul »
©
Table IV
Ul »
Figure imgf000322_0001
Table IV
Ul » »
Figure imgf000323_0001
Table IV
Ul »
Ul
Figure imgf000324_0001
Table IV
Ul »
4-
Figure imgf000325_0001
Table IV
Ul »
Figure imgf000326_0001
Table IV
Ul »
Figure imgf000327_0001
Table IV
Ul »
-4
Figure imgf000328_0001
Table IV
Ul Ki
90
Figure imgf000329_0001
Table IV
Ul »
Figure imgf000330_0001
Table IV
Ul Ul
©
Figure imgf000331_0001
Table IV
Ul Ul
Figure imgf000332_0001
Table IV
Ul Ul »
Figure imgf000333_0001
Table IV
Figure imgf000334_0001
Table IV
Ul Ul
Figure imgf000335_0001
Table IV
Figure imgf000336_0001
Table IV
Figure imgf000337_0001
Table IV
Ul Ul -4
Figure imgf000338_0001
Table IV
Ul Ul
00
Figure imgf000339_0001
Table IV
Figure imgf000340_0001
Table IV
Ul 4-
©
Figure imgf000341_0001
Table IV
Figure imgf000342_0001
Table IV
Ul 4- Ki
Figure imgf000343_0001
Table IV
Ul 4- Ui
Figure imgf000344_0001
Table IV
Figure imgf000345_0001
Table IV
Figure imgf000346_0001
Table
Figure imgf000347_0001
Table IV
Ul 4- -4
Figure imgf000348_0001
Table IV
l 4-
90
Figure imgf000349_0001
Table IV
Figure imgf000350_0001
Table IV
Figure imgf000351_0001
Table IV
Figure imgf000352_0001
Table IV
Figure imgf000353_0001
Table IV
Figure imgf000354_0001
Table IV
Figure imgf000355_0001
Table IV
Figure imgf000356_0001
Table IV
Figure imgf000357_0001
Table IV
Figure imgf000358_0001
Table IV
Figure imgf000359_0001
Table IV
Figure imgf000360_0001
Table IV
Figure imgf000361_0001
Table IV
Figure imgf000362_0001
Table IV
Figure imgf000363_0001
Table IV
Figure imgf000364_0001
Table IV
Figure imgf000365_0001
Table IV
Figure imgf000366_0001
Table IV
Figure imgf000367_0001
Table IV
Figure imgf000368_0001
Table IV
Figure imgf000369_0001
Table IV
Figure imgf000370_0001
Table IV
Ul -4
©
Figure imgf000371_0001
Table IV
Figure imgf000372_0001
Table IV
l -4 >
Figure imgf000373_0001
Table IV
Ui ~4
Ui
Figure imgf000374_0001
Table IV
Figure imgf000375_0001
Table IV
Figure imgf000376_0001
Table IV
Figure imgf000377_0001
Table IV
Ul -4 -4
Figure imgf000378_0001
Table IV
Ul -4
00
Figure imgf000379_0001
Table IV
Figure imgf000380_0001
Table IV
Figure imgf000381_0001
Table IV
Ul
00
Figure imgf000382_0001
Table IV
Ul
00 »
Figure imgf000383_0001
Table IV
Ul
00 Ul
Figure imgf000384_0001
Table IV
Figure imgf000385_0001
Table IV
Ul 00
Figure imgf000386_0001
Table IV
Ul
00
Figure imgf000387_0001
Table IV
l 00 -4
Figure imgf000388_0001
Table IV
Ul
00 00
Figure imgf000389_0001
Table IV
Figure imgf000390_0001
Table IV
Figure imgf000391_0001
Table IV
Ul v©
Figure imgf000392_0001
Table IV
Ul »
Figure imgf000393_0001
Table IV
l v© l
Figure imgf000394_0001
Table IV
Figure imgf000395_0001
Table IV
Ul
Figure imgf000396_0001
Table IV
Figure imgf000397_0001
Table IV
Figure imgf000398_0001
Table IV
Figure imgf000399_0001
Table IV
Ul
Figure imgf000400_0001
Table IV
© ©
Figure imgf000401_0001
Table IV
4-
©
Figure imgf000402_0001
Table IV
© »
Figure imgf000403_0001
Table IV
4-
© Ul
Figure imgf000404_0001
Table IV
4- © 4-
Figure imgf000405_0001
Table IV
Figure imgf000406_0001
Table IV
Figure imgf000407_0001
Table IV
©
-4
Figure imgf000408_0001
Table IV
Figure imgf000409_0001
Table IV
© v©
Figure imgf000410_0001
Table IV
Figure imgf000411_0001
Table IV
Figure imgf000412_0001
Table IV
Figure imgf000413_0001
Table IV
Figure imgf000414_0001
Table IV
Figure imgf000415_0001
Table IV
Figure imgf000416_0001
Table IV
Figure imgf000417_0001
Table IV
Figure imgf000418_0001
Table IV
Figure imgf000419_0001
Table IV
Figure imgf000420_0001
Table IV
4- >
©
Figure imgf000421_0001
Table IV
Figure imgf000422_0001
Table IV
- > >
Figure imgf000423_0001
Table IV
»
Ul
Figure imgf000424_0001
Table IV
Figure imgf000425_0001
Table IV
Figure imgf000426_0001
Table IV
Figure imgf000427_0001
Table IV
»
-4
Figure imgf000428_0001
Table IV
Figure imgf000429_0001
Table IV
» ©
Figure imgf000430_0001
Table IV
Ul
©
Figure imgf000431_0001
Table IV
4- Ul
Figure imgf000432_0001
Table IV
Ul »
Figure imgf000433_0001
Table IV
Ul Ul
Figure imgf000434_0001
Table IV
Figure imgf000435_0001

Claims

CLAIMS:
1. A pharmaceutical co-crystal composition, comprising: an API and a co-crystal former, wherein the API is a liquid or a solid at room temperature and the co-crystal former is a solid at room temperature, and wherein the API and co-crystal former are hydrogen bonded to each other.
2. The pharmaceutical co-crystal composition according to claim 1, wherein:
(a) the co-crystal former is selected from a co-crystal former of Table I or Table II;
(b) the API is selected from an API of Table TV;
(c) the API is selected from an API of Table TV and the co-crystal former is selected from a co-crystal former of Table I or Table II;
(d) the API is a liquid at room temperature;
(e) the API is a solid at room temperature;
(f) the API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(g) the co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; (h) the difference in pKa between the API and the co-crystal former does not exceed 2; (i) the solubility of the co-crystal is increased as compared to the API;
(j) the dose response of the co-crystal is increased as compared to the API;
(k) the dissolution of the co-crystal is increased as compared to the API;
(1) the bioavailability of the co-crystal is increased as compared to the
API; (m) the stability of the co-crystal is increased as compared to the API;
(n) a difficult to salt or unsaltable API is incorporated into the co-crystal;
(o) the hygroscopicity of the co-crystal is decreased as compared to the
API; (p) an amorphous API is crystallized as a component of the co-crystal;
(q) the form diversity of the co-crystal is decreased as compared to the
API; or (r) the morphology of the co-crystal is modulated as compared to the API.
3. A pharmaceutical co-crystal composition, comprising: an API, a co-crystal former, and a third molecule; wherein the API is a liquid or a solid at room temperature and the co-crystal former is a solid at room temperature, and wherein the API and the third molecule are bonded to each other, and further wherein the co-crystal former and the third molecule are hydrogen bonded to each other.
4. The pharmaceutical co-crystal composition according to claim 3, wherein:
(a) the co-crystal former is selected from a co-crystal former of Table I or Table II;
(b) the API is selected from an API of Table TV;
(c) the API is selected from an API of Table IV and the co-crystal former is selected from a co-crystal former of Table I or Table II;
(d) the API is a liquid at room temperature; (e) the API is a solid at room temperature;
(f) the API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(g) the co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; or
(h) the difference in pKa between the API and the co-crystal former does not exceed 2; (i) the solubility of the co-crystal is increased as compared to the API;
(j) the dose response of the co-crystal is increased as compared to the
API; (k) the dissolution of the co-crystal is increased as compared to the API;
(1) the bioavailability of the co-crystal is increased as compared to the
API; (m) the stability of the co-crystal is increased as compared to the API;
(n) a difficult to salt or unsaltable API is incorporated into the co-crystal;
(o) the hygroscopicity of the co-crystal is decreased as compared to the
API; (p) an amorphous API is crystallized as a component of the co-crystal;
(q) the form diversity of the co-crystal is decreased as compared to the
API; or (r) the morphology of the co-crystal is modulated as compared to the API.
5. A pharmaceutical co-crystal composition, comprising: a first and a second API, wherein each API is either a liquid or a solid at room temperature, and wherein the APIs are hydrogen bonded to a molecule.
6. The pharmaceutical co-crystal composition according to claim 5, wherein:
(a) the first API is hydrogen bonded to the second API;
(b) an API is selected from an API of Table TV;
(c) each API is selected from an API of Table TV;
(d) an API is a liquid at room temperature and the other API is a solid at room temperature;
(e) each API is a solid at room temperature;
(f) an API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(g) each API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; (h) the difference in pKa between the first API and the second API does not exceed 2; (i) the solubility of the co-crystal is increased as compared to an API;
(j) the dose response of the co-crystal is increased as compared to an API;
(k) the dissolution of the co-crystal is increased as compared to an API;
(1) the bioavailability of the co-crystal is increased as compared to an
API; (m) the stability of the co-crystal is increased as compared to an API;
(n) a difficult to salt or unsaltable API is incorporated into the co-crystal;
(o) the hygroscopicity of the co-crystal is decreased as compared to an
API; (p) an amorphous API is crystallized as a component of the co-crystal;
(q) the form diversity of the co-crystal is decreased as compared to an
API; or (r) the morphology of the co-crystal is modulated as compared to an API.
7. A pharmaceutical co-crystal composition, comprising: a first and a second co- crystal former, wherein each co-crystal former is a solid at room temperature, and wherem both co-crystal formers are hydrogen bonded to a molecule.
8. The pharmaceutical co-crystal composition according to claim 7, wherein:
(a) the first co-crystal former is hydrogen bonded to the second co-crystal former;
(b) a co-crystal former is selected from a co-crystal former of Table I or Table II;
(c) each co-crystal former is selected from a co-crystal former of Table I or Table II; (d) a co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phospliinic acid, sulfomc acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(e) each co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(f) the difference in pKa between the first co-crystal former and the second co-crystal former does not exceed 2;
(g) the solubility of the co-crystal is increased as compared to a co-crystal former;
(h) the dose response of the co-crystal is increased as compared to a co- crystal former;
(i) the dissolution of the co-crystal is increased as compared to a co- crystal former;
(j) the bioavailability of the co-crystal is increased as compared to a co- crystal former;
(k) the stability of the co-crystal is increased as compared to a co-crystal former;
(1) a difficult to salt or unsaltable API is incorporated into the co-crystal; (m) the hygroscopicity of the co-crystal is decreased as compared to a co- crystal former;
(n) an amorphous API is crystallized as a component of the co-crystal;
(o) the form diversity of the co-crystal is decreased as compared to a co- crystal former; or
(p) the morphology of the co-crystal is modulated as compared to a co- crystal former.
9. The pharmaceutical co-crystal composition according to claim 1, wherein the API is selected from celecoxib, carbamazepine, itraconazole, olanzapine, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin, or ibuprofen.
10. The pharmaceutical co-crystal composition according to claim 1, 3, 5, or 7, further comprising a pharmaceutically acceptable diluent, excipient, or carrier.
11. A co-crystal comprising an API and a co-crystal former selected from the group consisting of:
(a) carbamazepine and saccharin;
(b) carbamazepine and nicotinamide;
(c) carbamazepine and trimesic acid;
(d) celecoxib and nicotinamide;
(e) olanzapine and nicotinamide;
(f) celecoxib and 18-crown-6;
(g) itraconazole and succinic acid; (h) itraconazole and fumaric acid; (i) itraconazole and L-tartaric acid; (j) itraconazole and L-malic acid;
(k) itraconazoleHCl and DL-tartaric acid;
(1) modafinil and malonic acid; (m) modafinil and glycolic acid;
(n) modafinil and maleic acid;
(o) topiramate and 18-crown-6;
(p) 5-fluorouracil and urea;
(q) hydrochlorothiazide and nicotinic acid;
(r) hydrochlorothiazide and 18-crown-6;
(s) hydrochlorothiazide and piperazine;
(t) acetaminophen and 4,4 '-bipyridine;
(u) phenytoin and pyridone;
(v) aspirin and 4,4 '-bipyridine;
(w) ibuprofen and 4,4 '-bipyridine;
(x) flurbiprofen and 4,4 '-bipyridine;
(y) flurbiprofen and trans- l,2-bis(4-pyridyl) ethylene;
(z) carbamazepine and p-phthalaldehyde;
(aa) carbamazepine and 2,6-pyridinecarboxylic acid;
(bb) carbamazepine and 5-nitroisophthalic acid;
(cc) carbamazepine and 1,3,5,7-adamantane tetracarboxylic acid; and
(dd) carbamazepine and benzoquinone.
12. A process for preparing a pharmaceutical co-crystal composition comprising an API and a co-crystal former, comprising:
(a) providing an API and a co-crystal former, wherein the API is a liquid or a solid at room temperature and the co-crystal former is a solid at room temperature;
(b) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions, so as to form a solid phase, wherein the API and co-crystal former are hydrogen bonded to each other;
(c) isolating co-crystals formed thereby; and (d) incorporating the co-crystals into a pharmaceutical composition.
13. The process of claim 12, wherein:
(a) the co-crystal former is selected from a co-crystal former of Table I or Table II;
(b) the API is selected from an API of Table TV;
(c) the API is selected from an API of Table TV and the co-crystal former is selected from a co-crystal former of Table I or Table II;
(d) the API is a liquid at room temperature;
(e) the API is a solid at room temperature;
(f) the API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(g) the co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; or
(h) the difference in pKa between the API and the co-crystal former does not exceed 2.
14. A process for preparing a pharmaceutical co-crystal composition comprising an API, a co-crystal former, and a third molecule, comprising:
(a) providing an API and a co-crystal former, wherein the API is a liquid or a solid at room temperature and the co-crystal former is a solid at room temperature;
(b) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions, so as to form a solid phase, wherein the API and the third molecule are bonded to each other, and further wherein the co-crystal former and the third molecule are hydrogen bonded to each other;
(c) isolating co-crystals formed thereby; and
(d) incorporating the co-crystals into a pharmaceutical composition.
15. The process of claim 14, wherein:
(a) the co-crystal former is selected from a co-crystal former of Table I or Table II;
(b) the API is selected from an API of Table TV;
(c) the API is selected from an API of Table IV and the co-crystal former is selected from a co-crystal former of Table I or Table II;
(d) the API is a liquid at room temperature;
(e) the API is a solid at room temperature;
(f) the API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfomc acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; (g) the co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; or
(h) the difference in pKa between the API and the co-crystal former does not exceed 2.
16. A process for preparing a pharmaceutical co-crystal composition comprising a first and a second API, comprising:
(a) providing a first and a second API, wherein each API is either a liquid or a solid at room temperature;
(b) grinding, heating, co-subliming, co-melting, or contacting in solution the APIs under crystallization conditions, so as to form a solid phase, wherein the APIs are hydrogen bonded to a molecule;
(c) isolating co-crystals formed thereby; and
(d) incorporating the co-crystals into a pharmaceutical composition.
17. The process of claim 16, wherein:
(a) the first API is hydrogen bonded to the second API;
(b) an API is selected from an API of Table TV;
(c) each API is selected from an API of Table IV;
(d) an API is a liquid at room temperature and the other API is a solid at room temperature;
(e) each API is a solid at room temperature;
(f) an API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(g) each API has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; or
(h) the difference in pKa between the first API and the second API does not exceed 2.
18. A process for preparing a pharmaceutical co-crystal composition comprising a first and a second co-crystal former, comprising:
(a) providing a first and a second co-crystal former, wherein each co- crystal former is a solid at room temperature;
(b) grinding, heating, co-subliming, co-melting, or contacting in solution the co-crystal formers under crystallization conditions, so as to form a solid phase, wherein both co-crystal formers are hydrogen bonded to a molecule;
(c) isolating co-crystals formed thereby; and
(d) incorporating the co-crystals into a pharmaceutical composition.
19. The process of claim 18, wherein: (a) the first co-crystal former is hydrogen bonded to the second co-crystal former;
(b) a co-crystal former is selected from a co-crystal former of Table I or Table II;
(c) each co-crystal former is selected from a co-crystal former of Table I or Table II;
(d) a co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyπole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine;
(e) each co-crystal former has at least one functional group selected from the group consisting of: ether, thioether, alcohol, thiol, aldehyde, ketone, thioketone, nitrate ester, phosphate ester, thiophosphate ester, ester, thioester, sulfate ester, carboxylic acid, phosphonic acid, phosphinic acid, sulfonic acid, amide, primary amine, secondary amine, ammonia, tertiary amine, imine, thiocyanate, cyanamide, oxime, nitrile, diazo, organohalide, nitro, S-heterocyclic ring, thiophene, N-heterocyclic ring, pyrrole, O-heterocyclic ring, furan, epoxide, peroxide, hydroxamic acid, imidazole, and pyridine; or
(f) the difference in pKa between the first co-crystal former and the second co-crystal former does not exceed 2.
20. The process of claim 12, wherein the API is selected from celecoxib, carbamazepine, itraconazole, olanzapine, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin, or ibuprofen.
21. The process of claim 12, further comprising: incorporating a pharmaceutically acceptable diluent, excipient, or carrier.
22. A process of preparing a co-crystal comprising an API and a co-crystal former, comprising:
(a) providing an API and a co-crystal former;
(b) grinding, heating, co-subliming, co-melting, or contacting in solution the API with the co-crystal former under crystallization conditions, so as to form a solid phase; and
(c) isolating co-crystals formed thereby; wherein the API and the co-crystal former, respectively, are selected from the group consisting of: carbamazepine and saccharin, carbamazepine and nicotinamide, carbamazepine and trimesic acid, celecoxib and nicotinamide, olanzapine and nicotinamide, celecoxib and 18-crown-6, itraconazole and succinic acid, itraconazole and fumaric acid, itraconazole and tartaric acid, itraconazole and malic acid, itraconazoleHCl and tartaric acid, modafinil and malonic acid, modafinil and glycolic acid, modafinil and maleic acid, topiramate and 18-crown-6, 5-fluorouracil and urea, hydrochlorothiazide and nicotinic acid, hydrochlorothiazide and 18-crown-6, hydrochlorothiazide and piperazine, acetaminophen and 4,4 '-bipyridine, phenytoin and pyridone, aspirin and 4,4 '-bipyridine, ibuprofen and 4,4 '-bipyridine, flurbiprofen and 4,4'-bipyridine, flurbiprofen and trans- l,2-bis(4-pyridyl) ethylene, carbamazepine and p-phthalaldehyde, carbamazepine and 2,6-pyridinecarboxylic acid, carbamazepine and 5-nitroisophthalic acid, carbamazepine and 1,3,5,7-adamantane tetracarboxylic acid, and carbamazepine and benzoquinone.
23. A process for modulating the solubility of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound; (b) isolating the co-crystal, wherein the co-crystal has a modulated solubility as compared to the API; and
(c) incorporating the co-crystal having modulated solubility into a pharmaceutical composition.
24. The process of claim 23, wherein the solubility of the co-crystal is increased as compared to the API.
25. A process for modulating the dose response of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has a modulated dose response as compared to the API; and
(c) incorporating the co-crystal having modulated dose response into a pharmaceutical composition.
26. The process of claim 25, wherein the dose response of the co-crystal is increased as compared to the API.
27. A process for modulating the dissolution of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has a modulated dissolution as compared to the API; and (c) incorporating the co-crystal having modulated dissolution into a pharmaceutical composition.
28. The process of claim 27, wherein the dissolution of the co-crystal is increased as compared to the API.
29. A process for modulating the bioavailability of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has a modulated bioavailability as compared to the API; and
(c) incorporating the co-crystal having modulated bioavailability into a pharmaceutical composition.
30. The process of claim 29, wherein the bioavailability of the co-crystal is increased as compared to the API.
31. A process for increasing the stability of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has increased stability as compared to the API; and
(c) incorporating the co-crystal having increased stability into a pharmaceutical composition.
32. A process for the incorporation of a difficult to salt or unsaltable API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal;
(c) incorporating the co-crystal having a difficult to salt or unsaltable API into a pharmaceutical composition.
33. A process for decreasing the hygroscopicity of an API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has decreased hygroscopicity as compared to the API; and
(c) , incorporating the co-crystal having decreased hygroscopicity into a pharmaceutical composition.
34. A process for crystallizing an amorphous API for use in a pharmaceutical composition, which process comprises:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal;
(c) incorporating the co-crystal into a pharmaceutical composition.
35. A process for decreasing the form diversity of an API for use in a pharmaceutical composition, which process includes:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has decreased form diversity as compared to the API; and
(c) incorporating the co-crystal having decreased foπn diversity into a pharmaceutical composition.
36. A process for modulating the morphology of an API for use in a pharmaceutical composition, which process includes:
(a) grinding, heating, co-subliming, co-melting, or contacting in solution the API with a co-crystal forming compound under crystallization conditions, so as to form a co-crystal of the API and the co-crystal forming compound;
(b) isolating the co-crystal, wherein the co-crystal has a different morphology as compared to the API; and
(c) incorporating the co-crystal having modulated morphology into a pharmaceutical composition.
37. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an amino-pyridine functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) water;
(e) an alcohol;
(f) a primary amine; (g) a secondary amine;
(h) a carbonyl;
(i) a sulfoxo moiety;
(j) an ether;
(k) an ester;
(1) an aromatic N;
(m) a cyano moiety;
(n) a nitro moiety;
(o) a chloride moiety;
(p) a bromide moiety;
(q) a primary amide where the interaction distance is between about 2.97 and about 3.07 angstroms; (r) a secondary amide where the interaction distance is between about 2.70 and about 3.20 angstroms; (s) a secondary amide where the interaction distance is between about 2.75 and about 3.17 angstroms; (t) a carboxylic acid where the interaction distance is between about 2.72 and about 3.07 angstroms; (u) a carboxylic acid where the interaction distance is between about 2.54 and about 2.82 angstroms; (v) water where the interaction distance is between about 2.72 and about 3.15 angstroms; (w) water where the interaction distance is between about 2.65 and about 3.15 angstroms; (x) an alcohol where the interaction distance is between about 2.78 and about
3.14 angstroms; (y) an alcohol where the interaction distance is between about 2.63 and about
3.06 angstroms; (z) a primary amine where the interaction distance is between about 2.85 and about 3.25 angstroms; (aa) a secondary amine where the interaction distance is between about 2.83 and about 3.25 angstroms; (bb) a carbonyl where the interaction distance is between about 2.87 and about
3.10 angstroms; (cc) a sulfoxo moiety where the interaction distance is between about 2.70 and about 3.10 angstroms; (dd) an ether where the interaction distance is between about 2.84 and about
3.20 angstroms; (ee) an ester where the interaction distance is about 3.09 angstroms; (ff) an ester where the interaction distance is between about 2.85 and about
3.16 angstroms; (gg) an aromatic N where the interaction distance is between about 2.78 and about 3.25 angstroms; (hh) a cyano moiety where the interaction distance is between about 2.83 and about 3.30 angstroms; (ii) a nitro moiety where the interaction distance is between about 2.85 and about 3.28 angstroms; (jj) a chloride moiety where the interaction distance is between about 3.10 and about 3.45 angstroms; or (kk) a bromide moiety where the interaction distance is between about 3.27 and about 3.48 angstroms.
38. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a primary amine functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) water; (g) an alcohol;
(h) a carbonyl;
(i) a sulfoxo moiety;
(j) a sulfonyl;
(k) an ether;
(1) an ester;
(m) an aromatic N;
(n) a cyano moiety;
(o) a nitro moiety;
(p) a chloride moiety;
(q) a bromide moiety;
(r) a primary amide where the interaction distance is between about 2.73 and about 3.20 angstroms; (s) a secondary amide where the interaction distance is between about 2.65 and about 3.20 angstroms; (t) a carboxylic acid where the interaction distance is between about 2.74 and about 3.15 angstroms; (u) a carboxylic acid where the interaction distance is between about 2.72 and about 3.12 angstroms; (v) an amino-pyridine where the interaction distance is between about 3.10 and about 3.24 angstroms; (w) a sulfonamide where the interaction distance is between about 2.86 and about 3.17 angstroms; (x) water where the interaction distance is between about 2.65 and about 3.17 angstroms; (y) an alcohol where the interaction distance is between about 2.63 and about
3.26 angstroms; (z) a carbonyl where the interaction distance is between about 2.64 and about
3.15 angstroms; (aa) a sulfoxo moiety where the interaction distance is between about 2.70 and about 3.10 angstroms; (bb) a sulfonyl where the interaction distance is between about 2.93 and about
3.12 angstroms; (cc) an ether where the interaction distance is between about 2.75 and about
3.25 angstroms; (dd) an ester where the interaction distance is between about 2.90 and about
3.20 angstroms; (ee) an ester where the interaction distance is between about 2.74 and about
3.27 angstroms; (ff) an aromatic N where the interaction distance is between about 2.92 and about 3.26 angstroms; (gg) a cyano moiety where the interaction distance is between about 2.83 and about 3.30 angstroms; (hh) a nitro moiety where the interaction distance is between about 2.75 and about-3.17 angstroms; (ii) a chloride moiety where the interaction distance is between about 3.07 and about 3.50 angstroms; or (jj) a bromide moiety where the interaction distance is between about 3.23 and about 3.60 angstroms.
39. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a primary sulfonamide functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) water;
(b) an alcohol;
(c) a primary amine;
(d) a secondary amine;
(e) a sulfonyl;
(f) an ether;
(g) an ester;
(h) a cyano moiety; (i) a nitro moiety; (j) a chloride moiety;
(k) water where the interaction distance is about 2.87 angstroms;
(1) an alcohol where the interaction distance is between about 2.85 and about
3.07 angstroms; (m) a primary amine where the interaction distance is between about 2.85 and about 3.20 angstroms; (n) a secondary amine where the interaction distance is between about 2.85 and about 3.20 angstroms; (o) a sulfonyl where the interaction distance is between about 2.85 and about
3.20 angstroms; (p) an ether where the interaction distance is between about 2.90 and about
3.20 angstroms; (q) an ester where the interaction distance is between about 2.85 and about
3.12 angstroms; (r) a cyano moiety where the interaction distance is about 3.00 angstroms; (s) a nitro moiety where the interaction distance is between about 3.00 and about 3.20 angstroms; or (t) a chloride moiety where the interaction distance is between about 3.20 and about 3.32 angstroms.
40. The phannaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a primary amide functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a secondary amide;
( ) a carboxylic acid;
(c) an amino-pyridine;
(d) an aromatic N;
(e) water;
(f) an alcohol;
(g) a secondary amine;
(h) a carbonyl; (i) a sulfonyl;
(j) an ether;
(k) an ester;
(1) a cyano moiety;
(m) a nitro moiety;
(n) a chloride moiety;
(o) a bromide moiety;
(p) a secondary amide where the interaction distance is between about 2.70 and about 3.15 angstroms; (q) a carboxylic acid where the interaction distance is between about 2.40 and about 2.80 angstroms; (r) a carboxylic acid where the interaction distance is between about 2.80 and about 3.25 angstroms; (s) an amino-pyridine where the interaction distance is between about 2.90 and about 3.20 angstroms; (t) an amino-pyridine where the interaction distance is between about 2.80 and about 3.10 angstroms; (u) an aromatic N where the interaction distance is between about 2.90 and about 3.21 angstroms; (v) water where the interaction distance is between about 2.60 and about 3.00 angstroms; (w) water where the interaction distance is between about 2.70 and about 3.07 angstroms; (x) an alcohol where the interaction distance is between about 2.50 and about
3.00 angstroms; (y) an alcohol where the interaction distance is between about 2.70 and about
3.10 angstroms; (z) a secondary amine where the interaction distance is between about 2.80 and about 3.10 angstroms; (aa) a secondary amine where the interaction distance is between about 3.00 and about 3.15 angstroms; (bb) a carbonyl where the interaction distance is between about 2.80 and about
3.15 angstroms; (cc) a sulfonyl where the interaction distance is between about 2.90 and about
3.00 angstroms; (dd) an ether where the interaction distance is between about 2.80 and about
3.10 angstroms; (ee) an ester where the interaction distance is between about 2.70 and about
3.05 angstroms; (ff) a cyano moiety where the interaction distance is between about 3.00 and about 3.30 angstroms; (gg) a nitro moiety where the interaction distance is between about 2.90 and about 3.07 angstroms; (hh) a chloride moiety where the interaction distance is between about 3.10 and about 3.60 angstroms; or (ii) a bromide moiety where the interaction distance is between about 3.30 and about 3.80 angstroms.
41. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a secondary amide functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a carboxylic acid;
(c) an amino-pyridine;
( a sulfonamide;
(e) an aromatic N;
(f> water;
(g) an alcohol;
GO a primary amine;
(i) a secondary amine:
G) a carbonyl;
(k) a sulfonyl; (1) an ether;
(m) an ester;
(n) a cyano moiety;
(o) a nitro moiety;
(p) a chloride moiety;
(q) a bromide moiety;
(r) a primary amide where the interaction distance is between about 2.70 and about 3.15 angstroms; (s) a carboxylic acid where the interaction distance is between about 2.70 and about 3.10 angstroms; (t) a carboxylic acid where the interaction distance is between about 2.40 and about 3.05 angstroms; (u) an amino-pyridine where the interaction distance is between about 2.70 and about 3.20 angstroms; (v) an amino-pyridine where the interaction distance is between about 2.75 and about 3.17 angstroms; (w) a sulfonamide where the interaction distance is between about 2.70 and about 3.00 angstroms; (x) an aromatic N where the interaction distance is between about 2.60 and about 3.15 angstroms; (y) water where the interaction distance is between about 2.40 and about 3.10 angstroms; (z) water where the interaction distance is between about 2.60 and about 3.10 angstroms; (aa) an alcohol where the interaction distance is between about 2.50 and about
3.04 angstroms; (bb) an alcohol where the interaction distance is between about 2.50 and about
3.20 angstroms; (cc) a primary amine where the interaction distance is between about 2.65 and about 3.20 angstroms; (dd) a secondary amine where the interaction distance is between about 2.60 and about 3.15 angstroms; (ee) a carbonyl where the interaction distance is between about 2.70 and about
3.07 angstroms; (ff) a sulfonyl where the interaction distance is between about 2.60 and about
3.25 angstroms; (gg) an ether where the interaction distance is between about 2.70 and about
3.16 angstroms; (hh) an ester where the interaction distance is between about 2.80 and about
3.16 angstroms; (ii) a cyano moiety where the interaction distance is between about 2.90 and about 3.30 angstroms; (jj) a nitro moiety where the interaction distance is between about 2.80 and about 3.10 angstroms; (kk) a chloride moiety where the interaction distance is between about 2.90 and about 3.40 angstroms; or (11) a bromide moiety where the interaction distance is between about 3.10 and about 3.50 angstroms.
42. The pharmaceutical co-crystal composition accordmg to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an alcohol functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) an aromatic N;
(g) water;
(h) a primary amine; (i) a secondary amine; (j) a carbonyl;
(k) a sulfonyl;
(1) an ether;
(m) an ester;
(n) a cyano moiety;
(o) a nitro moiety;
(p) a chloride moiety;
(q) a bromide moiety;
(r) a primary amide where the interaction distance is between about 2.50 and about 3.00 angstroms; (s) a primary amide where the interaction distance is between about 2.70 and about 3.10 angstroms; (t) a secondary amide where the interaction distance is between about 2.50 and about 3.04 angstroms; (u) a secondary amide where the interaction distance is between about 2.50 and about 3.20 angstroms; (v) a carboxylic acid where the interaction distance is between about 2.50 and about 3.00 angstroms; (w) a carboxylic acid where the interaction distance is between about 2.40 and about 2.90 angstroms; (x) an amino-pyridine where the interaction distance is between about 2.60 and about 3.06 angstroms; (y) an amino-pyridine where the interaction distance is between about 2.75 and about 3.15 angstroms; (z) a sulfonamide where the interaction distance is between about 2.80 and about 3.07 angstroms; (aa) an aromatic N where the interaction distance is between about 2.50 and about 3.00 angstroms; (bb) water where the interaction distance is between about 2.40 and about 3.03 angstroms; (cc) a primary amine where the interaction distance is between about 2.60 and about 3.15 angstroms; (dd) a secondary amine where the interaction distance is between about 2.60 and about 3.15 angstroms; (ee) a carbonyl where the interaction distance is between about 2.40 and about
3.05 angstroms; (ff) a sulfonyl where the interaction distance is between about 2.40 and about
3.15 angstroms; (gg) an ether where the interaction distance is between about 2.40 and about
3.00 angstroms; (hh) an ester where the interaction distance is between about 2.50 and about
3.10 angstroms; (ii) a cyano moiety where the interaction distance is between about 2.40 and about 3.10 angstroms; (jj) a nitro moiety where the interaction distance is between about 2.45 and about 3.05 angstroms; (kk) a chloride moiety where the interaction distance is between about 2.60 and about 3.30 angstroms; or (11) a bromide moiety where the interaction distance is between about 3.00 and about 3.50 angstroms.
43. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a carboxylic acid functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) an amino-pyridine;
(d) an aromatic N;
(e) water;
(f) an alcohol;
(g) a primary amine; (h) a secondary amine;
(i) a carbonyl;
(j) an ether;
(k) an ester;
(1) a cyano moiety;
(m) a nitro moiety;
(n) a chloride moiety;
(o) a bromide moiety;
(p) a primary amide where the interaction distance is between about 2.80 and about 3.25 angstroms; (q) a primary amide where the interaction distance is between about 2.40 and about 2.80 angstroms; (r) a secondary amide where the interaction distance is between about 2.70 and about 3.10 angstroms; (s) a secondary amide where the interaction distance is between about 2.40 and about 3.05 angstroms; (t) an amino-pyridine where the interaction distance is between about 2.50 and about 2.80 angstroms; (u) an amino-pyridine where the interaction distance is between about 2.70 and about 3.00 angstroms; (v) an aromatic N where the interaction distance is between about 2.54 and about 2.94 angstroms; (w) water where the interaction distance is between about 2.50 and about 3.00 angstroms; (x) water where the interaction distance is between about 2.40 and about 3.00 angstroms; (y) an alcohol where the interaction distance is between about 2.50 and about
3.00 angstroms; (z) an alcohol where the interaction distance is between about 2.50 and about
2.90 angstroms; (aa) a primary amine where the interaction distance is between about 2.70 and about 3.10 angstroms; (bb) a secondary amine where the interaction distance is between about 2.70 and about 3.10 angstroms; (cc) a carbonyl where the interaction distance is between about 2.40 and about
3.00 angstroms; (dd) an ether where the interaction distance is between about 2.50 and about
3.00 angstroms; (ee) an ester where the interaction distance is between about 2.40 and about
3.05 angstroms; (ff) an ester where the interaction distance is between about 2.40 and about
3.10 angstroms; (gg) a cyano moiety where the interaction distance is between about 2.50 and about 2.80 angstroms; (hh) a nitro moiety where the interaction distance is between about 2.70 and about 3.05 angstroms; (ii) a chloride moiety where the interaction distance is between about 2.80 and about 3.20 angstroms; or (jj) a bromide moiety where the interaction distance is between about 3.00 and about 3.30 angstroms.
44. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a carbonyl functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a secondary sulfonamide:
(f) water;
(g) an alcohol; (h) a primary amine;
(i) a secondary amine;
(j) a primary amide where the interaction distance is between about 2.83 and about 3.15 angstroms; (k) a secondary amide where the interaction distance is between about 2.70 and about 3.07 angstroms; (1) a carboxylic acid where the interaction distance is between about 2.40 and about 3.00 angstroms; (m) an amino-pyridine where the interaction distance is between about 2.87 and about 3.10 angstroms; (n) a secondary sulfonamide where the interaction distance is between about
2.76 and about 3.22 angstroms; (o) water where the interaction distance is between about 2.55 and about 3.05 angstroms; (p) an alcohol where the interaction distance is between about 2.40 and about
3.05 angstroms; (q) a primary amine where the interaction distance is between about 2.64 and about 3.15 angstroms; or (r) a secondary amine where the interaction distance is between about 2.64 and about 3.15 angstroms.
45. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a cyano group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a primary sulfonamide;
(f) a secondary sulfonamide;
(g) water; (h) an alcohol;
(i) a primary amine;
(j) a secondary amine;
(k) a primary amide where the interaction distance is between about 3.01 and about 3.30 angstroms; (1) a secondary amide where the interaction distance is between about 2.90 and about 3.30 angstroms; (m) a carboxylic acid where the interaction distance is between about 2.57 and about 3.00 angstroms; (n) an amino-pyridine where the interaction distance is between about 2.84 and about 3.33 angstroms; (o) a primary sulfonamide where the interaction distance is about 2.99 angstroms; (p) a secondary sulfonamide where the interaction distance is between about
2.83 and about 3.00 angstroms; (q) water where the interaction distance is between about 2.78 and about 3.20 angstroms; (r) an alcohol where the interaction distance is between about 2.72 and about
3.13 angstroms; (s) a primary amine where the interaction distance is between about 2.84 and about 3.27 angstroms; or (t) a secondary amine where the interaction distance is between about 2.84 and about 3.30 angstroms.
46. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a sulfonyl group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a primary sulfonamide;
(d) a secondary sulfonamide; (e) water;
(f) an alcohol;
(g) a primary amine; (h) a secondary amine;
(i) a primary amide where the interaction distance is about 2.92 angstroms; (j) a secondary amide where the interaction distance is between about 2.95 and about 3.25 angstroms; (k) a primary sulfonamide where the interaction distance is between about
2.85 and about 3.10 angstroms; (1) a secondary sulfonamide where the interaction distance is between about
2.85 and about 3.20 angstroms; (m) water where the interaction distance is between about 2.84 and about 3.00 angstroms; (n) an alcohol where the interaction distance is between about 2.65 and about
3.15 angstroms; (o) a primary amine where the interaction distance is between about 2.93 and about 3.32 angstroms; or (p) a secondary amide where the interaction distance is between about 2.75 and about 3.32 angstroms.
47. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an aromatic N as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) water;
(f) an alcohol;
(g) a primary amine; (h) a secondary amine; (i) a primary amide where the interaction distance is between about 2.90 and about 3.21 angstroms; (j) a secondary amide where the interaction distance is between about 2.60 and about 3.15 angstroms; (k) a carboxylic acid where the interaction distance is between about 2.54 and about 2.94 angstroms; (1) an amino-pyridine where the interaction distance is between about 2.70 and about 3.20 angstroms; (m) water where the interaction distance is between about 2.60 and about 3.15 angstroms; (n) an alcohol where the interaction distance is between about 2.50 and about
3.00 angstroms; (o) a primary amine where the interaction distance is between about 2.92 and about 3.26 angstroms; or (p) a secondary amine where the interaction distance is between about 2.73 and about 3.25 angstroms.
48. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an ether functional group as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) water;
(g) an alcohol;
(h) a primary amine; (i) a secondary amine;
(j) a primary amide where the interaction distance is between about 2.80 and about 3.10 angstroms; (k) a secondary amide where the interaction distance is between about 2.70 and about 3.16 angstroms; (1) a carboxylic acid where the interaction distance is between about 2.50 and about 3.02 angstroms; (m) an amino-pyridine where the interaction distance is between about 2.80 and about 3.20 angstroms; (n) a sulfonamide where the interaction distance is less than about 3.20 angstroms; (o) water where the interaction distance is between about 2.40 and about 3.15 angstroms; (p) an alcohol where the interaction distance is between about 2.40 and about
3.00 angstroms; (q) a primary amine where the interaction distance is between about 2.75 and about 3.25 angstroms; or (r) a secondary amine where the interaction distance is between about 2.60 and about 3.25 angstroms.
49. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a chloride moiety as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) water;
(g) an alcohol;
(h) a primary amine; (i) a secondary amine;
(j) a primary amide where the interaction distance is between about 3.10 and about 3.60 angstroms; (k) a secondary amide where the interaction distance is between about 2.90 and about 3.30 angstroms; (1) a carboxylic acid where the interaction distance is between about 2.80 and about 3.30 angstroms; (m) an amino-pyridine where the interaction distance is between about 3.10 and about 3.45 angstroms; (n) a sulfonamide where the interaction distance is less than about 3.35 angstroms; (o) water where the interaction distance is between about 2.70 and about 3.30 angstroms; (p) an alcohol where the interaction distance is between about 2.50 and about
3.30 angstroms; (q) a primary amine where the interaction distance is between about 3.00 and about 3.50 angstroms; or (r) a secondary amine where the interaction distance is between about 2.90 and about 3.40 angstroms.
50. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an organochloride moiety as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) water;
(g) an alcohol;
(h) a primary amine; (i) a secondary amine;
(j) a primary amide where the interaction distance is between about 3.18 and about 3.21 angstroms; (k) a secondary amide where the interaction distance is between about 3.20 and about 3.27 angstroms; (1) a carboxylic acid where the interaction distance is between about 2.90 and about 3.23 angstroms; (m) an amino-pyridine where the interaction distance is between about 3.28 and about 3.33 angstroms; (n) a sulfonamide where the interaction distance is less than about 3.50 angstroms; (o) water where the interaction distance is between about 2.79 and about 3.26 angstroms; (p) an alcohol where the interaction distance is between about 2.90 and about
3.29 angstroms; (q) a primary amine where the interaction distance is between about 3.21 and about 3.29 angstroms; or (r) a secondary amine where the interaction distance is between about 3.26 and about 3.30 angstroms.
51. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises a bromide moiety as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) an alcohol;
(f) a primary amine;
(g) a secondary amine;
(h) a primary amide where the interaction distance is between about 3.30 and about 3.80 angstroms; (i) a secondary amide where the interaction distance is between about 3.10 and about 3.80 angstroms; (j) a carboxylic acid where the interaction distance is between about 3.00 and about 3.30 angstroms; (k) an amino-pyridine where the interaction distance is between about 3.20 and about 3.50 angstroms; (1) an alcohol where the interaction distance is between about 3.00 and about
3.50 angstroms; (m) a primary amine where the interaction distance is between about 3.20 and about 3.60 angstroms; or (n) a secondary amine where the interaction distance is between about 3.10 and about 3.60 angstroms.
52. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an organobromide moiety as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) a sulfonamide;
(f) water;
(g) an alcohol;
(h) a primary amine;
(i) a secondary amine;
(j) a primary amide where the interaction distance is less than about 3.50 angstroms; (k) a secondary amide where the interaction distance is less than about 3.50 angstroms; • (1) a carboxylic acid where the interaction distance is between about 3.01 and about 3.31 angstroms; (m) an amino-pyridine where the interaction distance is less than about 3.50 angstroms; (n) a sulfonamide where the interaction distance is less than about 3.50 angstroms;
(0) water where the interaction distance is between about 3.14 and about 3.27 angstroms;
(p) an alcohol where the interaction distance is between about 2.90 and about
3.36 angstroms; (q) a primary amine where the interaction distance is less than about 3.50 angstroms; or (r) a secondary amine where the interaction distance is between about 3.20 and about 3.39 angstroms.
53. The pharmaceutical co-crystal composition according to claims 1, 3, 5, or 7, wherein the API or co-crystal former comprises an organoiodide moiety as a hydrogen bonded moiety and another hydrogen bonded moiety comprises:
(a) a primary amide;
(b) a secondary amide;
(c) a carboxylic acid;
(d) an amino-pyridine;
(e) an aromatic N;
(f) an alcohol;
(g) a primary amine; (h) a secondary amine;
(i) a primary amide where the interaction distance is less than about 3.80 angstroms; (j) a secondary amide where the interaction distance is less than about 3.80 angstroms; (k) a carboxylic acid where the interaction distance is less than about 3.80 angstroms;
(1) an amino-pyridine where the interaction distance is less than about 3.80 angstroms; (m) an aromatic N where the interaction distance is between about 2.70 and about 3.23 angstroms; (n) an alcohol where the interaction distance is between about 2.90 and about
3.48 angstroms; (o) a primary amine where the interaction distance is between about 3.25 and about 3.42 angstroms; or (p) a secondary amine where the interaction distance is between about 2.71 and about 2.87 angstroms.
54. The pharmaceutical co-crystal composition according to claims 1 or 3, wherein the API forms a dimeric primary amide structure via hydrogen bonds with an R2 2 (8) motif, and further wherein the composition comprises:
(a) at least one hydrogen bond donor;
(b) at least two hydrogen bond donors;
(c) at least three hydrogen bond donors;
(d) at least four hydrogen bond donors;
(e) at least one hydrogen bond acceptor;
(f) at least two hydrogen bond acceptors;
(g) at least one hydrogen bond donor and one hydrogen bond acceptor; (h) at least two hydrogen bond donors and one hydrogen bond acceptor; (i) at least one hydrogen bond donor and two hydrogen bond acceptors;
(j) at least two hydrogen bond donors and two hydrogen bond acceptors; or (k) at least three hydrogen bond donors and one hydrogen bond acceptor.
55. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises peaks at 3.77, 9.63, and 17.78 degrees; (ii) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises peaks at 9.63, 20.44, and 22.10 degrees; (iii) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises peaks at 14.76 and 21.19 degrees; (iv) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises peaks at 3.77 and 19.31 degrees; (v) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises peaks at 17.78 and 20.44 degrees; (vi) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises a peak at 3.77 degrees; or (vii) said co-crystal is a celecoxib :nicotinamide co-crystal and said X- ray diffraction pattern comprises a peak at 17.78 degrees;
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a celecoxib :nicotinamide co-crystal and said DSC thermogram comprises an endothermic transition at about 130 degrees C; or
(c) the co-crystal is characterized by a Raman spectrum comprising peaks expressed in terms of cm"1, wherein:
(i) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises peaks at 1599, 1162, and 1044; (ii) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises peaks at 1618, 1044, and 796; (iii) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises peaks at 1599 and 1044; (iv) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises a peak at 1044; (v) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises a peak at 1618; or (vi) said co-crystal is a celecoxib :nicotinamide co-crystal and said
Raman spectrum comprises a peak at 1599.
56. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 8.73, 13.13, and 18.45 degrees; (ii) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 8.73, 11.89, and 17.75
.degrees; (iii) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 16.37, 18.45, and 23.11 degrees; (iv) said co-crystal is a celecoxib :18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 17.75 and 20.75 degrees; (v) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 8.73 and 13.13 degrees; (vi) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises peaks at 11.89 and 22.37 degrees; (vii) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises a peak at 8.73 degrees; (viii) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises a eak at 11.89 degrees; or (ix) said co-crystal is a celecoxib: 18-crown-6 co-crystal and said X-ray diffraction pattern comprises a peak at 17.75 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a celecoxib: 18-crown-6 co-crystal and said DSC thermogram comprises an endothermic transition at about 190 degrees C.
57. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 11.07, 13.83, and 18.03 degrees; (ii) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 10.79, 16.13, and 18.51 degrees; (iii) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 12.17, 18.03, and 21.43 degrees; (iv) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 11.07 and 18.03 degrees; (v) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 12.17 and 18.51 degrees; (vi) ' said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 16.13 and 21.43 degrees; (vii) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises a peak at 11.07 degrees; or (viii) said co-crystal is a topiramate: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises a peak at 13.83 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a topiramate: 18-crown-6 co-crystal and said DSC thermogram comprises an endothermic transition at about 135 degrees C.
58. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is an olanzapine:nicotinamide foπn I co-crystal and said X-ray diffraction pattern comprises peaks at 4.89, 8.65, and 17.15 degrees; (ii) said co-crystal is an olanzapine :nicotinamide form I co-crystal and said X-ray diffraction pattern comprises peaks at 17.15, 23.95, and
25.53 degrees; (iii) said co-crystal is an olanzapine:nicotinamide form I co-crystal and said X-ray diffraction pattern comprises peaks at 8.65, 19.71, and
26.71 degrees; (iv) said co-crystal is an olanzapinemicotinamide form I co-crystal and said X-ray diffraction pattern comprises peaks at 4.89 and 17.15 degrees; (v) said co-crystal is an olanzapine:nicotinamide foπn I co-crystal and said X-ray diffraction pattern comprises peaks at 8.65 and 23.95 degrees; (vi) said co-crystal is an olanzapine:nicotinamide form I co-crystal and said X-ray diffraction pattern comprises peaks at 23.95 and 25.53 degrees; (vii) said co-crystal is an olanzapine:nicotinamide form I co-crystal and said X-ray diffraction pattern comprises a peak at 4.89 degrees; or (viii) said co-crystal is an olanzapine micotinamide form I co-crystal and said X-ray diffraction pattern comprises a peak at 17.15 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is an olanzapine icotinamide form I co-crystal and said DSC thermogram comprises an endothermic transition at about 126 degrees C.
59. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is an olanzapine:nicotinamide form π co-crystal and said X-ray diffraction pattern comprises peaks at 8.65, 17.53, and 24.19 degrees; (b) said co-crystal is an olanzapine:nicotinarnide form II co-crystal and said X-ray diffraction pattern comprises peaks at 11.87, 14.53, and 19.69 degrees;
(c) said co-crystal is an olanzapine icotinamide form II co-crystal and said X-ray diffraction pattern comprises peaks at 8.65, 17.53, and 18.09 degrees;
(d) said co-crystal is an olanzapine :nicotinamide foπn II co-crystal and said X-ray diffraction pattern comprises peaks at 11.87 and 17.53 degrees;
(e) said co-crystal is an olanzapinemicotinamide form II co-crystal and said X-ray diffraction pattern comprises peaks at 8.65 and 14.53 degrees;
(f) said co-crystal is an olanzapine icotinamide form II co-crystal and said X-ray diffraction pattern comprises peaks at 11.87 and 24.19 degrees;
(g) said co-crystal is an olanzapine micotinamide form TL co-crystal and said X-ray diffraction pattern comprises a peak at 8.65 degrees;
(h) said co-crystal is an olanzapinemicotinamide form II co-crystal and said X-ray diffraction pattern comprises a peak at 17.53 degrees; or
(i) said co-crystal is an olanzapine icotinamide form II co-crystal and said X-ray diffraction pattern comprises a peak at 11.87 degrees.
60. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is an olanzapine :nicotinamide form III co-crystal and said X-ray diffraction pattern comprises peaks at 6.41, 12.85, and 18.67 degrees;
(b) said co-crystal is an olanzapine icotinamide form Til co-crystal and said X-ray diffraction pattern comprises peaks at 12.85, 21.85, and 24.37 degrees; (c) said co-crystal is an olanzapine :nicotinamide form III co-crystal and said X-ray diffraction pattern comprises peaks at 14.91, 18.67, and 21.85 degrees;
(d) said co-crystal is an olanzapine:nicotinamide form III co-crystal and said X-ray diffraction pattern comprises peaks at 6.41 and 12.85 degrees;
(e) said co-crystal is an olanzapine icotinamide form III co-crystal and said X-ray diffraction pattern comprises peaks at 6.41 and
, 18.67 degrees;
(f) said co-crystal is an olanzapinemicotinamide form III co-crystal and said X-ray diffraction pattern comprises peaks at 12.85 and 18.67 degrees;
(g) said co-crystal is an olanzapine icotinamide form III co-crystal and said X-ray diffraction pattern comprises a peak at 6.41 degrees;
(h) said co-crystal is an olanzapinemicotinamide form III co-crystal and said X-ray diffraction pattern comprises a peak at 12.85 degrees; or (i) said co-crystal is an olanzapine:nicotinamide form III co-crystal and said X-ray diffraction pattern comprises a peak at 18.67 degrees.
61. The. co-crystal according to claim 11 , wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a cw-itraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 3.01, 16.17, and 17.29 degrees; (ii) said co-crystal is a czAitraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.01, 15.87, and 24.47 degrees; (iii) said co-crystal is a cw-itraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 9.05, 20.41, and
22.27 degrees; (iv) said co-crystal is a cw-irraconazole:succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 3.01 and 17.29 degrees; (v) said co-crystal is a czAitraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.01 and 16.17 degrees; (vi) said co-crystal is a czAitraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 9.05 and 22.27 degrees; (vii) said co-crystal is a cw-itraconazole^uccinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 3.01 degrees; (viii) said co-crystal is a cw-itraconazole:succinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 16.17 degrees; or (ix) said co-crystal is a cω-itraconazole: succinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 17.29 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a czAitraconazole: succinic acid co-crystal and said DSC thermogram comprises an endothermic transition at about 160 degrees C.
62. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a czAitraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.61, 5.89, and 10.57 degrees; (ii) said co-crystal is a c/Aitraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 9.23, 19.05, and 20.79 degrees; (iii) said co-crystal is a czAitraconazoleώmaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 15.51, 16.23, and
16.93 degrees; (iv) said co-crystal is a ctAitraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.61 and 20.79 degrees; (v) said co-crystal is a czAitraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 5.89 and 19.05 degrees; (vi) said co-crystal is a
Figure imgf000484_0001
acid co-crystal and said X-ray diffraction pattern comprises peaks at 10.57 and 16.23 degrees; (vii) said co-crystal is a czAitraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 4.61 degrees; (viii) said co-crystal is a cw-itraconazole: fumaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 5.89 degrees; (ix) said co-crystal is a ct,s-ifraconazole:fumaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 10.57 degrees; or (x) said co-crystal is a cw-ifraconazole:fumaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 19.05 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a czAitraconazole: fumaric acid co-crystal and said DSC thermogram comprises an endothermic transition at about 180 degrees C.
63. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a c/>s-itraconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.13, 6.19, and 8.49 degrees; (ii) said co-crystal is a c7Aitraconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.19, 16.13, and
17.23 degrees; (iii) said co-crystal is a ctΛ traconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.49, 18.07, and
20.79 degrees; (iv) said co-crystal is a czAitraconazole: L-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.13 and 8.49 degrees; (v) said co-crystal is a cw-itraconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.19 and 20.79 degrees; (vi) said co-crystal is a cts-itraconazoleA-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 16.13 and 17.23 degrees; (vii) said co-crystal is a czAitraconazole:L-tartaric acid, co-crystal and said X-ray diffraction pattern comprises a peak at 4.13 degrees; (viii) said co-crystal is a czAitraconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 6.19 degrees; or (ix) said co-crystal is a czAitraconazole:L-tartaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 8.49 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a czAitraconazole:L-tartaric acid co-crystal and said DSC thermogram comprises an endothermic transition at about 181 degrees C.
64. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a cts-itraconazole:L-malic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.07, 8.85, and 17.05 degrees; (ii) said co-crystal is a czAitraconazole.-L-maric acid co-crystal and said X-ray diffraction pattern comprises peaks at 15.93, 20.49, and
22.85 degrees; (iii) said co-crystal is a czAitraconazole.-L-malic acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.85, 15.93 and
26.17 degrees; (iv) said co-crystal is a cw-itraconazole:L-malic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.07 and 17.05 degrees; (v) said co-crystal is a czAitraconazole: L-malic acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.85 and 21.27 degrees; (vi) said co-crystal is a czAitraconazole:L-malic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.07 and 8.85 degrees; (vii) said co-crystal is a czAitraconazole:L-malic acid co-crystal and said X-ray diffraction pattern comprises a peak at 6.07 degrees; (viii) said co-crystal is a czAitraconazole:L-malic acid co-crystal and said X-ray diffraction pattern comprises a peak at 8.85 degrees; or (ix) said co-crystal is a cw-itraconazole.'L-malic acid co-crystal and said X-ray diffraction pattern comprises a peak at 17.05 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a cM-itraconazole:L-malic acid co-crystal and said DSC thermogram comprises an endothermic transition at about 154 degrees C.
65. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a czΛMtraconazoleHCkDL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 3.73, 10.95, and 13.83 degrees; (ii) said co-crystal is a ezAitraconazoleHCkDL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 16.53, 17.75, and 19.65 degrees; (iii) said co-crystal is a cw-itraconazoleHCl:DL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 10.95, 16.53, and 21.11 degrees; (iv) said co-crystal is a cw-itraconazoleHC DL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 3.73 and
10.95 degrees; (v) said co-crystal is a cw-itraconazoleHCl:DL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 13.83 and
17.75 degrees; (vi) said co-crystal is a czAitraconazoleHCkDL-tartaric acid co-crystal and said X-ray diffraction pattern comprises peaks at 16.53 and
19.65 degrees; (vii) said co-crystal is a czAitraconazoleHC DL-tartaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 3.73 degrees; (viii) said co-crystal is a cw-itraconazoleHC DL-tartaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 10.95 degrees; or (ix) said co-crystal is a czAifraconazoleHCkDL-tartaric acid co-crystal and said X-ray diffraction pattern comprises a peak at 17.75 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a czAitraconazoleHCkDL-tartaric acid co-crystal and said DSC thermogram comprises an endothermic transition at about 162 degrees C.
66. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a modafinikmalonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 5.11, 9.35, and
16.87 degrees; (ii) said co-crystal is a modafinil:malonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 16.87, 18.33, and
19.53 degrees; (iii) said co-crystal is a modafinil:malonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 9.35, 19.53, and
22.89 degrees; (iv) said co-crystal is a modafini malonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 5.11 and 9.35 degrees; (v) said co-crystal is a modafinil:malonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 16.87 and 19.53 degrees; (vi) said co-crystal is a modafinil malonic acid form I co-crystal and said X-ray diffraction pattern comprises peaks at 18.33 and 22.89 degrees; (vii) said co-crystal is a modafinil :malonic acid form I co-crystal and said X-ray diffraction pattern comprises a peak at 5.11 degrees; (viii) said co-crystal is a modafinikmalonic acid form I co-crystal and said X-ray diffraction pattern comprises a peak at 9.35 degrees; or (ix) said co-crystal is a modafini malonic acid form I co-crystal and said X-ray diffraction pattern comprises a peak at 16.87 degrees;
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a modafinikmalonic acid form I co-crystal and said DSC thermogram comprises an endothermic transition at about 106 degrees C; or
(c) the co-crystal is characterized by a Raman spectrum comprising peaks expressed in terms of cm"1, wherein:
(i) said co-crystal is a modafinil malonic acid form I co-crystal and said Raman spectrum comprises peaks at 1004, 633, and 265; (ii) said co-crystal is a modafinihmalonic acid form I co-crystal and said Raman spectrum comprises peaks at 1032, 1601, and 767; (iii) said co-crystal is a modafinil:malonic acid form I co-crystal and said Raman spectrum comprises peaks at 1004 and 633; (iv) said co-crystal is a modafinihmalonic acid form I co-crystal and said Raman spectrum comprises peaks at 1183 and 767; or (v) said co-crystal is a modafinihmalonic acid form I co-crystal and said Raman spectrum comprises peaks at 1601 and 718.
67. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is a modafinihmalonic acid forni II co-crystal and said X-ray diffraction pattern comprises peaks at 5.90, 9.54, and 20.01 degrees;
(b) said co-crystal is a modafini malonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 15.79, 18.02, and 21.66 degrees; '
(c) said co-crystal is a modafini malonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 9.54, 20.01, and 25.30 degrees;
(d) said co-crystal is a modafinil malonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 5.90 and 9.54 degrees;
(e) said co-crystal is a modafini malonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 5.90 and 20.01 degrees;
(f) said co-crystal is a modafini malonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 9.54 and 20.01 degrees; (g) said co-crystal is a modafinihmalonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 5.90 degrees; or
(h) said co-crystal is a modafinikmalonic acid form II co-crystal and said X-ray diffraction pattern comprises peaks at 9.54 degrees.
68. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction patteπi comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is a modafinil: lycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 9.51, 15.97, and 20.03 degrees;
(b) said co-crystal is a modafinil: glycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 14.91, 19.01, and 22.75 degrees;
(c) said co-crystal is a modafϊni glycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 15.97, 25.03, and 25.71 degrees;
(d) said co-crystal is a modafinihglycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 9.51 and 15.97 degrees;
(e) said co-crystal is a modafinil: glycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 20.03 and 25.03 degrees;
(f) said co-crystal is a modafinil: glycolic acid co-crystal and said X-ray diffraction pattern comprises peaks at 15.97 and 25.03 degrees;
(g) said co-crystal is a modafinil: glycolic acid co-crystal and said X-ray diffraction pattern comprises a peak at 9.51 degrees;
(h) said co-crystal is a modafinil:glycolic acid co-crystal and said X-ray diffraction pattern comprises a peak at 15.97 degrees; or
(i) said co-crystal is a modafini glycolic acid co-crystal and said X-ray diffraction pattern comprises a peak at 20.03 degrees.
69. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (a) said co-crystal is a modafinil:maleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.69, 6.15, and 9.61 degrees;
(b) said co-crystal is a modafinihmaleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 10.23, 19.97, and 21.83 degrees;
(c) said co-crystal is a modafinil :maleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.69, 10.23, and 21.83 degrees;
(d) said co-crystal is a modafϊnikmaleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.69 and 19.97 degrees;
(e) said co-crystal is a modafinikmaleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 6.15 and 9.61 degrees;
(f) said co-crystal is a modafinil :maleic acid co-crystal and said X-ray diffraction pattern comprises peaks at 4.69 and 6.15 degrees;
(g) said co-crystal is a modafinil :maleic acid co-crystal and said X-ray diffraction pattern comprises a peak at 4.69 degrees;
(h) said co-crystal is a modafinil maleic acid co-crystal and said X-ray diffraction pattern comprises apeak at 9.61 degrees; or
(i) said co-crystal is a modafinil maleic acid co-crystal and said X-ray diffraction pattern comprises a peak at 19.97 degrees.
70. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 11.23, 13.27, and 16.93 degrees; (ii) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 12.69, 20.37, and 25.55 degrees; (iii) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 17.93, 23.65, and 26.87 degrees; (iv) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 11.23 and 16.93 degrees; (v) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 23.65 and 32.49 degrees; (vi) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises peaks at 13.27 and 25.55 degrees; (vii) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises a peak at 11.23 degrees; (viii) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises a peak at 16.93 degrees; or (ix) said co-crystal is a 5-fluorouracil:urea co-crystal and said X-ray diffraction pattern comprises a peak at 25.55 degrees;
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a 5-fluorouracil:urea co-crystal and said DSC thermogram comprises an endothermic transition at about 208 degrees C; or
(c) the co-crystal is characterized by a Raman spectrum comprising peaks expressed in terms of cm"1, wherein:
(i) said co-crystal is a 5-fluorouracil:urea co-crystal and said Raman spectrum comprises peaks at 1347, 1024, and 757; (ii) said co-crystal is a 5-fluorouracil:urea co-crystal and said Raman spectrum comprises peaks at 644, 545, and 472; (iii) said co-crystal is a 5-fluorouracil:urea co-crystal and said Raman spectrum comprises peaks at 1680 and 1347; (iv) said co-crystal is a 5-fluorouracil:urea co-crystal and said Raman spectrum comprises peaks at 1347 and 757; or (v) said co-crystal is a 5-fluorouracil:urea co-crystal and said Raman spectrum comprises peaks at 1024 and 757.
71. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (a) said co-crystal is a hydrochlorothiazide icotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.57, 13.23, and 21.13 degrees;
(b) said co-crystal is a hydrochlorothiazide:nicotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 14.31, 17.89, and 26.57 degrees;
(c) said co-crystal is a hydrochlorothiazide:nicotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.57, 21.13, and 25.73 degrees;
(d) said co-crystal is a hydrochlorothiazide cotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 8.57 and 21.13 degrees;
(e) said co-crystal is a hydrochlorothiazidemcotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 13.23 and 26.57 degrees;
(f) said co-crystal is a hydrochlorothiazide icotinic acid co-crystal and said X-ray diffraction pattern comprises peaks at 17.89 and 24.41 degrees;
(g) said co-crystal is a hydrochlorothiazide icotinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 8.57 degrees;
(h) said co-crystal is a hydrochlorothiazidemcotinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 13.23 degrees; or
(i) said co-crystal is a hydrochlorothiazidemcotinic acid co-crystal and said X-ray diffraction pattern comprises a peak at 21.13 degrees.
72. The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 9.97, 11.57, and 15.67 degrees;
(b) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 14.53, 19.05, and 20.31 degrees;
(c) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 16.61, 20.65, and 23.63 degrees; (d) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 9.97 and 10.43 degrees;
(e) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 12.83 and 15.67 degrees;
(f) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises peaks at 14.53 and 20.31 degrees;
(g) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises apeak at 10.43 degrees;
(h) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises a peak at 12.83 degrees; or
(i) said co-crystal is a hydrochlorothiazide: 18-crown-6 co-crystal and said X- ray diffraction pattern comprises a peak at 20.31 degrees.
73.' The co-crystal according to claim 11, wherein the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(a) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 6.85, 13.75, and 18.71 degrees;
(b) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 15.93, 23.27, and 24.17 degrees;
(c) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 18.17, 20.93, and 27.75 degrees;
(d) said co-crystal is a hydrochlorothiazide :piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 6.85 and 18.71 degrees;
(e) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 13.75 and 23.27 degrees;
(f) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises peaks at 15.93 and 24.17 degrees;
(g) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises a peak at 6.85 degrees; (h) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises a peak at 13.75 degrees; or
(i) said co-crystal is a hydrochlorothiazide:piperazine co-crystal and said X- ray diffraction pattern comprises a peak at 18.71 degrees.
74. The co-crystal according to claim 11, wherein the co-crystal is characterized by a DSC thermogram, wherein said co-crystal is an acetaminophen:4,4-bipyridine:water co- crystal and said DSC thermogram comprises an endothermic transition at about 58 degrees C.
75. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is a phenytoi pyridone co-crystal and said X-ray diffraction pattern comprises peaks at 5.2, 15.1, and 16.7 degrees; (ii) said co-crystal is a phenytoimpyridone co-crystal and said X-ray diffraction pattern comprises peaks at 11.1, 16.2, and 17.8 degrees; (iii) said co-crystal is a phenytoimpyridone co-crystal and said X-ray diffraction pattern comprises peaks at 5.2 and 15.1 degrees; (iv) said co-crystal is a phenytoimpyridone co-crystal and said X-ray diffraction pattern comprises peaks at 15.1 and 19.4 degrees; (v) said co-crystal is a phenytoimpyridone co-crystal and said X-ray diffraction pattern comprises a peak at 5.2 degrees; or (vi) said co-crystal is a phenytoimpyridone co-crystal and said X-ray diffraction pattern comprises a peak at 15.1 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a phenytoimpyridone co-crystal and said DSC thermogram comprises an endothermic transition at about 233 degrees C.
76. The co-crystal according to claim 11, wherein the co-crystal is characterized by a DSC thermogram, wherein said co-crystal is an aspirin:4,4-bipyridine co-crystal and said DSC thermogram comprises an endothermic transition at about 95 degrees C.
77. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is an ibuprofen: 4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 3.4 and 6.9 degrees; (ii) said co-crystal is an ibuprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 3.4 and 10.4 degrees; (iii) said co-crystal is an ibuprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 10.4 and 17.3 degrees; (iv) said co-crystal is an ibuprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises a peak at 3.4 degrees; or (v) said co-crystal is an ibuprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises a peak at 10.4 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is an ibuprofen:4,4-bipyridine co-crystal and said DSC theπnogram comprises an endothermic transition at about 119 degrees C.
78. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a flurbiprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 16.8, 18.1, and 20.0 degrees; (ii) said co-crystal is a flurbiprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 18.1, 21.3, and 25.0 degrees; (iii) said co-crystal is a flurbiprofen: 4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 16.8 and 19.0 degrees; (iv) said co-crystal is a flurbiprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises peaks at 17.1 and 21.3 degrees; (v) said co-crystal is a flurbiprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises apeak at 16.8 degrees; or (vi) said co-crystal is a flurbiprofen:4,4-bipyridine co-crystal and said
X-ray diffraction pattern comprises a peak at 19.0 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a flurbiprofen:4,4-bipyridine co-crystal and said DSC thermogram comprises an endothermic transition at about 163 degrees C.
79. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises peaks at
3.6, 17.3, and 18.4 degrees; (ii) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises peaks at
17.3, 19.1, and 23.8 degrees; (iii) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises peaks at
18.1 and 22.3 degrees; (iv) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises peaks at 3.6 and 18.4 degrees; (v) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises a peak at
3.6 degrees; or (vi) said co-crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said X-ray diffraction pattern comprises a peak at 19.1 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a flurbiprofen:trans-l,2-bis-(4-pyridyl) ethylene co-crystal and said DSC thermogram comprises an endothermic transition at about 164 degrees C.
80. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in teπns of 2-theta angles, wherein:
(i) said co-crystal is a carbamazepine :p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises peaks at 8.5, 11.9, and
15.1 degrees; (ii) said co-crystal is a carbamazepine:p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises peaks at 10.6, 14.4, and
18.0 degrees; (iii) said co-crystal is a carbamazepine :p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises peaks at 11.9 and 23.7 degrees; (iv) said co-crystal is a carbamazepine:p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises peaks at 8.5 and 14.4 degrees; (v) said co-crystal is a carbamazepine :p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises a peak at 8.5 degrees; or (vi) said co-crystal is a carbamazepine:p-phthalaldehyde co-crystal and said X-ray diffraction pattern comprises a peak at 11.9 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a carbamazepine :p-phthalaldehyde co-crystal and said DSC thermogram comprises an endothermic transition at about 128 degrees C.
81. The co-crystal according to claim 11 , wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is a carbamazepine :nicotinamide co-crystal and said
X-ray diffraction pattern comprises peaks at 8.8, 13.2, and 15.6 degrees; (ii) said co-crystal is a carbamazepine icotinamide co-crystal and said
X-ray diffraction pattern comprises peaks at 13.2, 15.6, and 20.4 degrees; (iii) said co-crystal is a carbamazepine :nicotinamide co-crystal and said
X-ray diffraction pattern comprises peaks at 8.8 and 26.4 degrees; (iv) said co-crystal is a carbamazepine micotinamide co-crystal and said
X-ray diffraction pattern comprises peaks at 13.2 and 15.6 degrees; (v) said co-crystal is a carbamazepine icotinamide co-crystal and said
X-ray diffraction pattern comprises a peak at 8.8 degrees; or (vi) said co-crystal is a carbamazepine:nicotinamide co-crystal and said
X-ray diffraction pattern comprises a peak at 15.6 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a carbamazepine :nicotinamide co-crystal and said DSC thermogram comprises an endothermic transition at about 157 degrees C.
82. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein: (i) said co-crystal is a carbamazepine:saccharin co-crystal and said X- ray diffraction pattern comprises peaks at 6.9, 13.6, and 15.3 degrees; (ii) said co-crystal is a carbamazepine: saccharin co-crystal and said X- ray diffraction pattern comprises peaks at 14.0, 20.2, and 28.3 degrees; (iii) said co-crystal is a carbamazepine: saccharin co-crystal and said X- ray diffraction pattern comprises peaks at 12.2 and 21.3 degrees; (iv) said co-crystal is a carbamazepine:saccharin co-crystal and said X- ray diffraction pattern comprises peaks at 14.0 and 20.2 degrees; (v) said co-crystal is a carbamazepine:saccharin co-crystal and said X- ray diffraction pattern comprises a peak at 14.0 degrees; or (vi) said co-crystal is a carbamazepine:saccharin co-crystal and said X- ray diffraction pattern comprises a peak at 21.3 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a carbamazepine:saccharin co-crystal and said DSC thermogram comprises an endothermic transition at about 177 degrees C.
The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises peaks at 10.14 and 17.44 degrees;
(ii) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises peaks at 15.29 and 21.17 degrees;
(iii) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises peaks at 10.14 and 15.29 degrees;
(iv) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises peaks at 21.17 and 31.41 degrees;
(v) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises a peak at 10.14 degrees; or (vi) said co-crystal is a carbamazepine:5-nitroisophthalic acid co- crystal and said X-ray diffraction pattern comprises a peak at 17.44 degrees; or (b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a carbamazepine:5-nitroisophthalic acid co-crystal and said DSC thermogram comprises an endotheπnic transition at about 191 degrees C.
84. The co-crystal according to claim 11, wherein:
(a) the co-crystal is characterized by a powder X-ray diffraction pattern comprising peaks expressed in terms of 2-theta angles, wherein:
(i) said co-crystal is a carbamazepine:trimesic acid co-crystal and said
X-ray diffraction pattern comprises peaks at 10.89, 12.23, and
16.25 degrees; (ii) said co-crystal is a carbamazepine :trimesic acid co-crystal and said
X-ray diffraction patteni comprises peaks at 10.89, 17.05, and
18.47 degrees; (iii) said co-crystal is a carbamazepine:trimesic acid co-crystal and said
X-ray diffraction pattern comprises peaks at 12.23 and 17.05 degrees; (iv) said co-crystal is a carbamazepine:trimesic acid co-crystal and said
X-ray diffraction pattern comprises peaks at 10.89 and 21.95 degrees; (v) said co-crystal is a carbamazepine:trimesic acid co-crystal and said
X-ray diffraction pattern comprises a peak at 10.89 degrees; or (vi) said co-crystal is a carbamazepine:trimesic acid co-crystal and said
X-ray diffraction pattern comprises a peak at 16.25 degrees; or
(b) the co-crystal is characterized by a DSC thermogram, wherein said co- crystal is a carbamazepine:trimesic acid co-crystal and said DSC thermogram comprises an endothermic transition at about 273 degrees C.
85. The co-crystal of claim 1, specifically excluding a co-crystal selected from the group consisting of: nabumetone:2,3-naphthalenediol, fluoxetine HChbenzoic acid, fluoxetine HCksuccinic acid, acetaminophe piperazine, acetaminophemtheophylline, theophylline: salicylic acid, theophylline:p-hydroxybenzoic acid, theophylline: sorbic acid, theophylline: l-hydroxy-2-naphthoic acid, theophylline:glycolic acid, theophylline:2,5- dihydroxybenzoic acid, theophyllinexhloroacetic.acid, bis(diphenylhydantoin):9- ethyladenine acetylacetone solvate, bis(diphenylhydantoin):9-ethyladenine 2,4- pentanedione solvate, 5,5-diphenylbarbituric acid:9-ethyladenine, bis(diphenylhydantoin) : 9-ethyladenine, 4-aminobenzoic acid:4-aminobenzonitrile, sulfadimidine: salicylic acid, 8-hydroxyquinolinium 4-mtrobenzoate:4-nitrobenzoic acid, sulfaproxyline : caffeine, retro-inverso-isopropyl (2R,3 S)-4-cyclohexyl-2-hydroxy-3 -(N- ((2R)-2-morpholinocarbonyhnethyl-3-(l-naphthyl)propionyl)-L- histidylamino)butyrate: cinnamic acid monohydrate, benzoic acid sonicotinamide, 3-(2- N',N'-(dimethylhydrazino)-4-thiazolylmethylthio)-N"-sulfamoylpropionamidine:maleic acid, diglycine hydrochloride (C2H5NO2:C2H6NO +Cl"), octadecanoic acid:3- pyridinecarboxamide, cis-N-(3-methyl-l-(2-(l,2,3,4-tetrahydro)naphthyl)-piperidin-4- yl)-N-phenylpropanamide hydrochloride:oxalic acid, trans-N-(3-methyl-l-(2-(l,2,3,4- tetrahydro)naphthyl)-piperidin-4-ylium)-N-phenylpropanamide oxalate:oxalic acid dihydrate, bis(l-(3-((4-(2-isopropoxyphenyl)-l-piperazinyl)methyl)benzoyl)piperidine) succinate: succinic acid, bis(p-cyanophenyl)imidazolylmethane:succinic acid, cis-l-((4- (l-imidazolylmethyl)cyclohexyl)methyl)imidazole:succinic acid, (+)-2-(5,6-dimethoxy- l,2,3,4-tetrahydro-l-naphthyl)imidazoline:(+)-dibenzoyl-D-tartaric acid, raclopride:tartaric acid, 2,6-diamino-9-ethylpurine:5,5-diethylbarbituric acid, 5,5- diethylbarbituric acid:bis(2-aminopyridine), 5,5-diethylbarbituric acid:acetamide, 5,5- diethylbarbituric acid:KI3, 5,5-diethylbarbituric acid:urea, bis(barbital):hexamethylphosphoramide, 5,5-diethylbarbituric acid:imidazole, barbitahl- methylimidazole, 5,5-diethylbarbituric acid:N-methyl-2-pyridone, 2,4-diamino-5-(3,4,5- trimethoxybenzyl)-pyrimidine:5,5-diethylbarbituric acid, bis(barbital): caffeine, bis(barbital) : 1 -methylimidazole, bis(beta-cyclodextrin) :bis(barbital) hydrate, tefrakis(beta-cyclodextrin):tetrakis(barbital), 9-ethyladenine:5,5-diethylbarbituric acid, barbital:N'-(f-cyanophenyl)-N-(p-iodophenyl)melamine, barbital:2-amino-4-(7n- bromophenylamino)-6-chloro-l ,3,5-triazine, 5,5-diethylbarbituric acid:N,N'- diphenylmelamine, 5,5-diethylbarbituric acid:N,N'-bis(p-chlorophenyl)melamine, N,N'- bis(J9-bromophenyl)melamine:5,5-diethylbarbituric acid, 5,5-diethylbarbituric acid:N,N'- bis(p-iodophenyl)melamine, 5,5-diethylbarbituric acid:N,N'-bis(p-tolyl)melamine, 5,5- diethylbarbituric acid:N,N'-bis(m-tolyl)melamine, 5,5-diethylbarbituric acid:N,N'-bis(m- chlorophenyl)rnelarnine, N,N'-Bis(z«-methylphenyl)melamine:barbital, N,N'-bis(m- chlorophenyl)melarnine:barbital tetrahydrofuran solvate, 5,5-diethylbarbituric acid:N,N'- bis(tert-butyl)melamine, 5,5-diethylbarbituric acid:N,N'-di(tert-butyl)melamine, 6,6'- diquinolyl ether:5,5-diethylbarbituric acid, 5-tert-butyl-2,4,6- triaminopyrimidine: diethylbarbituric acid, N,N'-bis(4- carboxymethylphenyl)melamine:barbital ethanol solvate, N,N'-bis(4-tert- butylphenyl)melamine:barbital, tris(5,17-N,N'-bis(4-amino-6-(butylamino)-l,3,5-triazin- 2-yl)diamino- 11 ,23-dinitro-25,26,27,28- tetrapropoxycalix(4)arene):hexakis(diethylbarbituric acid) toluene solvate, N,N'-bis(m- fluorophenyl)melamine:barbital, N,N'-bis(m-bromophenyl)melamine:barbital acetone solvate, N,N'-bis(m-iodophenyl)melamine:barbital acetonitrile solvate, N,N'-bis(m- trifluoromethylphenyl)melamine:barbital acetonitrile solvate, aminopyrine:barbital, N,N'-bis(4-fluorophenyl)melamine:barbital, N,N'-bis(4- trifluoromethylphenyl)melamine:barbital, 2,4-diamino-5-(3,4,5- trimethoxybenzyl)pyrimidine:barbital, hydroxybutyrate:hydroxyvalerate, 2- aminopyrimidine:succinic acid, l,3-bis(((6-methylpyrid-2- yl)amino)carbonyl)benzene:glutaric acid, 5-tert-butyl-2,4,6- triaminopyrimidine:diethylbarbituric acid, bis(dithiobiuret-S,S ')nickel(II) :diuracil, platinum 3,3 '-dihydroxymethyl-2,2' -bipyridine dichloride:AgF3CSO3, 4,4'- bipyridyhisophthalic acid, 4,4'-bipyridyl:l,4-naphthalenedicarboxylic acid, 4,4'- bipyridyl:l,3,5-cyclohexane-tricarboxylic acid, 4,4'-bipyridyl:tricaballylic acid, urotropin:azelaic acid, insulin:C8-HI (octanoyl-Ne-LysB29-human insulin), isonicotinamide: cinnamic acid, isonicotinamide:3-hydroxybenzoic acid, isonicotinamide:3-N,N-dimethylaminobenzoic acid, isonicotinamide:3,5- bis(trifluoromethyl)-benzoic acid, isonicotinamide:d,l-mandelic acid, isonicotinamide hloroacetic acid, isonicotinamide: fumaric acid monoethyl ester, isonicotinamide: 12-bromododecanoic acid, isonicotinamide: fumaric acid, isonicotinamide: succinic acid, isonicotinamide:4-ketopimelic acid, isonicotinamide:thiodiglycolic acid, 1,3,5-cyclohexane-tricarboxylic acid:hexamethyltetramine, 1,3,5-cyclohexane-tricarboxylic acid:4,7-phenanthroline, 4,7- phenanthroline: oxalic acid, 4,7-phenantl roline:terephthalic acid, 4,7-phenanthroline: 1 ,3,5-cyclohexane-tricarboxylic acid, 4,7-phenanthroline: 1 ,4-naphthalenedicarboxylic acid, pyrazine:methanoic acid, pyrazine:ethanoic acid, pyrazine:propanoic acid, pyrazine:butanoic acid, pyrazine:pentanoic acid, pyrazine:hexanoic acid, pyrazine:heptanoic acid, pyrazine:octanoic acid, pyrazine onanoic acid, pyrazine:decanoic acid, diammine-(deoxy-quanyl-quanyl-N7,N7)-platinum:tris(glycine) hydrate, 2-aminopyrimidine:p-phenylenediacetic acid, bis(2-aminopyrimidin-l- ium)fumarate:fumaric acid, 2-aminopyrimidine:indole-3-acetic acid, 2- aminopyrimidine:N-methylpyrrole-2-carboxylic acid, 2-aminopyrimidine:thiophen-2- carboxylic acid, 2-aminopyrimidine:(+)-camphoric acid, 2,4,6-Trinitrobenzoic acid:2- aminopyrimidine, 2-aminopyrimidine:4-aminobenzoic acid, 2- aminopyrimidine:bis(phenoxyacetic acid), 2-aminopyrimidine:(2,4- dichlorophenoxy)acetic acid, 2-aminopyrimidine:(3,4-dichlorophenoxy)acetic acid, 2- aminopyrimidine:indole-2-carboxylic acid, 2-aminopyrimidine:terephthalic acid, 2- aminopyrimidine:bis(2-nitrobenzoic acid), 2-aminopyrimidine:bis(2-aminobenzoic acid), 2-aminopyrimidine:3-aminobenzoic acid, 2-hexeneoic acid:isonicotinamide, 4- nitrobenzoic acid: isonicotinamide, 3,5-dinitrobenzoic acid:isonicotinamide:4- methylbenzoic acid, 2-amino-5-nitropyrimidine:2-amino-3-nitropyridine, 3,5- dinitrobenzoic acid:4-chlorobenzamide, 3-dimethylaminobenzoic acid:4- chlorobenzamide, fumaric acid:4-chlorobenzamide, oxine:4-nitrobenzoic acid, oxine:3,5- dinitrobenzoic acid, oxine:3,5-dinitrosalicylic acid, 3-[2-(N',N'-dimethylhydrazino)-4- thiazolylmethylthio]-N -sulfamoylpropionamidine:maleic acid, 5-fluorouracil:9- ethylhypoxanthine, 5-fluorouracil:cytosine dihydrate, 5-fluorouracil:theophylline monohydrate, stearic acid:nicotinamide, cis-l-{[4-(l- imidazolyhnethyl)cyclohexyl]methyl}imidazole:succinic acid, CGS18320B:succinic acid, sulfaproxyline: caffeine, 4-aminobenzoic acid:4-aminobenzonitrile, 3,5- dinitrobenzoic acid:isonicotinamide:3-methylbenzoic acid, 3,5-dinitrobenzoic acid:isonicotinamide:4-(dimethylamino)benzoic acid, 3,5-dinitrobenzoic acid:isonicotinamide:4-hydroxy-3-methoxycinnamic acid, isonicotinamide: oxalic acid, isonicotinamidemalonic acid, isonicotinamide :succinic acid, isonicotinamide :glutaric acid, isonicotinamide :adipic acid, benzoic acid:isonicotinamide, mazapertine:succinate, betaine:dichloromtrophenol, betainepyridine:dichloronitrophenol, betainepyridine:pentachlorophenol, 4- {2-[ 1 -(2-hydroxyethyl)-4-pyridylidene]- ethylidene}-cyclo-hexa-2,5-dien-l-one:methyl 2,4-dihydroxybenzoate, 4-{2-[l-(2- hydroxyethyl)-4-pyridylidene]-ethylidene}-cyclo-hexa-2,5-dien-l-one:2,4- dihydroxypropiophenone, 4- {2-[ 1 -(2-hydroxyethyl)-4-pyridylidene]-ethylidene} -cyclo- hexa-2,5-dien-l-one:2,4-dihydroxyacetophenone, squaric acid:4,4'-dipyridylacetylene, squaric acid:l,2-bis(4-pyridyl)ethylene, chloranilic acid:l,4-bis[(4- pyridyl)ethynyl]benzene, 4,4' -bipyridine :phthalic acid, 4,4'-dipyridylacetylene:phthalic acid, bis(pentamethylcyclopentadienyl)iron:bromanilic acid, bis(pentamethylcyclopentadienyl)iron: chloranilic acid, bis(pentamethylcyclopentadienyl)iron:cyananilic acid, pyrazinotetrathiafulvalene:chloranilic acid, phenohpentafluorophenol, co-crystals of itraconazole, and co-crystals of topiramate.
PCT/US2004/006288 2002-02-15 2004-02-26 Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen WO2004078163A2 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
EP04715190A EP1631260A2 (en) 2003-02-28 2004-02-26 Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
JP2006508979A JP2007524596A (en) 2003-02-28 2004-02-26 Co-crystal pharmaceutical composition
CA002514733A CA2514733A1 (en) 2003-02-28 2004-02-26 Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US10/546,963 US20070059356A1 (en) 2002-05-31 2004-02-26 Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US10/551,014 US20060223794A1 (en) 2002-02-15 2004-03-31 Novel olanzapine forms and related methods of treatment
PCT/US2004/009947 WO2004089313A2 (en) 2003-04-01 2004-03-31 Novel olanzapine forms and related methods of treatment
US10/926,842 US7446107B2 (en) 2002-02-15 2004-08-26 Crystalline forms of conazoles and methods of making and using the same
PCT/US2004/028456 WO2005092884A1 (en) 2004-02-26 2004-09-01 Novel crystalline forms of conazoles and methods of making and using the same
EP04782868A EP1718640A4 (en) 2004-02-26 2004-09-01 Novel crystalline forms of conazoles and methods of making and using the same
JP2007500742A JP2007525502A (en) 2004-02-26 2004-09-01 Novel crystalline forms of conazoles and methods for their production and use
EP20040783308 EP1670753A4 (en) 2003-09-04 2004-09-04 Modafinil compositions
JP2006525508A JP4842819B2 (en) 2003-09-04 2004-09-04 Modafinil composition
IL173575A IL173575A0 (en) 2003-09-04 2006-02-07 Modafinil compositions
NO20060669A NO20060669L (en) 2003-09-11 2006-02-10 Modafinilsammensetninger
US12/234,420 US20090088443A1 (en) 2002-02-15 2008-09-19 Novel crystalline forms of conazoles and methods of making and using the same
IL199140A IL199140A (en) 2003-09-04 2009-06-03 Modafinil compositions
US12/792,415 US20100311701A1 (en) 2002-02-15 2010-06-02 Pharmaceutical Co-Crystal Compositions

Applications Claiming Priority (26)

Application Number Priority Date Filing Date Title
US45121303P 2003-02-28 2003-02-28
US60/451,213 2003-02-28
PCT/US2003/006662 WO2003074474A2 (en) 2002-03-01 2003-03-03 Multiple-component solid phases containing at least one active pharmaceutical ingredient
USPCT/US03/06662 2003-03-03
US45602703P 2003-03-18 2003-03-18
US60/456,027 2003-03-18
US46396203P 2003-04-18 2003-04-18
US60/463,962 2003-04-18
US10/449,307 2003-05-30
US10/449,307 US7078526B2 (en) 2002-05-31 2003-05-30 CIS-itraconazole crystalline forms and related processes, pharmaceutical compositions and methods
USPCT/US03/19574 2003-06-20
US10/601,092 US20050025791A1 (en) 2002-06-21 2003-06-20 Pharmaceutical compositions with improved dissolution
PCT/US2003/019574 WO2004000284A1 (en) 2002-06-21 2003-06-20 Pharmaceutical compositions with improved dissolution
US10/601,092 2003-06-20
US48706403P 2003-07-11 2003-07-11
US60/487,064 2003-07-11
USPCT/US03/27772 2003-09-04
PCT/US2003/027772 WO2004078161A1 (en) 2003-02-28 2003-09-04 Pharmaceutical co-crystal compositions of drugs such as carbamazeptine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US10/660,202 2003-09-11
US10/660,202 US7927613B2 (en) 2002-02-15 2003-09-11 Pharmaceutical co-crystal compositions
US50820803P 2003-10-02 2003-10-02
US60/508,208 2003-10-02
PCT/US2003/041273 WO2004061433A1 (en) 2002-12-30 2003-12-24 Pharmaceutical compositions with improved dissolution
USPCT/US03/41273 2003-12-24
US54275204P 2004-02-06 2004-02-06
US60/542,752 2004-02-06

Related Parent Applications (6)

Application Number Title Priority Date Filing Date
PCT/US2003/006662 Continuation-In-Part WO2003074474A2 (en) 2002-02-15 2003-03-03 Multiple-component solid phases containing at least one active pharmaceutical ingredient
US10/449,307 Continuation-In-Part US7078526B2 (en) 2002-02-15 2003-05-30 CIS-itraconazole crystalline forms and related processes, pharmaceutical compositions and methods
US10/601,092 Continuation-In-Part US20050025791A1 (en) 2002-02-15 2003-06-20 Pharmaceutical compositions with improved dissolution
US10/637,829 Continuation-In-Part US20040053853A1 (en) 2002-02-15 2003-08-08 Topiramate salts and compositions comprising and methods of making and using the same
PCT/US2003/027772 Continuation-In-Part WO2004078161A1 (en) 2002-02-15 2003-09-04 Pharmaceutical co-crystal compositions of drugs such as carbamazeptine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US10/660,202 Continuation-In-Part US7927613B2 (en) 2002-02-15 2003-09-11 Pharmaceutical co-crystal compositions

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US10/601,092 Continuation-In-Part US20050025791A1 (en) 2002-02-15 2003-06-20 Pharmaceutical compositions with improved dissolution
US10/926,842 Continuation-In-Part US7446107B2 (en) 2002-02-15 2004-08-26 Crystalline forms of conazoles and methods of making and using the same
US12/792,415 Continuation US20100311701A1 (en) 2002-02-15 2010-06-02 Pharmaceutical Co-Crystal Compositions

Publications (2)

Publication Number Publication Date
WO2004078163A2 true WO2004078163A2 (en) 2004-09-16
WO2004078163A3 WO2004078163A3 (en) 2005-01-20

Family

ID=56290526

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/006288 WO2004078163A2 (en) 2002-02-15 2004-02-26 Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen

Country Status (4)

Country Link
EP (1) EP1631260A2 (en)
JP (1) JP2007524596A (en)
CA (1) CA2514733A1 (en)
WO (1) WO2004078163A2 (en)

Cited By (384)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1511490A2 (en) * 2002-05-31 2005-03-09 Transform Pharmaceuticals, Inc. Novel conazole crystalline forms and related processes, pharmaceutical compositions and methods
EP1670753A2 (en) * 2003-09-04 2006-06-21 Cephalon, Inc. Modafinil compositions
WO2006103011A1 (en) * 2005-03-30 2006-10-05 Aicuris Gmbh & Co. Kg Pharmaceutical preparation of n -[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-n-methyl-2-[4-(2-pyridinyl)phenyl] acetamide
WO2006138421A2 (en) * 2005-06-15 2006-12-28 Elan Pharma International Limited Nanoparticulate azelnidipine formulations
WO2008054609A2 (en) * 2006-10-04 2008-05-08 The Regents Of The University Of Michigan Dissolution and precipitation of cocrystals with ionizable components
WO2008063284A2 (en) * 2006-10-10 2008-05-29 Janssen Pharmaceutica Nv Novel crystal of (s)-(+)-2-(2-chlorophenyl)-2-hydroxy-ethyl carbamate
EP1951393A2 (en) * 2005-10-31 2008-08-06 The Regents of the University of Michigan Reaction co-crystallization of molecular complexes or co-crystals
EP1962600A2 (en) * 2005-12-08 2008-09-03 SSCI, Inc. Metronidazole cocrystals and imipramine cocrystals
WO2008108639A1 (en) * 2007-03-08 2008-09-12 Avantium Holding B.V. Co-crystalline forms of carbamazepine
WO2008153945A2 (en) * 2007-06-06 2008-12-18 University Of South Florida Nutraceutical co-crystal compositions
US7507823B2 (en) 2004-05-06 2009-03-24 Bristol-Myers Squibb Company Process of making aripiprazole particles
WO2009094155A1 (en) * 2008-01-22 2009-07-30 Thar Pharmaceuticals In vivo studies of crystalline forms of meloxicam
WO2009140466A2 (en) * 2008-05-14 2009-11-19 Dr. Reddy's Laboratories Ltd. Linezolid co-crystals
EP2123626A1 (en) * 2008-05-21 2009-11-25 Laboratorios del Dr. Esteve S.A. Co-crystals of duloxetine and co-crystal formers for the treatment of pain
WO2009152347A2 (en) * 2008-06-13 2009-12-17 Bionevia Pharmaceuticals Inc. Crystalline forms of zotepine hydrochloride
WO2010043412A1 (en) * 2008-10-17 2010-04-22 Laboratorios Del Dr. Esteve, S.A. Co-crystals of tramadol and nsaids
WO2010017343A3 (en) * 2008-08-06 2010-05-14 Bionevia Pharmaceuticals, Inc. Flupirtine hydrochloride maleic acid cocrystal
EP2199274A1 (en) * 2008-12-16 2010-06-23 Laboratorios Del. Dr. Esteve, S.A. Co-crystals of tramadol and paracetamol
WO2010090491A2 (en) * 2009-02-09 2010-08-12 주식회사 한독약품 Novel salts of adefovir dipivoxil and preparation method thereof
US7790905B2 (en) 2002-02-15 2010-09-07 Mcneil-Ppc, Inc. Pharmaceutical propylene glycol solvate compositions
WO2010115981A1 (en) 2009-04-10 2010-10-14 Novartis Ag 7-azadispiro [3.0.4.1] decane-8-carboxamides as hepatitis c virus inhibitors
WO2010116248A1 (en) 2009-04-10 2010-10-14 Novartis Ag Organic compounds and their uses
WO2010130773A2 (en) 2009-05-15 2010-11-18 Novartis Ag Benzoxazolone derivatives as aldosterone symthase inhibitors
WO2010130796A1 (en) 2009-05-15 2010-11-18 Novartis Ag Aryl pyridine as aldosterone synthase inhibitors
WO2010136493A1 (en) 2009-05-28 2010-12-02 Novartis Ag Substituted aminopropionic derivatives as neprilysin inhibitors
EP2262488A1 (en) * 2008-04-07 2010-12-22 Mutual Pharmaceutical Company, Inc. Colchicine solid complex; methods of making; and methods of use thereof
WO2011009943A1 (en) 2009-07-24 2011-01-27 Novartis Ag Oxazine derivatives and their use as bace inhibitors for the treatment of neurological disorders
WO2011009852A2 (en) 2009-07-21 2011-01-27 Novartis Ag Pharmaceutical compositions
US7883713B2 (en) 2001-06-22 2011-02-08 Aicuris Gmbh & Co. Kg Topical application of thiazolyl amides
EP2281558A1 (en) * 2009-08-06 2011-02-09 Laboratorios Del. Dr. Esteve, S.A. Pharmaceutical compounds of O-Desmethyl-Tramadol and COX-inhibitors
WO2011015652A1 (en) 2009-08-07 2011-02-10 Novartis Ag 3-heteroarylmethyl-imidazo[1,2-b]pyridazin-6-yl derivatives as c-met tyrosine kinase modulators
WO2011018454A1 (en) 2009-08-12 2011-02-17 Novartis Ag Heterocyclic hydrazone compounds and their uses to treat cancer and inflammation
WO2011020861A1 (en) 2009-08-20 2011-02-24 Novartis Ag Heterocyclic oxime compounds
WO2011023677A1 (en) 2009-08-26 2011-03-03 Novartis Ag Tetra-substituted heteroaryl compounds and their use as mdm2 and/or mdm4 modulators
WO2011026911A1 (en) 2009-09-04 2011-03-10 Novartis Ag Bipyridines useful for the treatment of proliferative diseases
WO2011026917A1 (en) 2009-09-04 2011-03-10 Novartis Ag Heteroaryl compounds as kinase inhibitors
WO2011026904A1 (en) 2009-09-04 2011-03-10 Novartis Ag Pyrazinylpyridines useful for the treatment of proliferative diseases
WO2011029842A1 (en) 2009-09-10 2011-03-17 Novartis Ag Sulfonamides as inhibitors of bcl-2 family proteins for the treatment of cancer
WO2011029915A1 (en) 2009-09-10 2011-03-17 Novartis Ag Ether derivatives of bicyclic heteroaryls
US7927613B2 (en) 2002-02-15 2011-04-19 University Of South Florida Pharmaceutical co-crystal compositions
WO2011054828A1 (en) 2009-11-04 2011-05-12 Novartis Ag Heterocyclic sulfonamide derivatives useful as mek inhibitors
EP2325172A1 (en) * 2009-11-02 2011-05-25 Laboratorios Del. Dr. Esteve, S.A. Co-crystals of celecoxib and L-proline
WO2011061168A1 (en) 2009-11-17 2011-05-26 Novartis Ag Aryl-pyridine derivatives as aldosterone synthase inhibitors
WO2011061271A1 (en) 2009-11-20 2011-05-26 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel nep inhibitors
WO2011064376A1 (en) 2009-11-30 2011-06-03 Novartis Ag Imidazole derivatives as aldosterone synthase inhibitors
WO2011073316A1 (en) 2009-12-18 2011-06-23 Novartis Ag 4-aryl-butane-1,3-diamides
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
WO2011076747A1 (en) 2009-12-21 2011-06-30 Novartis Ag Diaza-spiro[5.5]undecanes as orexin receptor antagonists
WO2011076744A1 (en) 2009-12-21 2011-06-30 Novartis Ag Disubstituted heteroaryl-fused pyridines
WO2011080176A1 (en) 2009-12-31 2011-07-07 Novartis Ag Pyrazine derivatives and their use in the treatment of neurological disorders
WO2011092290A1 (en) 2010-02-01 2011-08-04 Novartis Ag Pyrazolo[5,1b]oxazole derivatives as crf-1 receptor antagonists
WO2011092293A2 (en) 2010-02-01 2011-08-04 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
WO2011095452A1 (en) 2010-02-02 2011-08-11 Novartis Ag Aryl benzylamine compounds
WO2011095450A1 (en) 2010-02-02 2011-08-11 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
WO2011104266A1 (en) 2010-02-25 2011-09-01 Novartis Ag Dimeric iap inhibitors
WO2011113894A1 (en) 2010-03-19 2011-09-22 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf
WO2011131709A1 (en) 2010-04-21 2011-10-27 Novartis Ag Furopyridine compounds and uses thereof
WO2011144666A1 (en) 2010-05-20 2011-11-24 Novartis Ag 2,4-dioxo-1,4-dihydro-2h-quinazolin-3-yl-sulfonamide derivatives
WO2011157787A1 (en) 2010-06-17 2011-12-22 Novartis Ag Biphenyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2011157793A1 (en) 2010-06-17 2011-12-22 Novartis Ag Piperidinyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2011097372A3 (en) * 2010-02-03 2011-12-29 Aptuit Laurus Private Limited Pterostilbene cocrystals
WO2012003189A1 (en) 2010-06-29 2012-01-05 Irm Llc Compositions and methods for modulating the wnt signaling pathway
WO2012004299A1 (en) 2010-07-06 2012-01-12 Novartis Ag Tetrahydro-pyrido-pyrimidine derivatives
WO2012007539A1 (en) 2010-07-14 2012-01-19 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2012006953A1 (en) 2010-07-13 2012-01-19 Novartis Ag Oxazine derivatives and their use in the treatment of neurological disorders
WO2012010663A1 (en) 2010-07-22 2012-01-26 Novartis Ag 2,3,5-trisubstituted thiophene compounds and uses thereof
WO2012017020A1 (en) 2010-08-04 2012-02-09 Novartis Ag N-((6-amino-pyridin-3-yl)methyl)-heteroaryl-carboxamides as inhibitors of plasma kallikrein
WO2011158110A3 (en) * 2010-04-28 2012-03-15 Nuformix Limited Cilostazol cocrystals and compositions
WO2012035023A1 (en) 2010-09-13 2012-03-22 Novartis Ag Triazine-oxadiazoles
WO2012035158A1 (en) 2010-09-17 2012-03-22 Novartis Ag Pyrazine derivatives as enac blockers
EP2432320A1 (en) * 2009-05-20 2012-03-28 Nutracryst Therapeutics Private Limited Pharmaceutical co-crystals of quercetin
WO2012040230A1 (en) 2010-09-20 2012-03-29 Envivo Pharmaceuticals, Inc. Imidazotriazinone compounds
WO2012048235A1 (en) 2010-10-08 2012-04-12 Novartis Ag Vitamin e formulations of sulfamide ns3 inhibitors
WO2012055888A1 (en) 2010-10-26 2012-05-03 Novartis Ag Diaza-spiro[5.5]undecanes useful as orexin receptor antagonists
US8183290B2 (en) 2002-12-30 2012-05-22 Mcneil-Ppc, Inc. Pharmaceutically acceptable propylene glycol solvate of naproxen
WO2012065958A1 (en) 2010-11-16 2012-05-24 Novartis Ag Method of treating contrast-induced nephropathy
WO2012065953A1 (en) 2010-11-16 2012-05-24 Novartis Ag Substituted carbamoylcycloalkyl acetic acid derivatives as nep inhibitors
WO2012065956A1 (en) 2010-11-16 2012-05-24 Novartis Ag Substituted amino bisphenyl pentanoic acid derivatives as nep inhibitors
WO2012080271A1 (en) 2010-12-13 2012-06-21 Novartis Ag Dimeric iap inhibitors
WO2012080260A1 (en) 2010-12-13 2012-06-21 Novartis Ag Dimeric iap inhibitors
WO2012087520A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2012087521A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2012084873A1 (en) 2010-12-20 2012-06-28 Novartis Ag 4- (hetero) aryl - ethynyl - octahydro - indole - 1 - carboxylic acid esters
WO2012087519A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating fxr
WO2012093101A1 (en) 2011-01-04 2012-07-12 Novartis Ag Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration (amd)
WO2012095463A1 (en) 2011-01-12 2012-07-19 Novartis Ag Oxazine derivatives and their use in the treatment of neurological disorders
WO2012095521A1 (en) 2011-01-13 2012-07-19 Novartis Ag Bace-2 inhibitors for the treatment of metabolic disorders
WO2012095469A1 (en) 2011-01-13 2012-07-19 Novartis Ag Novel heterocyclic derivatives and their use in the treatment of neurological disorders
WO2012098501A1 (en) * 2011-01-21 2012-07-26 Ranbaxy Laboratories Limited Febuxostat co-crystals
WO2012101058A1 (en) 2011-01-24 2012-08-02 Novartis Ag 4-tolyl-ethynyl-octahydro-indole-1-ester derivatives
WO2012101063A1 (en) 2011-01-28 2012-08-02 Novartis Ag N-acyl pyridine biaryl compounds and their uses
WO2012101487A1 (en) 2010-12-22 2012-08-02 Novartis Ag Di/tri-aza-spiro-c9-c11alkanes
WO2012101065A2 (en) 2011-01-28 2012-08-02 Novartis Ag Pyrimidine biaryl amine compounds and their uses
WO2012101064A1 (en) 2011-01-28 2012-08-02 Novartis Ag N-acyl pyrimidine biaryl compounds as protein kinase inhibitors
WO2012101066A1 (en) 2011-01-28 2012-08-02 Novartis Ag Pyridine biaryl amine compounds and their uses
WO2012104823A2 (en) 2011-02-04 2012-08-09 Novartis Ag Pyridopyrimidinone compounds in the treatment of neurodegenerative diseases
WO2012104776A1 (en) 2011-01-31 2012-08-09 Novartis Ag Novel heterocyclic derivatives
WO2012107500A1 (en) 2011-02-10 2012-08-16 Novartis Ag [1, 2, 4] triazolo [4, 3 -b] pyridazine compounds as inhibitors of the c-met tyrosine kinase
WO2012116217A1 (en) 2011-02-25 2012-08-30 Irm Llc Compounds and compositions as trk inhibitors
US8258155B2 (en) 2008-06-30 2012-09-04 Mutual Pharmaceutical Company, Inc. Quinine sulfate/bisulfate solid complex; methods of making; and methods of use thereof
WO2012117224A1 (en) 2011-03-02 2012-09-07 Sabrepharm Limited Dipyridinium derivatives
WO2012120469A1 (en) 2011-03-08 2012-09-13 Novartis Ag Fluorophenyl bicyclic heteroaryl compounds
WO2012131633A1 (en) 2011-04-01 2012-10-04 Novartis Ag Pyrazolo pyrimidine derivatives
WO2012138648A1 (en) 2011-04-06 2012-10-11 Irm Llc Compositions and methods for modulating lpa receptors
WO2012140597A1 (en) 2011-04-14 2012-10-18 Novartis Ag Glycoside derivatives and their uses for the treatment of diabetes
WO2012140596A1 (en) 2011-04-14 2012-10-18 Novartis Ag Glycoside derivatives and uses thereof
US8298576B2 (en) 2006-11-17 2012-10-30 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
WO2012146371A1 (en) * 2011-04-28 2012-11-01 Zentiva, K.S. Pharmaceutically acceptable cocrystals of n-[2-(7-methoxy-1-naphtyl)ethyl]acetamide and methods of their preparation
WO2012156936A1 (en) 2011-05-17 2012-11-22 Novartis Ag Substituted indole derivatives for the treatment of immunological disorders
WO2012129568A3 (en) * 2011-03-24 2012-11-22 University Of South Florida Lithium compositions
WO2012164473A1 (en) 2011-05-27 2012-12-06 Novartis Ag 3-spirocyclic piperidine derivatives as ghrelin receptor agonists
WO2012175520A1 (en) 2011-06-20 2012-12-27 Novartis Ag Hydroxy substituted isoquinolinone derivatives
WO2012175487A1 (en) 2011-06-20 2012-12-27 Novartis Ag Cyclohexyl isoquinolinone compounds
WO2013008162A1 (en) 2011-07-08 2013-01-17 Novartis Ag Novel trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
WO2013008095A1 (en) 2011-07-08 2013-01-17 Novartis Ag Novel pyrrolo pyrimidine derivatives
WO2013008164A2 (en) 2011-07-08 2013-01-17 Novartis Ag Method of treating atherosclerosis in high triglyceride subjects
WO2013016197A1 (en) 2011-07-22 2013-01-31 Novartis Ag Tetrahydropyrido-pyridine and tetrahydropyrido-pyrimidine compounds and use thereof as c5a receptor modulators
WO2013014627A1 (en) 2011-07-27 2013-01-31 Novartis Ag Pyrazoline derivatives and their use as selective androgen receptor modulators
WO2013027188A1 (en) 2011-08-25 2013-02-28 Novartis Ag 2 -amino-4 - (pyridin- 2 -yl) - 5, 6 -dihydro-4h- 1, 3 -oxazine derivatives and their use as bace-1 and/or bace - 2 inhibitors
WO2013033620A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as pdgfr kinase inhibitors
WO2013030802A1 (en) 2011-09-01 2013-03-07 Novartis Ag Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
WO2013033203A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013033167A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013033070A1 (en) 2011-09-01 2013-03-07 Irm Llc COMPOUNDS AND COMPOSITIONS AS c-KIT KINASE INHIBITORS
WO2013033116A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013035047A1 (en) 2011-09-06 2013-03-14 Novartis Ag Benzothiazolone compound
US8399023B2 (en) 2009-07-31 2013-03-19 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
WO2013038381A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine/pyrazine amide derivatives
WO2013038373A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038378A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038386A1 (en) 2011-09-16 2013-03-21 Novartis Ag Heterocyclic compounds for the treatment of cystic fibrosis
WO2013038362A1 (en) 2011-09-15 2013-03-21 Novartis Ag 6 - substituted 3 - (quinolin- 6 - ylthio) - [1,2,4] triazolo [4, 3 -a] pyradines as tyrosine kinase
WO2013038390A1 (en) 2011-09-16 2013-03-21 Novartis Ag N-substituted heterocyclyl carboxamides
WO2013050987A1 (en) 2011-10-08 2013-04-11 Novartis Ag Carbamate/ urea derivatives containing piperidin and piperazin rings as h3 receptor inhibitors
WO2013054291A1 (en) 2011-10-13 2013-04-18 Novartis Ag Novel oxazine derivatives and their use in the treatment of disease
WO2013057711A1 (en) 2011-10-21 2013-04-25 Novartis Ag Quinazoline derivatives as pi3k modulators
WO2013057169A1 (en) 2011-10-19 2013-04-25 Universiteit Gent Pharmaceutical nanosuspension
WO2013061305A1 (en) 2011-10-28 2013-05-02 Novartis Ag Novel purine derivatives and their use in the treatment of disease
WO2013080120A1 (en) 2011-11-28 2013-06-06 Novartis Ag Novel trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
WO2013080141A1 (en) 2011-11-29 2013-06-06 Novartis Ag Pyrazolopyrrolidine compounds
WO2013093850A1 (en) 2011-12-22 2013-06-27 Novartis Ag Quinoline derivatives
WO2013093849A1 (en) 2011-12-22 2013-06-27 Novartis Ag Dihydro-benzo-oxazine and dihydro-pyrido-oxazine derivatives
WO2013105061A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused dihydropyrido [2,3 -b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105057A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused pyrroles as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105063A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105058A1 (en) 2012-01-13 2013-07-18 Novartis Ag 7,8- dihydropyrido [3, 4 - b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105065A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pah and related disorders
WO2013111108A1 (en) 2012-01-27 2013-08-01 Novartis Ag 5-membered heteroarylcarboxamide derivatives as plasma kallikrein inhibitors
WO2013111107A1 (en) 2012-01-27 2013-08-01 Novartis Ag Aminopyridine derivatives as plasma kallikrein inhibitors
US8507561B2 (en) 2009-01-22 2013-08-13 Absorption Pharmaceuticals, LLC Desensitizing drug product
US8519002B2 (en) 2008-04-07 2013-08-27 Takeda Pharmaceuticals U.S.A., Inc. Colchicine solid complex; methods of making; and methods of use thereof
WO2013128421A1 (en) 2012-03-02 2013-09-06 Novartis Ag Spirohydantoin compounds and their use as selective androgen receptor modulators
US8555875B2 (en) 2008-12-23 2013-10-15 Map Pharmaceuticals, Inc. Inhalation devices and related methods for administration of sedative hypnotic compounds
WO2013163508A1 (en) 2012-04-27 2013-10-31 Novartis Ag Tetrahydropyran dgat1 inhibitors
WO2013160873A1 (en) 2012-04-27 2013-10-31 Novartis Ag Cyclic bridgehead ether dgat1 inhibitors
WO2013164802A1 (en) 2012-05-04 2013-11-07 Novartis Ag Complement pathway modulators and uses thereof
WO2013164790A1 (en) 2012-05-03 2013-11-07 Novartis Ag L-malate salt of 2, 7 - diaza - spiro [4.5 ] dec- 7 - yle derivatives and crystalline forms thereof as ghrelin receptor agonists
WO2013171690A1 (en) 2012-05-16 2013-11-21 Novartis Ag Monocyclic heteroaryl cycloalkyldiamine derivatives
WO2013175417A1 (en) 2012-05-24 2013-11-28 Novartis Ag Pyrrolopyrrolidinone compounds
WO2013184757A1 (en) 2012-06-06 2013-12-12 Irm Llc Compounds and compositions for modulating egfr activity
WO2013184766A1 (en) 2012-06-06 2013-12-12 Irm Llc Compounds and compositions for modulating egfr activity
WO2013189910A1 (en) * 2012-06-22 2013-12-27 Basf Se Multicomponent crystals comprising imatinib mesilate and selected co-crystal formers
WO2013192345A1 (en) 2012-06-20 2013-12-27 Novartis Ag Complement pathway modulators and uses thereof
WO2014002051A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
WO2014002054A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014002057A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014002052A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014002058A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
WO2014002053A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014009833A2 (en) 2012-07-12 2014-01-16 Novartis Ag Complement pathway modulators and uses thereof
WO2014013469A1 (en) 2012-07-20 2014-01-23 Novartis Ag Carbamate/urea derivatives
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
WO2014028459A1 (en) 2012-08-13 2014-02-20 Novartis Ag 1,4-disubstituted pyridazine analogs and methods for treating smn-deficiency-related conditions
WO2014027300A1 (en) 2012-08-13 2014-02-20 Novartis Ag Bicyclic heteroaryl cycloalkyldiamine derivatives as inhibitors of spleen tyrosine kinases (syk)
WO2014033617A1 (en) 2012-08-31 2014-03-06 Novartis Ag 2'-ethynyl nucleoside derivatives for treatment of viral infections
WO2014033654A1 (en) 2012-08-30 2014-03-06 Novartis Ag Salts of benzothiazolone compound as beta-2-adrenoceptor agonist
WO2014033631A1 (en) 2012-08-31 2014-03-06 Novartis Ag N-(3-pyridyl) biarylamides as kinase inhibitors
WO2014033630A1 (en) 2012-08-31 2014-03-06 Novartis Ag Novel aminothiazole carboxamides as kinase inhibitors
WO2014037900A1 (en) 2012-09-07 2014-03-13 Novartis Ag Indole carboxamide derivatives and uses thereof
WO2014047020A1 (en) 2012-09-19 2014-03-27 Novartis Ag Dihydropyrrolidino-pyrimidines as kinase inhibitors
WO2014052619A1 (en) 2012-09-27 2014-04-03 Irm Llc Piperidine derivatives and compositions as modulators of gpr119 activity
WO2014049514A1 (en) 2012-09-25 2014-04-03 Novartis Ag Compounds for use in gastric complication
WO2014049540A2 (en) 2012-09-29 2014-04-03 Novartis Ag Cyclic peptides and use as medicines
US8697735B2 (en) 2008-07-25 2014-04-15 Bionevia Pharmaceuticals, Inc. Solid forms of epalrestat
WO2014072956A1 (en) 2012-11-12 2014-05-15 Novartis Ag Oxazolidin-2-one-pyrimidine derivatives
WO2014072911A1 (en) 2012-11-07 2014-05-15 Novartis Ag Substituted indole derivatives
WO2014078802A1 (en) 2012-11-19 2014-05-22 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014078813A1 (en) 2012-11-19 2014-05-22 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014082935A1 (en) 2012-11-30 2014-06-05 Novartis Ag Cyclic nucleoside derivatives and uses thereof
WO2014093606A1 (en) 2012-12-13 2014-06-19 Novartis Ag Pyridone derivatives and uses thereof in the treatment of tuberculosis
WO2014091446A1 (en) 2012-12-13 2014-06-19 Novartis Ag Pyrimido [4,5-b]quinoline-4,5 (3h,10h)-diones as nonsense mutation suppressors
WO2014097151A2 (en) 2012-12-19 2014-06-26 Novartis Ag Autotaxin inhibitors
WO2014099880A1 (en) 2012-12-19 2014-06-26 Novartis Ag Aryl-substituted fused bicyclic pyridazine compounds
WO2014097148A1 (en) 2012-12-19 2014-06-26 Novartis Ag Tricyclic compounds for inhibiting the cftr channel
WO2014097147A1 (en) 2012-12-19 2014-06-26 Novartis Ag Tricyclic compounds as cftr inhibitors
WO2014115077A1 (en) 2013-01-22 2014-07-31 Novartis Ag Substituted purinone compounds
WO2014116845A1 (en) 2013-01-23 2014-07-31 Novartis Ag Thiadiazole analogs thereof and methods for treating smn-deficiency-related-conditions
WO2014115080A1 (en) 2013-01-22 2014-07-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the p53/mdm2 interaction
WO2014126979A1 (en) 2013-02-14 2014-08-21 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as nep (neutral endopeptidase) inhibitors
WO2014125413A1 (en) 2013-02-13 2014-08-21 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2014128612A1 (en) 2013-02-20 2014-08-28 Novartis Ag Quinazolin-4-one derivatives
WO2014132205A1 (en) 2013-02-28 2014-09-04 Novartis Ag Formulation comprising benzothiazolone compound
WO2014143638A1 (en) 2013-03-14 2014-09-18 Novartis Ag 2-(1h-indol-4-ylmethyl)-3h-imidazo[4,5-b]pyridine-6-carbonitrile derivatives as complement factor b inhibitors useful for the treatment of ophthalmic diseases
WO2014151784A1 (en) 2013-03-15 2014-09-25 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014151616A1 (en) 2013-03-14 2014-09-25 Novartis Ag Biaryl amide compounds as kinase inhibitors
WO2014151729A1 (en) 2013-03-15 2014-09-25 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014160649A1 (en) 2013-03-29 2014-10-02 Novartis Ag Hydroxamic acid derivatives as lpxc inhibitors for the treatment of bacterial infections
US8859607B2 (en) 2007-02-09 2014-10-14 Basf Se Crystalline complexes of agriculturally active organic compounds
WO2014167528A1 (en) 2013-04-11 2014-10-16 Novartis Ag Spiropyrazolopyridine derivatives and uses thereof for the treatment of viral infections
WO2014178040A1 (en) 2013-04-29 2014-11-06 Mapi Pharma Ltd. Co-crystals of dapagliflozin
WO2014184778A1 (en) 2013-05-17 2014-11-20 Novartis Ag Pyrimidin-4-yl)oxy)-1h-indole-1-carboxamide derivatives and use thereof
WO2014191894A1 (en) 2013-05-27 2014-12-04 Novartis Ag Imidazopyrrolidinone derivatives and their use in the treatment of disease
WO2014191906A1 (en) 2013-05-28 2014-12-04 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
WO2014191911A1 (en) 2013-05-28 2014-12-04 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
WO2014191896A1 (en) 2013-05-27 2014-12-04 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
US8906948B2 (en) 2008-09-06 2014-12-09 Bionevia, LLC Choline cocrystal of epalrestat
WO2015008230A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors comprising a heteroaromatic ring-benzyl-amide-cycle core
WO2015009616A1 (en) 2013-07-15 2015-01-22 Novartis Ag Piperidinyl indole derivatives and their use as complement factor b inhibitors
WO2015008229A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors
WO2015059668A1 (en) 2013-10-25 2015-04-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
WO2015066241A1 (en) 2013-10-30 2015-05-07 Novartis Ag 2-benzyl-benzimidazole complement factor b inhibitors and uses thereof
WO2015066413A1 (en) 2013-11-01 2015-05-07 Novartis Ag Oxazolidinone hydroxamic acid compounds for the treatment of bacterial infections
WO2015066188A1 (en) 2013-11-01 2015-05-07 Novartis Ag Aminoheteroaryl benzamides as kinase inhibitors
WO2015075665A1 (en) 2013-11-21 2015-05-28 Novartis Ag Pyrrolopyrrolone derivatives and their use as bet inhibitors
WO2015079417A1 (en) 2013-11-29 2015-06-04 Novartis Ag Novel amino pyrimidine derivatives
WO2015092740A1 (en) 2013-12-20 2015-06-25 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2015095477A1 (en) 2013-12-19 2015-06-25 Irm Llc [1,2,4]triazolo[1,5-a]pyrimidine derivatives as protozoan proteasome inhibitors for the treatment of parasitic diseases such as leishmaniasis
WO2015095301A2 (en) 2013-12-17 2015-06-25 Irm Llc Cytotoxic peptides and conjugates thereof
WO2015102929A1 (en) 2013-12-30 2015-07-09 Novartis Ag Tricyclic sulfonamide derivatives
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
WO2015148379A1 (en) 2014-03-24 2015-10-01 Novartis Ag Monobactam organic compounds for the treatment of bacterial infections
WO2015070177A3 (en) * 2013-11-11 2015-10-08 Collaborative Medicinal Development, Llc Metal complexes and methods of treatment
WO2015161052A1 (en) 2014-04-17 2015-10-22 Novartis Ag Polycyclic herg activators
US9169279B2 (en) 2009-07-31 2015-10-27 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
WO2015162461A1 (en) 2014-04-24 2015-10-29 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162456A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162558A1 (en) 2014-04-24 2015-10-29 Novartis Ag Autotaxin inhibitors
WO2015164458A1 (en) 2014-04-22 2015-10-29 Novartis Ag Isoxazoline hydroxamic acid derivatives as lpxc inhibitors
WO2015162459A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015173659A2 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015173656A1 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015175487A1 (en) 2014-05-13 2015-11-19 Novartis Ag Compounds and compositions for inducing chondrogenesis
WO2015175796A1 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015177646A1 (en) 2014-05-14 2015-11-26 Novartis Ag Carboxamide derivatives
WO2015181747A1 (en) 2014-05-28 2015-12-03 Novartis Ag Novel pyrazolo pyrimidine derivatives and their use as malt1 inhibitors
WO2015186062A1 (en) 2014-06-03 2015-12-10 Novartis Ag PYRIMIDO[4,5-b]QUINOLINE-4,5(3H,10H)-DIONE DERIVATIVES
WO2015186063A1 (en) 2014-06-03 2015-12-10 Novartis Ag Naphthyridinedione derivatives
WO2015186061A1 (en) 2014-06-03 2015-12-10 Novartis Ag Pyridopyrimidinedione derivatives
WO2015189791A1 (en) 2014-06-13 2015-12-17 Novartis Ag Auristatin derivatives and conjugates thereof
WO2016001878A1 (en) 2014-07-02 2016-01-07 Novartis Ag Cyclohexen-1-yl-pyridin-2-yl-1h-pyrazole-4-carboxylic acid derivatives and the use thereof as soluble guanylate cyclase activators
WO2016001876A1 (en) 2014-07-02 2016-01-07 Novartis Ag Thiophen-2-yl-pyridin-2-yl-1h-pyrazole-4-carboxylic acid derivatives and the use thereof as soluble guanylate cyclase activators
WO2016001875A1 (en) 2014-07-02 2016-01-07 Novartis Ag Indane and indoline derivatives and the use thereof as soluble guanylate cyclase activators
WO2016020836A1 (en) 2014-08-06 2016-02-11 Novartis Ag Quinolone derivatives as antibacterials
CN105399665A (en) * 2015-12-11 2016-03-16 吉林大学珠海学院 Deferiprone pharmaceutical cocrystal with p-hydroxybenzoic acid as precursor and preparation method thereof
CN105399664A (en) * 2015-12-11 2016-03-16 吉林大学珠海学院 Deferiprone pharmaceutical cocrystal with 2,5-dihydroxybenzoic acid as precursor and preparation method thereof
WO2016038581A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as raf kinase inhibitors
WO2016038582A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as raf kinase inhibitors
WO2016038583A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as kinase inhibitors
WO2016049266A1 (en) * 2014-09-24 2016-03-31 Pain Therapeutics, Inc. 4:3 naltrexone: 5-methyl-2-furaldehyde cocrystal
CN105541701A (en) * 2015-12-11 2016-05-04 吉林大学珠海学院 Pharmaceutical cocrystal of deferiprone with maleic acid as precursor, and preparation method thereof
US9340565B2 (en) 2010-11-24 2016-05-17 Thar Pharmaceuticals, Inc. Crystalline forms
WO2016079669A1 (en) 2014-11-19 2016-05-26 Novartis Ag Labeled amino pyrimidine derivatives
WO2016088082A1 (en) 2014-12-05 2016-06-09 Novartis Ag Amidomethyl-biaryl derivatives complement factor d inhibitors and uses thereof
WO2016097995A1 (en) 2014-12-16 2016-06-23 Novartis Ag Isoxazole hydroxamic acid compounds as lpxc inhibitors
WO2016103155A1 (en) 2014-12-23 2016-06-30 Novartis Ag Triazolopyrimidine compounds and uses thereof
US9388134B2 (en) 2005-11-09 2016-07-12 Novartis, Ag Compounds containing S-N-valeryl-N-{[2′-(1H-tetrazole-5-yl)-biphenyl-4-yl]-methyl)-valine and (2R,4S)-5-biphenyl-4-yl-4-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester moieties and cations
EP3048100A1 (en) 2009-05-28 2016-07-27 Novartis AG Substituted aminobutyric derivatives as neprilysin inhibitors
EP3064502A1 (en) 2012-01-26 2016-09-07 Novartis AG Imidazopyrrolidinone compounds
WO2016151499A1 (en) 2015-03-25 2016-09-29 Novartis Ag Formylated n-heterocyclic derivatives as fgfr4 inhibitors
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
WO2016174616A1 (en) 2015-04-30 2016-11-03 Novartis Ag Fused tricyclic pyrazole derivatives useful for modulating farnesoid x receptors
WO2016203432A1 (en) 2015-06-17 2016-12-22 Novartis Ag Antibody drug conjugates
US9532993B2 (en) 2011-11-25 2017-01-03 Nuformix Limited Aprepitant L-proline solvates—compositions and cocrystals
CN106432136A (en) * 2016-06-30 2017-02-22 中国药科大学 Hydrochlorothiazide and atenolol co-amorphous system and preparation method thereof
WO2017053936A1 (en) 2015-09-24 2017-03-30 Pain Therapeutic, Inc. Cocrystals of naloxone and naltrexone
WO2017081641A1 (en) 2015-11-13 2017-05-18 Novartis Ag Novel pyrazolo pyrimidine derivatives
WO2017089985A1 (en) 2015-11-26 2017-06-01 Novartis Ag Diamino pyridine derivatives
WO2017103677A1 (en) 2015-12-16 2017-06-22 Druggability Technologies Ip Holdco Limited Complexes of celecoxib and its salts and derivatives process for the preparation thereof and pharmaceutical compositions containing them
WO2017103888A1 (en) 2015-12-18 2017-06-22 Novartis Ag Indane derivatives and the use thereof as soluble guanylate cyclase activators
WO2017103824A1 (en) 2015-12-18 2017-06-22 Novartis Ag Tricyclic compounds and compositions as kinase inhibitors
WO2017125898A1 (en) 2016-01-21 2017-07-27 Novartis Ag Compounds and compositions for the treatment of cryptosporidiosis
US9725453B2 (en) 2012-03-19 2017-08-08 Ironwood Pharmaceuticals, Inc. Imidazotriazinone compounds
WO2017140821A1 (en) 2016-02-19 2017-08-24 Novartis Ag Tetracyclic pyridone compounds as antivirals
US9744137B2 (en) 2006-08-31 2017-08-29 Supernus Pharmaceuticals, Inc. Topiramate compositions and methods of enhancing its bioavailability
WO2017149463A1 (en) 2016-03-01 2017-09-08 Novartis Ag Cyano-substituted indole compounds and uses thereof as lsd1 inhibitors
WO2017153919A1 (en) 2016-03-08 2017-09-14 Novartis Ag Tricyclic compounds useful to treat orthomyxovirus infections
WO2017163186A1 (en) 2016-03-24 2017-09-28 Novartis Ag Alkynyl nucleoside analogs as inhibitors of human rhinovirus
WO2017175185A1 (en) 2016-04-08 2017-10-12 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2017216685A1 (en) 2016-06-16 2017-12-21 Novartis Ag Pentacyclic pyridone compounds as antivirals
WO2017216686A1 (en) 2016-06-16 2017-12-21 Novartis Ag 8,9-fused 2-oxo-6,7-dihydropyrido-isoquinoline compounds as antivirals
WO2017221100A1 (en) 2016-06-20 2017-12-28 Novartis Ag Imidazopyrimidine compounds useful for the treatment of cancer
WO2017221092A1 (en) 2016-06-20 2017-12-28 Novartis Ag Triazolopyridine compounds and uses thereof
WO2018014829A1 (en) 2016-07-20 2018-01-25 Novartis Ag Aminopyridine derivatives and their use as selective alk-2 inhibitors
WO2018042316A1 (en) 2016-08-29 2018-03-08 Novartis Ag N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
WO2018042303A1 (en) 2016-08-29 2018-03-08 Novartis Ag Fused tricyclic pyridazinone compounds useful to treat orthomyxovirus infections
WO2018047081A1 (en) 2016-09-09 2018-03-15 Novartis Ag Compounds and compositions as inhibitors of endosomal toll-like receptors
WO2018047109A1 (en) 2016-09-09 2018-03-15 Novartis Ag Polycyclic pyridone compounds as antivirals
WO2018055551A1 (en) 2016-09-23 2018-03-29 Novartis Ag Aza-indazole compounds for use in tendon and/or ligament injuries
WO2018055550A1 (en) 2016-09-23 2018-03-29 Novartis Ag Indazole compounds for use in tendon and/or ligament injuries
WO2018060926A1 (en) 2016-09-28 2018-04-05 Novartis Ag Beta-lactamase inhibitors
WO2018073753A1 (en) 2016-10-18 2018-04-26 Novartis Ag Fused tetracyclic pyridone compounds as antivirals
WO2018087677A1 (en) 2016-11-10 2018-05-17 Novartis Ag Bmp potentiators
WO2018141749A1 (en) 2017-02-01 2018-08-09 Medivir Ab Therapeutic applications of malt1 inhibitors
WO2018172997A1 (en) 2017-03-24 2018-09-27 Novartis Ag Isoxazole carboxamide compounds and uses thereof
US10093691B2 (en) 2009-07-31 2018-10-09 Grunenthal Gmbh Crystallization method and bioavailability
WO2018198079A1 (en) 2017-04-27 2018-11-01 Novartis Ag Fused indazole pyridone compounds as antivirals
WO2018203302A1 (en) 2017-05-05 2018-11-08 Novartis Ag Tricyclic 2-quinolinones as antibacterials
WO2018234978A1 (en) 2017-06-19 2018-12-27 Novartis Ag Substituted 5-cyanoindole compounds and uses thereof
US10195218B2 (en) 2016-05-31 2019-02-05 Grunenthal Gmbh Crystallization method and bioavailability
WO2019087162A1 (en) 2017-11-06 2019-05-09 Novartis Ag Polycyclic herg activators
WO2019087163A1 (en) 2017-11-06 2019-05-09 Novartis Ag Polycyclic herg activators
WO2019097479A1 (en) 2017-11-17 2019-05-23 Novartis Ag Novel dihydroisoxazole compounds and their use for the treatment of hepatitis b
WO2019102256A1 (en) 2017-11-24 2019-05-31 Novartis Ag Pyridinone derivatives and their use as selective alk-2 inhibitors
WO2019124507A1 (en) 2017-12-21 2019-06-27 杏林製薬株式会社 Therapeutic agent for nocturnal pollakiuria
WO2019123285A1 (en) 2017-12-20 2019-06-27 Novartis Ag Fused tricyclic pyrazolo-dihydropyrazinyl-pyridone compounds as antivirals
WO2019154416A1 (en) 2018-02-07 2019-08-15 Novartis Ag Substituted bisphenyl butanoic ester derivatives as nep inhibitors
WO2019166950A1 (en) 2018-02-28 2019-09-06 Novartis Ag 10-(di(phenyl)methyl)-4-hydroxy-8,9,9a,10-tetrahydro-7h-pyrrolo[1 ',2':4,5]pyrazino[1,2-b]pyridazine-3,5-dione derivatives and related compounds as inhibitors of the orthomyxovirus replication for treating influenza
WO2019166951A1 (en) 2018-02-28 2019-09-06 Novartis Ag Indole-2-carbonyl compounds and their use for the treatment of hepatitis b
US10463653B2 (en) 2014-10-03 2019-11-05 Novartis Ag Use of ring-fused bicyclic pyridyl derivatives as FGFR4 inhibitors
WO2019223632A1 (en) 2018-05-22 2019-11-28 Js Innomed Holdings Ltd. Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
WO2019244049A1 (en) 2018-06-19 2019-12-26 Novartis Ag Cyanotriazole compounds and uses thereof
WO2020016594A1 (en) 2018-07-19 2020-01-23 Benevolentai Bio Limited Imidazo[1,2-b]pyridazine derivatives as trk inhibitors
EP3610883A1 (en) 2015-10-21 2020-02-19 Hox Therapeutics Limited Peptides
WO2020035779A1 (en) 2018-08-17 2020-02-20 Novartis Ag Urea compounds and compositions as smarca2/brm atpase inhibitors
WO2020039209A1 (en) 2018-08-23 2020-02-27 Benevolentai Bio Limited Imidazo[1,2-b]pyridazines as trk inhibitors
WO2020053654A1 (en) 2018-09-12 2020-03-19 Novartis Ag Antiviral pyridopyrazinedione compounds
WO2020058913A1 (en) 2018-09-21 2020-03-26 Novartis Ag Isoxazole carboxamide compounds and uses thereof
WO2020075046A1 (en) * 2018-10-09 2020-04-16 Novartis Ag Solid forms of n-(4-fluoro-3-(6-(3-methylpyridin-2-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-2-yl)phenyl)-2,4-dimethyloxazole-5-carboxamide
US10633344B2 (en) 2002-03-01 2020-04-28 University Of South Florida Multiple-component solid phases containing at least one active pharmaceutical ingredient
WO2020128768A1 (en) 2018-12-18 2020-06-25 Novartis Ag N-(pyridin-2-ylsulfonyl)cyclopropanecarboxamide derivatives and their use in the treatment of a cftr mediated disease
WO2020144604A1 (en) 2019-01-11 2020-07-16 Novartis Ag Lta4h inhibitors for the treatment of hidradenitis suppurativa
WO2020150626A1 (en) 2019-01-18 2020-07-23 Biogen Ma Inc. Imidazo[1,2-a]pyridinyl derivatives as irak4 inhibitors
WO2020154581A1 (en) * 2019-01-24 2020-07-30 Assia Chemical Industries Ltd Solid state forms of fedovapagon-salicyclic acid co-crystal
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
WO2020198077A1 (en) 2019-03-22 2020-10-01 Sumitomo Dainippon Pharma Oncology, Inc. Compositions comprising pkm2 modulators and methods of treatment using the same
WO2020250116A1 (en) 2019-06-10 2020-12-17 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf, copd, and bronchiectasis
WO2020263967A1 (en) 2019-06-27 2020-12-30 Biogen Ma Inc. 2h-indazole derivatives and their use in the treatment of disease
WO2020263980A1 (en) 2019-06-27 2020-12-30 Biogen Ma Inc. Imidazo[1,2-a]pyridinyl derivatives and their use in the treatment of disease
WO2021003417A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
WO2021038426A1 (en) 2019-08-28 2021-03-04 Novartis Ag Substituted 1,3-phenyl heteroaryl derivatives and their use in the treatment of disease
WO2021044351A1 (en) 2019-09-06 2021-03-11 Novartis Ag Methods of treating liver disease using lta4h inhibitors
CN112521292A (en) * 2020-12-18 2021-03-19 深圳市萱嘉生物科技有限公司 Eutectic crystal of betaine and organic acid and preparation method and application thereof
CN112552189A (en) * 2020-11-10 2021-03-26 中国海洋大学 Pharmaceutical co-crystal of amantadine hydrochloride and resveratrol and preparation method thereof
WO2021060949A1 (en) * 2019-09-26 2021-04-01 대봉엘에스 주식회사 Co-crystalline efinaconazole, and method for producing same
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
WO2021124172A1 (en) 2019-12-18 2021-06-24 Novartis Ag 3-(5-methoxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2021148807A1 (en) 2020-01-22 2021-07-29 Benevolentai Bio Limited Pharmaceutical compositions and their uses
WO2021148805A1 (en) 2020-01-22 2021-07-29 Benevolentai Bio Limited Topical pharmaceutical compositions comprising imidazo[1,2-b]pyridazine compounds
US11091489B2 (en) 2016-06-20 2021-08-17 Novartis Ag Crystalline forms of a triazolopyrimidine compound
EP3884938A1 (en) 2020-03-25 2021-09-29 ArtemiFlow GmbH 1,2,4-trioxane compounds and compositions comprising the same for use in the treatment of covid-19
WO2021214080A1 (en) 2020-04-20 2021-10-28 Novartis Ag Antiviral 1,3-di-oxo-indene compounds
WO2021214073A1 (en) 2020-04-20 2021-10-28 Novartis Ag Antiviral 1,3-di-oxo-indene compounds
CN113582927A (en) * 2020-04-30 2021-11-02 苏州恩华生物医药科技有限公司 Celecoxib and pregabalin co-amorphous compound and preparation method thereof
WO2021255608A1 (en) 2020-06-16 2021-12-23 Novartis Ag Methyl 2-methyl-5-oxo-1,4,5,7-tetradhydrofuro[3,4- b]pyridine-3-carboxylate compounds as cav1.2 activators
WO2021255607A1 (en) 2020-06-15 2021-12-23 Novartis Ag Methyl 2-(fluoromethyl)-5-oxo-4-phenyl-4,5,6,7-tetrahydro-1h-cyclopenta[b]pyridine-3-carboxylates and methyl 2-(fluoromethyl)-5-oxo-4-phenyl-1,4,5,7-tetrahydrofuro[3,4-b]pyridine-3-carboxylates as cav1.2 activators
CN113845438A (en) * 2021-10-12 2021-12-28 河北大学 Acetaminophen-piracetam pharmaceutical co-crystal and preparation method thereof
CN114031515A (en) * 2021-11-29 2022-02-11 河北大学 Paracetamol-ibuprofen pharmaceutical co-crystal and preparation method thereof
WO2022058902A1 (en) 2020-09-17 2022-03-24 Novartis Ag Compounds and compositions as sppl2a inhibitors
CN114644669A (en) * 2022-03-10 2022-06-21 华润紫竹药业有限公司 Preparation method and application of progesterone eutectic
WO2022140415A1 (en) 2020-12-22 2022-06-30 Biogen Ma Inc. 2h-indazole derivatives as irak4 inhibitors and their use in the treatment of disease
WO2022140425A1 (en) 2020-12-22 2022-06-30 Biogen Ma Inc. Imidazo[1,2-a]pyridine derivatives as irak4 inhibitors and their use in the treatment of disease
WO2022150446A1 (en) 2021-01-07 2022-07-14 Biogen Ma Inc. Tyk2 inhibitors
CN114835657A (en) * 2022-06-10 2022-08-02 大连工业大学 Method for regulating and controlling ethenzamide-saccharin eutectic crystal polycrystalline type based on ionic liquid
WO2022195355A1 (en) 2021-03-15 2022-09-22 Novartis Ag Benzisoxazole derivatives and uses thereof
WO2022195454A1 (en) 2021-03-15 2022-09-22 Novartis Ag Pyrazolopyridine derivatives and uses thereof
WO2022201097A1 (en) 2021-03-26 2022-09-29 Novartis Ag 1,3-substituted cyclobutyl derivatives and uses thereof
WO2022217232A1 (en) 2021-04-10 2022-10-13 Sunovion Pharmaceuticals Inc. Chromans and benzofurans as 5-ht1a and taar1 agonists
WO2022217248A1 (en) 2021-04-10 2022-10-13 Sunovion Pharmaceuticals Inc. Substituted sulfonamide-chroman compounds, and pharmaceutical compositions, and methods of use thereof
WO2022219546A1 (en) 2021-04-16 2022-10-20 Novartis Ag Heteroaryl aminopropanol derivatives as inhibitors of lta4h
WO2022234287A1 (en) 2021-05-06 2022-11-10 Benevolentai Bio Limited Imidazopyridazine derivatives useful as trk inhibitors
WO2022249060A1 (en) 2021-05-26 2022-12-01 Novartis Ag Triazolo-pyrimidine analogues for treating diseases connected to the inhibiton of werner syndrome recq helicase (wrn)
WO2022254362A1 (en) 2021-06-03 2022-12-08 Novartis Ag 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and medical uses thereof
EP4105208A1 (en) 2013-07-31 2022-12-21 Novartis AG 1,4-disubstituted pyridazine derivatives and their use for treating smn-deficiency-related conditions
WO2023275796A1 (en) 2021-07-01 2023-01-05 Novartis Ag Heterocyclic derivatives as sphingosine-1-phosphate 3 inhibitors
WO2023002443A1 (en) 2021-07-22 2023-01-26 Novartis Ag Substituted pyridone compounds useful to treat orthomyxovirus infections
WO2023049721A1 (en) 2021-09-23 2023-03-30 Sunovion Pharmaceuticals Inc. Methods of treating metabolic disorders
WO2023094965A1 (en) 2021-11-23 2023-06-01 Novartis Ag Naphthyridinone derivatives for the treatment of a disease or disorder
US11667613B2 (en) 2019-09-26 2023-06-06 Novartis Ag Antiviral pyrazolopyridinone compounds
WO2023111799A1 (en) 2021-12-13 2023-06-22 Novartis Ag PYRIDINE-3-CARBOXYLATE COMPOUNDS AS CaV1.2 ACTIVATORS
WO2023139534A1 (en) 2022-01-24 2023-07-27 Novartis Ag Spirocyclic piperidinyl derivatives as complement factor b inhibitors and uses thereof
WO2023187715A1 (en) 2022-04-01 2023-10-05 Novartis Ag Complement factor b inhibitors and uses thereof
US11806346B2 (en) 2020-05-13 2023-11-07 Chdi Foundation, Inc. HTT modulators for treating Huntington's disease
WO2024006493A1 (en) 2022-07-01 2024-01-04 Biogen Ma Inc. Tyk2 inhibitors
WO2024028782A1 (en) 2022-08-03 2024-02-08 Novartis Ag Nlrp3 inflammasome inhibitors

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5017103B2 (en) * 2004-06-17 2012-09-05 トランスフオーム・フアーマシユーチカルズ・インコーポレーテツド Pharmaceutical co-crystal composition and related methods of use
US7935817B2 (en) 2008-03-31 2011-05-03 Apotex Pharmachem Inc. Salt form and cocrystals of adefovir dipivoxil and processes for preparation thereof
GB0813709D0 (en) * 2008-07-26 2008-09-03 Univ Dundee Method and product
JP5558875B2 (en) * 2009-03-19 2014-07-23 日本曹達株式会社 Novel inclusion complex, epoxy resin composition and epoxy resin composition for semiconductor encapsulation
US9340536B2 (en) * 2012-06-15 2016-05-17 Basf Se Multicomponent crystals comprising dasatinib and selected co-crystal formers
KR101303803B1 (en) 2012-08-02 2013-09-04 순천향대학교 산학협력단 Preparation of co-crystals of various drug substances including cabamazepine-saccharin co-crystals by anti-solvent method
CA2988992A1 (en) * 2015-06-19 2016-12-22 Syn-Nat Products Enterprise LLC Pharmaceutical composition of carboplatin based co-crystals and use thereof
JP6929234B2 (en) * 2015-06-25 2021-09-01 シン−ナット プロダクツ エンタープライズ エルエルシー Pharmaceutical co-crystal composition and its uses
JP7348663B2 (en) * 2018-02-23 2023-09-21 センター フォー インテリジェント リサーチ イン クリスタル エンジニアリング,エセ.エレ. Co-crystals of ubiquinol and compositions containing them

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001042222A1 (en) * 1999-12-08 2001-06-14 Pharmacia Corporation Polymorphic crystalline forms of celecoxib
WO2001042221A1 (en) * 1999-12-08 2001-06-14 Pharmacia Corporation Solid-state form of celecoxib having enhanced bioavailability
WO2001051919A2 (en) * 2000-01-07 2001-07-19 Transform Pharmaceuticals, Inc. High-throughput formation, identification, and analysis of diverse solid-forms
WO2003033462A2 (en) * 2001-10-15 2003-04-24 The Regents Of The University Of Michigan Systems and methods for the generation of crystalline polymorphs
WO2003074474A2 (en) * 2002-03-01 2003-09-12 University Of South Florida Multiple-component solid phases containing at least one active pharmaceutical ingredient

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8500862D0 (en) * 1985-01-14 1985-02-20 Tate & Lyle Plc Composition
AU2003272270A1 (en) * 2003-02-28 2004-09-28 The Regents Of The University Of Michigan Pharmaceutical co-crystal compositions of drugs such as carbamazeptine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001042222A1 (en) * 1999-12-08 2001-06-14 Pharmacia Corporation Polymorphic crystalline forms of celecoxib
WO2001042221A1 (en) * 1999-12-08 2001-06-14 Pharmacia Corporation Solid-state form of celecoxib having enhanced bioavailability
WO2001051919A2 (en) * 2000-01-07 2001-07-19 Transform Pharmaceuticals, Inc. High-throughput formation, identification, and analysis of diverse solid-forms
WO2003033462A2 (en) * 2001-10-15 2003-04-24 The Regents Of The University Of Michigan Systems and methods for the generation of crystalline polymorphs
WO2003074474A2 (en) * 2002-03-01 2003-09-12 University Of South Florida Multiple-component solid phases containing at least one active pharmaceutical ingredient

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WEISSBUCH I ET AL: "Understanding and control of nucleation, growth, habit, dissolution and structure of two- and three-dimensional crystals using 'tailor-made' auxiliaries" 1995, ACTA CRYSTALLOGRAPHICA. SECTION B, STRUCTURAL SCIENCE, MUNKSGAARD, COPENHAGEN, DK, PAGE(S) 115-148 , XP002960386 ISSN: 0108-7681 the whole document abstract page 145, column 2, paragraph 2 *

Cited By (543)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7883713B2 (en) 2001-06-22 2011-02-08 Aicuris Gmbh & Co. Kg Topical application of thiazolyl amides
US7790905B2 (en) 2002-02-15 2010-09-07 Mcneil-Ppc, Inc. Pharmaceutical propylene glycol solvate compositions
US7927613B2 (en) 2002-02-15 2011-04-19 University Of South Florida Pharmaceutical co-crystal compositions
US10633344B2 (en) 2002-03-01 2020-04-28 University Of South Florida Multiple-component solid phases containing at least one active pharmaceutical ingredient
EP1511490A4 (en) * 2002-05-31 2009-03-11 Transform Pharmaceuticals Inc Novel conazole crystalline forms and related processes, pharmaceutical compositions and methods
EP1511490A2 (en) * 2002-05-31 2005-03-09 Transform Pharmaceuticals, Inc. Novel conazole crystalline forms and related processes, pharmaceutical compositions and methods
US8183290B2 (en) 2002-12-30 2012-05-22 Mcneil-Ppc, Inc. Pharmaceutically acceptable propylene glycol solvate of naproxen
JP2007513867A (en) * 2003-09-04 2007-05-31 セフアロン・インコーポレーテツド Modafinil composition
EP1670753A4 (en) * 2003-09-04 2008-01-02 Cephalon Inc Modafinil compositions
JP4842819B2 (en) * 2003-09-04 2011-12-21 セフアロン・インコーポレーテツド Modafinil composition
EP1670753A2 (en) * 2003-09-04 2006-06-21 Cephalon, Inc. Modafinil compositions
US7507823B2 (en) 2004-05-06 2009-03-24 Bristol-Myers Squibb Company Process of making aripiprazole particles
US8784887B2 (en) 2005-03-30 2014-07-22 Aicuris Gmbh & Co. Kg Pharmaceutical preparation of N-[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-N-methyl-2-[4-(2-pyridinyl)phenyl]acetamide
AU2006228747B2 (en) * 2005-03-30 2011-11-03 Aicuris Gmbh & Co. Kg Pharmaceutical preparation of N -[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-N-methyl-2-[4-(2-pyridinyl)phenyl] acetamide
WO2006103011A1 (en) * 2005-03-30 2006-10-05 Aicuris Gmbh & Co. Kg Pharmaceutical preparation of n -[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-n-methyl-2-[4-(2-pyridinyl)phenyl] acetamide
WO2006138421A3 (en) * 2005-06-15 2008-07-10 Elan Pharma Int Ltd Nanoparticulate azelnidipine formulations
WO2006138421A2 (en) * 2005-06-15 2006-12-28 Elan Pharma International Limited Nanoparticulate azelnidipine formulations
EP1951393A2 (en) * 2005-10-31 2008-08-06 The Regents of the University of Michigan Reaction co-crystallization of molecular complexes or co-crystals
EP1951393A4 (en) * 2005-10-31 2010-05-19 Univ Michigan Reaction co-crystallization of molecular complexes or co-crystals
US9388134B2 (en) 2005-11-09 2016-07-12 Novartis, Ag Compounds containing S-N-valeryl-N-{[2′-(1H-tetrazole-5-yl)-biphenyl-4-yl]-methyl)-valine and (2R,4S)-5-biphenyl-4-yl-4-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester moieties and cations
US20160324821A1 (en) * 2005-11-09 2016-11-10 Lili Feng Compounds containing s-n-valeryl-n--valine and (2r,4s)-5-biphenyl-4-yl-4-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester moieties and cations
EP2340828B1 (en) 2005-11-09 2020-07-15 Novartis AG Pharmaceutical combinations of an angiotensin receptor antagonist and a nep inhibitor
US11096918B2 (en) 2005-11-09 2021-08-24 Novartis Pharmaceuticals Corporation Amorphous solid form of compounds containing S—N-valeryl-N-{[2′-(1H-tetrazole-5-yl)-biphenyl-4-yl]-methyl}-valine and (2R,4S)-5-biphenyl-4-yl-4-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester moieties and sodium cations
US11642329B2 (en) 2005-11-09 2023-05-09 Novartis Pharmaceuticals Corporation Amorphous solid form of compounds containing S—N-valeryl-N- {[2′-( 1 H-tetrazole-5-yl)-biphenyl-4-yl]-methyl}-valine and (2R,4S)-5-biphenyl-4-yl-4-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester moieties and sodium cations
EP1962600A2 (en) * 2005-12-08 2008-09-03 SSCI, Inc. Metronidazole cocrystals and imipramine cocrystals
US8163790B2 (en) 2005-12-08 2012-04-24 New Form Pharmaceuticals, Inc. Metronidazole cocrystals and imipramine cocrystals
EP1962600A4 (en) * 2005-12-08 2010-09-22 Ssci Inc Metronidazole cocrystals and imipramine cocrystals
US9744137B2 (en) 2006-08-31 2017-08-29 Supernus Pharmaceuticals, Inc. Topiramate compositions and methods of enhancing its bioavailability
WO2008054609A2 (en) * 2006-10-04 2008-05-08 The Regents Of The University Of Michigan Dissolution and precipitation of cocrystals with ionizable components
WO2008054609A3 (en) * 2006-10-04 2008-06-19 Univ Michigan Dissolution and precipitation of cocrystals with ionizable components
WO2008063284A3 (en) * 2006-10-10 2008-07-10 Transform Pharmaceuticals Inc Novel crystal of (s)-(+)-2-(2-chlorophenyl)-2-hydroxy-ethyl carbamate
WO2008063284A2 (en) * 2006-10-10 2008-05-29 Janssen Pharmaceutica Nv Novel crystal of (s)-(+)-2-(2-chlorophenyl)-2-hydroxy-ethyl carbamate
US8663683B2 (en) 2006-11-17 2014-03-04 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US10314790B2 (en) 2006-11-17 2019-06-11 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US9555004B2 (en) 2006-11-17 2017-01-31 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US9622983B2 (en) 2006-11-17 2017-04-18 Supernus Pharmaceutcals, Inc. Sustained-release formulations of topiramate
US8298576B2 (en) 2006-11-17 2012-10-30 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US9549940B2 (en) 2006-11-17 2017-01-24 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US8889191B2 (en) 2006-11-17 2014-11-18 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US8992989B2 (en) 2006-11-17 2015-03-31 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US8877248B1 (en) 2006-11-17 2014-11-04 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US8298580B2 (en) 2006-11-17 2012-10-30 Supernus Pharmaceuticals, Inc. Sustained-release formulations of topiramate
US8859607B2 (en) 2007-02-09 2014-10-14 Basf Se Crystalline complexes of agriculturally active organic compounds
WO2008108639A1 (en) * 2007-03-08 2008-09-12 Avantium Holding B.V. Co-crystalline forms of carbamazepine
WO2008153945A3 (en) * 2007-06-06 2009-02-12 Univ South Florida Nutraceutical co-crystal compositions
US20100204204A1 (en) * 2007-06-06 2010-08-12 University Of South Florida Nutraceutical co-crystal compositions
US10842797B2 (en) 2007-06-06 2020-11-24 University Of South Florida Nutraceutical co-crystal compositions
WO2008153945A2 (en) * 2007-06-06 2008-12-18 University Of South Florida Nutraceutical co-crystal compositions
US10376521B2 (en) 2007-06-06 2019-08-13 University Of South Florida Nutraceutical co-crystal compositions
US8124603B2 (en) 2008-01-22 2012-02-28 Thar Pharmaceuticals In vivo studies of crystalline forms of meloxicam
WO2009094155A1 (en) * 2008-01-22 2009-07-30 Thar Pharmaceuticals In vivo studies of crystalline forms of meloxicam
EP2262488A4 (en) * 2008-04-07 2012-07-04 Mutual Pharmaceutical Co Colchicine solid complex; methods of making; and methods of use thereof
US8519002B2 (en) 2008-04-07 2013-08-27 Takeda Pharmaceuticals U.S.A., Inc. Colchicine solid complex; methods of making; and methods of use thereof
EP2262488A1 (en) * 2008-04-07 2010-12-22 Mutual Pharmaceutical Company, Inc. Colchicine solid complex; methods of making; and methods of use thereof
WO2009140466A3 (en) * 2008-05-14 2010-03-04 Dr. Reddy's Laboratories Ltd. Linezolid co-crystals
WO2009140466A2 (en) * 2008-05-14 2009-11-19 Dr. Reddy's Laboratories Ltd. Linezolid co-crystals
WO2009141144A1 (en) 2008-05-21 2009-11-26 Laboratorios Del Dr. Esteve, S.A. Co-crystals of duloxetine and cox-inhibitors for the treatment of pain
US9084774B2 (en) 2008-05-21 2015-07-21 Laboratorios Del Dr. Esteve, S.A. Co-crystals of duloxetine and cox-inhibitors for the treatment of pain
CN102036946A (en) * 2008-05-21 2011-04-27 埃斯蒂文博士实验室股份有限公司 Co-crystals of duloxetine and Cox-inhibitors for the treatment of pain
US8501802B2 (en) 2008-05-21 2013-08-06 Laboratorios Del Dr. Esteve, S.A. Co-crystals of duloxetine and COX-INHIBITORs for the treatment of pain
EP2123626A1 (en) * 2008-05-21 2009-11-25 Laboratorios del Dr. Esteve S.A. Co-crystals of duloxetine and co-crystal formers for the treatment of pain
WO2009152347A3 (en) * 2008-06-13 2010-04-15 Bionevia Pharmaceuticals Inc. Crystalline forms of zotepine hydrochloride
WO2009152347A2 (en) * 2008-06-13 2009-12-17 Bionevia Pharmaceuticals Inc. Crystalline forms of zotepine hydrochloride
US8258155B2 (en) 2008-06-30 2012-09-04 Mutual Pharmaceutical Company, Inc. Quinine sulfate/bisulfate solid complex; methods of making; and methods of use thereof
US8697735B2 (en) 2008-07-25 2014-04-15 Bionevia Pharmaceuticals, Inc. Solid forms of epalrestat
US9447056B2 (en) 2008-07-25 2016-09-20 Bionevia Pharmaceuticals, Inc. Solid forms of epalrestat
WO2010017343A3 (en) * 2008-08-06 2010-05-14 Bionevia Pharmaceuticals, Inc. Flupirtine hydrochloride maleic acid cocrystal
US8906948B2 (en) 2008-09-06 2014-12-09 Bionevia, LLC Choline cocrystal of epalrestat
US10464912B2 (en) 2008-09-06 2019-11-05 Bionevia Pharmaceuticals, Inc. Choline cocrystal of epalrestat
WO2010043412A1 (en) * 2008-10-17 2010-04-22 Laboratorios Del Dr. Esteve, S.A. Co-crystals of tramadol and nsaids
CN102186465B (en) * 2008-10-17 2015-09-16 埃斯蒂文博士实验室股份有限公司 The eutectic of tramadol and NSAID
US10245276B2 (en) 2008-10-17 2019-04-02 Laboratorios Del Dr. Esteve, S.A. Co-crystals of tramadol and coxibs
US11478488B2 (en) 2008-10-17 2022-10-25 Esteve Pharmaceuticals, S.A. Co-crystals of tramadol and coxibs
US10548909B2 (en) 2008-10-17 2020-02-04 Esteve Pharmaceuticals, S.A. Co-crystals of tramadol and coxibs
RU2599717C2 (en) * 2008-10-17 2016-10-10 Лабораторьос Дель Др. Эстеве, С.А. Co-crystalline forms of tramadol and nsaid
EP2199274A1 (en) * 2008-12-16 2010-06-23 Laboratorios Del. Dr. Esteve, S.A. Co-crystals of tramadol and paracetamol
ES2371848A1 (en) * 2008-12-16 2012-01-10 Laboratorios Del Dr. Esteve, S. A. Cocrystals of tramadol and paracetamol
WO2010069561A1 (en) * 2008-12-16 2010-06-24 Laboratorios Del Dr. Esteve, S.A. Cocrystals of tramadol and paracetamol
US9161912B2 (en) 2008-12-23 2015-10-20 Map Pharmaceuticals, Inc. Inhalation devices and related methods for administration of sedative hypnotic compounds
US8555875B2 (en) 2008-12-23 2013-10-15 Map Pharmaceuticals, Inc. Inhalation devices and related methods for administration of sedative hypnotic compounds
US8563616B2 (en) 2009-01-22 2013-10-22 Absorption Pharmaceuticals, LLC Desensitizing drug product
US8507561B2 (en) 2009-01-22 2013-08-13 Absorption Pharmaceuticals, LLC Desensitizing drug product
US8637577B2 (en) 2009-01-22 2014-01-28 Absorption Pharmaceuticals, LLC Desensitizing drug product
WO2010090491A2 (en) * 2009-02-09 2010-08-12 주식회사 한독약품 Novel salts of adefovir dipivoxil and preparation method thereof
WO2010090491A3 (en) * 2009-02-09 2011-01-27 주식회사 한독약품 Novel salts of adefovir dipivoxil and preparation method thereof
WO2010115981A1 (en) 2009-04-10 2010-10-14 Novartis Ag 7-azadispiro [3.0.4.1] decane-8-carboxamides as hepatitis c virus inhibitors
WO2010116248A1 (en) 2009-04-10 2010-10-14 Novartis Ag Organic compounds and their uses
WO2010130773A2 (en) 2009-05-15 2010-11-18 Novartis Ag Benzoxazolone derivatives as aldosterone symthase inhibitors
WO2010130796A1 (en) 2009-05-15 2010-11-18 Novartis Ag Aryl pyridine as aldosterone synthase inhibitors
EP2432320A4 (en) * 2009-05-20 2013-03-06 Nutracryst Therapeutics Private Ltd Pharmaceutical co-crystals of quercetin
EP2432320A1 (en) * 2009-05-20 2012-03-28 Nutracryst Therapeutics Private Limited Pharmaceutical co-crystals of quercetin
EP2594557A1 (en) 2009-05-28 2013-05-22 Novartis AG Substituted aminopropionic derivatives as neprilysin inhibitors
EP2594556A1 (en) 2009-05-28 2013-05-22 Novartis AG Substituted aminopropionic derivatives as neprilysin inhibitors
WO2010136493A1 (en) 2009-05-28 2010-12-02 Novartis Ag Substituted aminopropionic derivatives as neprilysin inhibitors
EP3048100A1 (en) 2009-05-28 2016-07-27 Novartis AG Substituted aminobutyric derivatives as neprilysin inhibitors
WO2011009852A2 (en) 2009-07-21 2011-01-27 Novartis Ag Pharmaceutical compositions
WO2011009943A1 (en) 2009-07-24 2011-01-27 Novartis Ag Oxazine derivatives and their use as bace inhibitors for the treatment of neurological disorders
US8399023B2 (en) 2009-07-31 2013-03-19 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
US10323052B2 (en) 2009-07-31 2019-06-18 Grunenthal Gmbh Crystallization method and bioavailability
US9334296B2 (en) 2009-07-31 2016-05-10 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
US10093691B2 (en) 2009-07-31 2018-10-09 Grunenthal Gmbh Crystallization method and bioavailability
US8933057B2 (en) 2009-07-31 2015-01-13 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
US9169279B2 (en) 2009-07-31 2015-10-27 Thar Pharmaceuticals, Inc. Crystallization method and bioavailability
WO2011015360A1 (en) * 2009-08-06 2011-02-10 Laboratorios Del Dr. Esteve, S.A. Pharmaceutical compounds of o-desmethyl-tramadol and cox-inhibitors
EP2281558A1 (en) * 2009-08-06 2011-02-09 Laboratorios Del. Dr. Esteve, S.A. Pharmaceutical compounds of O-Desmethyl-Tramadol and COX-inhibitors
WO2011015652A1 (en) 2009-08-07 2011-02-10 Novartis Ag 3-heteroarylmethyl-imidazo[1,2-b]pyridazin-6-yl derivatives as c-met tyrosine kinase modulators
WO2011018454A1 (en) 2009-08-12 2011-02-17 Novartis Ag Heterocyclic hydrazone compounds and their uses to treat cancer and inflammation
WO2011020861A1 (en) 2009-08-20 2011-02-24 Novartis Ag Heterocyclic oxime compounds
WO2011023677A1 (en) 2009-08-26 2011-03-03 Novartis Ag Tetra-substituted heteroaryl compounds and their use as mdm2 and/or mdm4 modulators
WO2011026904A1 (en) 2009-09-04 2011-03-10 Novartis Ag Pyrazinylpyridines useful for the treatment of proliferative diseases
WO2011026917A1 (en) 2009-09-04 2011-03-10 Novartis Ag Heteroaryl compounds as kinase inhibitors
WO2011026911A1 (en) 2009-09-04 2011-03-10 Novartis Ag Bipyridines useful for the treatment of proliferative diseases
WO2011029915A1 (en) 2009-09-10 2011-03-17 Novartis Ag Ether derivatives of bicyclic heteroaryls
WO2011029842A1 (en) 2009-09-10 2011-03-17 Novartis Ag Sulfonamides as inhibitors of bcl-2 family proteins for the treatment of cancer
EP2325172A1 (en) * 2009-11-02 2011-05-25 Laboratorios Del. Dr. Esteve, S.A. Co-crystals of celecoxib and L-proline
WO2011054828A1 (en) 2009-11-04 2011-05-12 Novartis Ag Heterocyclic sulfonamide derivatives useful as mek inhibitors
WO2011061168A1 (en) 2009-11-17 2011-05-26 Novartis Ag Aryl-pyridine derivatives as aldosterone synthase inhibitors
EP2993169A1 (en) 2009-11-17 2016-03-09 Novartis AG Aryl-pyridine derivatives as aldosterone synthase inhibitors
WO2011061271A1 (en) 2009-11-20 2011-05-26 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel nep inhibitors
EP2735561A1 (en) 2009-11-20 2014-05-28 Novartis AG Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
WO2011064376A1 (en) 2009-11-30 2011-06-03 Novartis Ag Imidazole derivatives as aldosterone synthase inhibitors
WO2011073316A1 (en) 2009-12-18 2011-06-23 Novartis Ag 4-aryl-butane-1,3-diamides
WO2011076744A1 (en) 2009-12-21 2011-06-30 Novartis Ag Disubstituted heteroaryl-fused pyridines
WO2011076747A1 (en) 2009-12-21 2011-06-30 Novartis Ag Diaza-spiro[5.5]undecanes as orexin receptor antagonists
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
WO2011080176A1 (en) 2009-12-31 2011-07-07 Novartis Ag Pyrazine derivatives and their use in the treatment of neurological disorders
WO2011092293A2 (en) 2010-02-01 2011-08-04 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
WO2011092290A1 (en) 2010-02-01 2011-08-04 Novartis Ag Pyrazolo[5,1b]oxazole derivatives as crf-1 receptor antagonists
WO2011095452A1 (en) 2010-02-02 2011-08-11 Novartis Ag Aryl benzylamine compounds
WO2011095450A1 (en) 2010-02-02 2011-08-11 Novartis Ag Cyclohexyl amide derivatives as crf receptor antagonists
US8318807B2 (en) 2010-02-03 2012-11-27 Laurus Labs Private Limited Pterostilbene cocrystals
WO2011097372A3 (en) * 2010-02-03 2011-12-29 Aptuit Laurus Private Limited Pterostilbene cocrystals
US8513236B2 (en) 2010-02-03 2013-08-20 Laurus Labs Private Limited Pterostilbene cocrystals
US8415507B2 (en) 2010-02-03 2013-04-09 Laurus Labs Private Limited Pterostilbene cocrystals
EP2531196A4 (en) * 2010-02-03 2013-07-31 Laurus Labs Private Ltd Pterostilbene cocrystals
EP2531196A2 (en) * 2010-02-03 2012-12-12 Laurus Labs Private Limited Pterostilbene cocrystals
US8399712B2 (en) 2010-02-03 2013-03-19 Laurus Labs Private Limited Pterostilbene cocrystals
WO2011104266A1 (en) 2010-02-25 2011-09-01 Novartis Ag Dimeric iap inhibitors
US9365552B2 (en) 2010-03-19 2016-06-14 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US10117858B2 (en) 2010-03-19 2018-11-06 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
EP2845593A1 (en) 2010-03-19 2015-03-11 Novartis AG Pyridine and pyrazine derivative for the treatment of chronic obstructive pulmonary disease
US11911371B2 (en) 2010-03-19 2024-02-27 Novartis Ag Pyridine and pyrazine derivative for the treatment of chronic bronchitis
WO2011113894A1 (en) 2010-03-19 2011-09-22 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf
USRE46757E1 (en) 2010-03-19 2018-03-20 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
WO2011131709A1 (en) 2010-04-21 2011-10-27 Novartis Ag Furopyridine compounds and uses thereof
US8779146B2 (en) 2010-04-28 2014-07-15 Nuformix Limited Cilostazol cocrystals and compositions
WO2011158110A3 (en) * 2010-04-28 2012-03-15 Nuformix Limited Cilostazol cocrystals and compositions
CN102933205A (en) * 2010-04-28 2013-02-13 诺弗米克斯有限公司 Cilostazol cocrystals and compositions
WO2011144666A1 (en) 2010-05-20 2011-11-24 Novartis Ag 2,4-dioxo-1,4-dihydro-2h-quinazolin-3-yl-sulfonamide derivatives
WO2011157793A1 (en) 2010-06-17 2011-12-22 Novartis Ag Piperidinyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2011157787A1 (en) 2010-06-17 2011-12-22 Novartis Ag Biphenyl substituted 1,3-dihydro-benzoimidazol-2-ylideneamine derivatives
WO2012003189A1 (en) 2010-06-29 2012-01-05 Irm Llc Compositions and methods for modulating the wnt signaling pathway
WO2012004299A1 (en) 2010-07-06 2012-01-12 Novartis Ag Tetrahydro-pyrido-pyrimidine derivatives
WO2012006953A1 (en) 2010-07-13 2012-01-19 Novartis Ag Oxazine derivatives and their use in the treatment of neurological disorders
WO2012007539A1 (en) 2010-07-14 2012-01-19 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2012010663A1 (en) 2010-07-22 2012-01-26 Novartis Ag 2,3,5-trisubstituted thiophene compounds and uses thereof
WO2012017020A1 (en) 2010-08-04 2012-02-09 Novartis Ag N-((6-amino-pyridin-3-yl)methyl)-heteroaryl-carboxamides as inhibitors of plasma kallikrein
WO2012035023A1 (en) 2010-09-13 2012-03-22 Novartis Ag Triazine-oxadiazoles
WO2012035158A1 (en) 2010-09-17 2012-03-22 Novartis Ag Pyrazine derivatives as enac blockers
WO2012040230A1 (en) 2010-09-20 2012-03-29 Envivo Pharmaceuticals, Inc. Imidazotriazinone compounds
EP3181566A1 (en) 2010-09-20 2017-06-21 Ironwood Pharmaceuticals, Inc. Imidazotriazinone compounds
US9540380B2 (en) 2010-09-20 2017-01-10 Ironwood Pharmaceuticals, Inc. Imidazotriazinone compounds
WO2012048235A1 (en) 2010-10-08 2012-04-12 Novartis Ag Vitamin e formulations of sulfamide ns3 inhibitors
WO2012055888A1 (en) 2010-10-26 2012-05-03 Novartis Ag Diaza-spiro[5.5]undecanes useful as orexin receptor antagonists
WO2012065953A1 (en) 2010-11-16 2012-05-24 Novartis Ag Substituted carbamoylcycloalkyl acetic acid derivatives as nep inhibitors
WO2012065958A1 (en) 2010-11-16 2012-05-24 Novartis Ag Method of treating contrast-induced nephropathy
WO2012065956A1 (en) 2010-11-16 2012-05-24 Novartis Ag Substituted amino bisphenyl pentanoic acid derivatives as nep inhibitors
US10519176B2 (en) 2010-11-24 2019-12-31 Thar Pharma, Llc Crystalline forms
US9340565B2 (en) 2010-11-24 2016-05-17 Thar Pharmaceuticals, Inc. Crystalline forms
WO2012080260A1 (en) 2010-12-13 2012-06-21 Novartis Ag Dimeric iap inhibitors
WO2012080271A1 (en) 2010-12-13 2012-06-21 Novartis Ag Dimeric iap inhibitors
WO2012087520A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2012087519A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating fxr
WO2012084873A1 (en) 2010-12-20 2012-06-28 Novartis Ag 4- (hetero) aryl - ethynyl - octahydro - indole - 1 - carboxylic acid esters
WO2012087521A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2012101487A1 (en) 2010-12-22 2012-08-02 Novartis Ag Di/tri-aza-spiro-c9-c11alkanes
WO2012093101A1 (en) 2011-01-04 2012-07-12 Novartis Ag Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration (amd)
US9085555B2 (en) 2011-01-04 2015-07-21 Novartis Ag Complement pathway modulators and uses thereof
WO2012095463A1 (en) 2011-01-12 2012-07-19 Novartis Ag Oxazine derivatives and their use in the treatment of neurological disorders
WO2012095469A1 (en) 2011-01-13 2012-07-19 Novartis Ag Novel heterocyclic derivatives and their use in the treatment of neurological disorders
WO2012095521A1 (en) 2011-01-13 2012-07-19 Novartis Ag Bace-2 inhibitors for the treatment of metabolic disorders
WO2012098501A1 (en) * 2011-01-21 2012-07-26 Ranbaxy Laboratories Limited Febuxostat co-crystals
WO2012101058A1 (en) 2011-01-24 2012-08-02 Novartis Ag 4-tolyl-ethynyl-octahydro-indole-1-ester derivatives
WO2012101066A1 (en) 2011-01-28 2012-08-02 Novartis Ag Pyridine biaryl amine compounds and their uses
WO2012101065A2 (en) 2011-01-28 2012-08-02 Novartis Ag Pyrimidine biaryl amine compounds and their uses
WO2012101064A1 (en) 2011-01-28 2012-08-02 Novartis Ag N-acyl pyrimidine biaryl compounds as protein kinase inhibitors
WO2012101063A1 (en) 2011-01-28 2012-08-02 Novartis Ag N-acyl pyridine biaryl compounds and their uses
WO2012104776A1 (en) 2011-01-31 2012-08-09 Novartis Ag Novel heterocyclic derivatives
WO2012104823A2 (en) 2011-02-04 2012-08-09 Novartis Ag Pyridopyrimidinone compounds in the treatment of neurodegenerative diseases
WO2012107500A1 (en) 2011-02-10 2012-08-16 Novartis Ag [1, 2, 4] triazolo [4, 3 -b] pyridazine compounds as inhibitors of the c-met tyrosine kinase
WO2012116217A1 (en) 2011-02-25 2012-08-30 Irm Llc Compounds and compositions as trk inhibitors
WO2012117224A1 (en) 2011-03-02 2012-09-07 Sabrepharm Limited Dipyridinium derivatives
WO2012120469A1 (en) 2011-03-08 2012-09-13 Novartis Ag Fluorophenyl bicyclic heteroaryl compounds
US10130708B2 (en) 2011-03-24 2018-11-20 University Of South Florida Lithium cocrystal compositions
WO2012129568A3 (en) * 2011-03-24 2012-11-22 University Of South Florida Lithium compositions
WO2012131633A1 (en) 2011-04-01 2012-10-04 Novartis Ag Pyrazolo pyrimidine derivatives
WO2012138648A1 (en) 2011-04-06 2012-10-11 Irm Llc Compositions and methods for modulating lpa receptors
WO2012140596A1 (en) 2011-04-14 2012-10-18 Novartis Ag Glycoside derivatives and uses thereof
WO2012140597A1 (en) 2011-04-14 2012-10-18 Novartis Ag Glycoside derivatives and their uses for the treatment of diabetes
KR20140008370A (en) * 2011-04-28 2014-01-21 젠티바, 케이.에스. Pharmaceutically acceptable cocrystals of n-[2-(7-methoxy-1-naphtyl)ethyl]acetamide and methods of their preparation
US8927771B2 (en) 2011-04-28 2015-01-06 Zentiva K.S. Pharmaceutically acceptable cocrystals of N-[2-(7-methoxyl-1-naphtyl)ethyl]acetamide and methods of their preparation
WO2012146371A1 (en) * 2011-04-28 2012-11-01 Zentiva, K.S. Pharmaceutically acceptable cocrystals of n-[2-(7-methoxy-1-naphtyl)ethyl]acetamide and methods of their preparation
EA024422B1 (en) * 2011-04-28 2016-09-30 Зентива, К.С. Pharmaceutically acceptable cocrystals of n-[2-(7-methoxy-1-naphtyl)ethyl]acetamide and methods of their preparation
CN103501777B (en) * 2011-04-28 2015-08-19 赞蒂瓦有限合伙公司 The pharmaceutically acceptable eutectic of N-[2-(7-methoxy-1-naphthyl) ethyl] acetamide and its preparation method
CN103501777A (en) * 2011-04-28 2014-01-08 赞蒂瓦有限合伙公司 Pharmaceutically acceptable cocrystals of n-[2-(7-methoxy-1-naphtyl)ethyl]acetamide and methods of their preparation
WO2012156936A1 (en) 2011-05-17 2012-11-22 Novartis Ag Substituted indole derivatives for the treatment of immunological disorders
WO2012164473A1 (en) 2011-05-27 2012-12-06 Novartis Ag 3-spirocyclic piperidine derivatives as ghrelin receptor agonists
WO2012175520A1 (en) 2011-06-20 2012-12-27 Novartis Ag Hydroxy substituted isoquinolinone derivatives
WO2012175487A1 (en) 2011-06-20 2012-12-27 Novartis Ag Cyclohexyl isoquinolinone compounds
WO2013008095A1 (en) 2011-07-08 2013-01-17 Novartis Ag Novel pyrrolo pyrimidine derivatives
WO2013008162A1 (en) 2011-07-08 2013-01-17 Novartis Ag Novel trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
WO2013008164A2 (en) 2011-07-08 2013-01-17 Novartis Ag Method of treating atherosclerosis in high triglyceride subjects
WO2013016197A1 (en) 2011-07-22 2013-01-31 Novartis Ag Tetrahydropyrido-pyridine and tetrahydropyrido-pyrimidine compounds and use thereof as c5a receptor modulators
WO2013014627A1 (en) 2011-07-27 2013-01-31 Novartis Ag Pyrazoline derivatives and their use as selective androgen receptor modulators
EP3023423A1 (en) 2011-08-25 2016-05-25 Novartis AG 2 -amino-4 - (pyridin- 2 -yl) - 5, 6 -dihydro-4h- 1, 3 -oxazine derivatives and their use as bace-1 and/or bace - 2 inhibitors
WO2013027188A1 (en) 2011-08-25 2013-02-28 Novartis Ag 2 -amino-4 - (pyridin- 2 -yl) - 5, 6 -dihydro-4h- 1, 3 -oxazine derivatives and their use as bace-1 and/or bace - 2 inhibitors
WO2013033167A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013033203A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013030802A1 (en) 2011-09-01 2013-03-07 Novartis Ag Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
WO2013033620A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as pdgfr kinase inhibitors
WO2013033070A1 (en) 2011-09-01 2013-03-07 Irm Llc COMPOUNDS AND COMPOSITIONS AS c-KIT KINASE INHIBITORS
WO2013033116A1 (en) 2011-09-01 2013-03-07 Irm Llc Compounds and compositions as c-kit kinase inhibitors
WO2013035047A1 (en) 2011-09-06 2013-03-14 Novartis Ag Benzothiazolone compound
WO2013038362A1 (en) 2011-09-15 2013-03-21 Novartis Ag 6 - substituted 3 - (quinolin- 6 - ylthio) - [1,2,4] triazolo [4, 3 -a] pyradines as tyrosine kinase
WO2013038386A1 (en) 2011-09-16 2013-03-21 Novartis Ag Heterocyclic compounds for the treatment of cystic fibrosis
WO2013038381A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine/pyrazine amide derivatives
WO2013038373A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038378A1 (en) 2011-09-16 2013-03-21 Novartis Ag Pyridine amide derivatives
WO2013038390A1 (en) 2011-09-16 2013-03-21 Novartis Ag N-substituted heterocyclyl carboxamides
WO2013050987A1 (en) 2011-10-08 2013-04-11 Novartis Ag Carbamate/ urea derivatives containing piperidin and piperazin rings as h3 receptor inhibitors
WO2013054291A1 (en) 2011-10-13 2013-04-18 Novartis Ag Novel oxazine derivatives and their use in the treatment of disease
WO2013057169A1 (en) 2011-10-19 2013-04-25 Universiteit Gent Pharmaceutical nanosuspension
TWI580442B (en) * 2011-10-19 2017-05-01 傑特大學 Pharmaceutical nanosuspension
US10251866B2 (en) 2011-10-19 2019-04-09 Universiteit Gent Pharmaceutical nanosuspension
WO2013057711A1 (en) 2011-10-21 2013-04-25 Novartis Ag Quinazoline derivatives as pi3k modulators
WO2013061305A1 (en) 2011-10-28 2013-05-02 Novartis Ag Novel purine derivatives and their use in the treatment of disease
US9532993B2 (en) 2011-11-25 2017-01-03 Nuformix Limited Aprepitant L-proline solvates—compositions and cocrystals
WO2013080120A1 (en) 2011-11-28 2013-06-06 Novartis Ag Novel trifluoromethyl-oxadiazole derivatives and their use in the treatment of disease
WO2013080141A1 (en) 2011-11-29 2013-06-06 Novartis Ag Pyrazolopyrrolidine compounds
WO2013093850A1 (en) 2011-12-22 2013-06-27 Novartis Ag Quinoline derivatives
WO2013093849A1 (en) 2011-12-22 2013-06-27 Novartis Ag Dihydro-benzo-oxazine and dihydro-pyrido-oxazine derivatives
WO2013105063A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105065A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused piperidines as ip receptor agonists for the treatment of pah and related disorders
WO2013105061A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused dihydropyrido [2,3 -b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105057A1 (en) 2012-01-13 2013-07-18 Novartis Ag Fused pyrroles as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
WO2013105058A1 (en) 2012-01-13 2013-07-18 Novartis Ag 7,8- dihydropyrido [3, 4 - b] pyrazines as ip receptor agonists for the treatment of pulmonary arterial hypertension (pah) and related disorders
EP3272754A1 (en) 2012-01-26 2018-01-24 Novartis AG Imidazopyrrolidinone compounds
EP3064502A1 (en) 2012-01-26 2016-09-07 Novartis AG Imidazopyrrolidinone compounds
WO2013111108A1 (en) 2012-01-27 2013-08-01 Novartis Ag 5-membered heteroarylcarboxamide derivatives as plasma kallikrein inhibitors
WO2013111107A1 (en) 2012-01-27 2013-08-01 Novartis Ag Aminopyridine derivatives as plasma kallikrein inhibitors
WO2013128421A1 (en) 2012-03-02 2013-09-06 Novartis Ag Spirohydantoin compounds and their use as selective androgen receptor modulators
US9725453B2 (en) 2012-03-19 2017-08-08 Ironwood Pharmaceuticals, Inc. Imidazotriazinone compounds
US9969742B2 (en) 2012-03-19 2018-05-15 Ironwood Pharmaceuticals, Inc. Imidazotriazinone compounds
WO2013163508A1 (en) 2012-04-27 2013-10-31 Novartis Ag Tetrahydropyran dgat1 inhibitors
WO2013160873A1 (en) 2012-04-27 2013-10-31 Novartis Ag Cyclic bridgehead ether dgat1 inhibitors
WO2013164790A1 (en) 2012-05-03 2013-11-07 Novartis Ag L-malate salt of 2, 7 - diaza - spiro [4.5 ] dec- 7 - yle derivatives and crystalline forms thereof as ghrelin receptor agonists
WO2013164802A1 (en) 2012-05-04 2013-11-07 Novartis Ag Complement pathway modulators and uses thereof
WO2013171690A1 (en) 2012-05-16 2013-11-21 Novartis Ag Monocyclic heteroaryl cycloalkyldiamine derivatives
WO2013175417A1 (en) 2012-05-24 2013-11-28 Novartis Ag Pyrrolopyrrolidinone compounds
WO2013184766A1 (en) 2012-06-06 2013-12-12 Irm Llc Compounds and compositions for modulating egfr activity
WO2013184757A1 (en) 2012-06-06 2013-12-12 Irm Llc Compounds and compositions for modulating egfr activity
EP3257848A1 (en) 2012-06-06 2017-12-20 Novartis AG Compounds and compositions for modulating egfr activity
US9440957B2 (en) 2012-06-06 2016-09-13 Novartis Ag Compounds and compositions for modulating EGFR Activity
WO2013192345A1 (en) 2012-06-20 2013-12-27 Novartis Ag Complement pathway modulators and uses thereof
WO2013189910A1 (en) * 2012-06-22 2013-12-27 Basf Se Multicomponent crystals comprising imatinib mesilate and selected co-crystal formers
US9221789B2 (en) 2012-06-22 2015-12-29 Basf Se Multicomponent crystals comprising imatinib mesilate and selected co-crystal formers
WO2014002053A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
US9487483B2 (en) 2012-06-28 2016-11-08 Novartis Ag Complement pathway modulators and uses thereof
US9464081B2 (en) 2012-06-28 2016-10-11 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
US9388199B2 (en) 2012-06-28 2016-07-12 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014002058A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
WO2014002057A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
US9815819B2 (en) 2012-06-28 2017-11-14 Novartis Ag Complement pathway modulators and uses thereof
WO2014002052A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
WO2014002051A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
WO2014002054A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
US9550755B2 (en) 2012-07-12 2017-01-24 Novartis Ag Complement pathway modulators and uses thereof
WO2014009833A2 (en) 2012-07-12 2014-01-16 Novartis Ag Complement pathway modulators and uses thereof
WO2014013469A1 (en) 2012-07-20 2014-01-23 Novartis Ag Carbamate/urea derivatives
WO2014028459A1 (en) 2012-08-13 2014-02-20 Novartis Ag 1,4-disubstituted pyridazine analogs and methods for treating smn-deficiency-related conditions
EP3564233A1 (en) 2012-08-13 2019-11-06 Novartis AG 1,4-disubstituted pyridazine analogs and methods for treating smn-deficiency-related conditions
WO2014027300A1 (en) 2012-08-13 2014-02-20 Novartis Ag Bicyclic heteroaryl cycloalkyldiamine derivatives as inhibitors of spleen tyrosine kinases (syk)
WO2014033654A1 (en) 2012-08-30 2014-03-06 Novartis Ag Salts of benzothiazolone compound as beta-2-adrenoceptor agonist
EP3243817A1 (en) 2012-08-30 2017-11-15 Novartis AG Salts of benzothiazolone compound as beta-2-adrenoceptor agonist
WO2014033617A1 (en) 2012-08-31 2014-03-06 Novartis Ag 2'-ethynyl nucleoside derivatives for treatment of viral infections
WO2014033631A1 (en) 2012-08-31 2014-03-06 Novartis Ag N-(3-pyridyl) biarylamides as kinase inhibitors
WO2014033630A1 (en) 2012-08-31 2014-03-06 Novartis Ag Novel aminothiazole carboxamides as kinase inhibitors
WO2014037900A1 (en) 2012-09-07 2014-03-13 Novartis Ag Indole carboxamide derivatives and uses thereof
WO2014047020A1 (en) 2012-09-19 2014-03-27 Novartis Ag Dihydropyrrolidino-pyrimidines as kinase inhibitors
WO2014049514A1 (en) 2012-09-25 2014-04-03 Novartis Ag Compounds for use in gastric complication
WO2014052619A1 (en) 2012-09-27 2014-04-03 Irm Llc Piperidine derivatives and compositions as modulators of gpr119 activity
WO2014049540A2 (en) 2012-09-29 2014-04-03 Novartis Ag Cyclic peptides and use as medicines
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
WO2014072911A1 (en) 2012-11-07 2014-05-15 Novartis Ag Substituted indole derivatives
KR102153763B1 (en) 2012-11-12 2020-09-09 노파르티스 아게 Oxazolidin-2-one-pyrimidine derivatives
WO2014072956A1 (en) 2012-11-12 2014-05-15 Novartis Ag Oxazolidin-2-one-pyrimidine derivatives
KR20150082295A (en) * 2012-11-12 2015-07-15 노파르티스 아게 Oxazolidin-2-one-pyrimidine derivatives
EP3199158A1 (en) 2012-11-12 2017-08-02 Novartis AG Oxazolidin-2-one-pyrimidine derivatives for use in the treatment of skin fibrosis, scleroderma, hypertrophic scars or keloids
WO2014078813A1 (en) 2012-11-19 2014-05-22 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014078802A1 (en) 2012-11-19 2014-05-22 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014082935A1 (en) 2012-11-30 2014-06-05 Novartis Ag Cyclic nucleoside derivatives and uses thereof
WO2014093606A1 (en) 2012-12-13 2014-06-19 Novartis Ag Pyridone derivatives and uses thereof in the treatment of tuberculosis
WO2014091446A1 (en) 2012-12-13 2014-06-19 Novartis Ag Pyrimido [4,5-b]quinoline-4,5 (3h,10h)-diones as nonsense mutation suppressors
WO2014097151A2 (en) 2012-12-19 2014-06-26 Novartis Ag Autotaxin inhibitors
WO2014099880A1 (en) 2012-12-19 2014-06-26 Novartis Ag Aryl-substituted fused bicyclic pyridazine compounds
WO2014097148A1 (en) 2012-12-19 2014-06-26 Novartis Ag Tricyclic compounds for inhibiting the cftr channel
WO2014097147A1 (en) 2012-12-19 2014-06-26 Novartis Ag Tricyclic compounds as cftr inhibitors
WO2014115080A1 (en) 2013-01-22 2014-07-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the p53/mdm2 interaction
WO2014115077A1 (en) 2013-01-22 2014-07-31 Novartis Ag Substituted purinone compounds
EP3736271A1 (en) 2013-01-23 2020-11-11 Novartis AG Thiadiazole analogs and their use for treating diseases associated with a deficiency of smn motor neurons
WO2014116845A1 (en) 2013-01-23 2014-07-31 Novartis Ag Thiadiazole analogs thereof and methods for treating smn-deficiency-related-conditions
WO2014125413A1 (en) 2013-02-13 2014-08-21 Novartis Ag Ip receptor agonist heterocyclic compounds
WO2014126979A1 (en) 2013-02-14 2014-08-21 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as nep (neutral endopeptidase) inhibitors
WO2014128612A1 (en) 2013-02-20 2014-08-28 Novartis Ag Quinazolin-4-one derivatives
WO2014132205A1 (en) 2013-02-28 2014-09-04 Novartis Ag Formulation comprising benzothiazolone compound
US8889190B2 (en) 2013-03-13 2014-11-18 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
US10363224B2 (en) 2013-03-13 2019-07-30 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules
WO2014143638A1 (en) 2013-03-14 2014-09-18 Novartis Ag 2-(1h-indol-4-ylmethyl)-3h-imidazo[4,5-b]pyridine-6-carbonitrile derivatives as complement factor b inhibitors useful for the treatment of ophthalmic diseases
WO2014151616A1 (en) 2013-03-14 2014-09-25 Novartis Ag Biaryl amide compounds as kinase inhibitors
EP3299367A1 (en) 2013-03-14 2018-03-28 Novartis AG Biaryl amide compounds as kinase inhibitors
US10172878B2 (en) 2013-03-15 2019-01-08 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules
US9555005B2 (en) 2013-03-15 2017-01-31 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
WO2014151784A1 (en) 2013-03-15 2014-09-25 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014151729A1 (en) 2013-03-15 2014-09-25 Irm Llc Compounds and compositions for the treatment of parasitic diseases
WO2014160649A1 (en) 2013-03-29 2014-10-02 Novartis Ag Hydroxamic acid derivatives as lpxc inhibitors for the treatment of bacterial infections
WO2014167528A1 (en) 2013-04-11 2014-10-16 Novartis Ag Spiropyrazolopyridine derivatives and uses thereof for the treatment of viral infections
WO2014178040A1 (en) 2013-04-29 2014-11-06 Mapi Pharma Ltd. Co-crystals of dapagliflozin
US9006188B2 (en) 2013-04-29 2015-04-14 Mapi Pharma Ltd. Co-crystals of dapagliflozin
WO2014184778A1 (en) 2013-05-17 2014-11-20 Novartis Ag Pyrimidin-4-yl)oxy)-1h-indole-1-carboxamide derivatives and use thereof
WO2014191894A1 (en) 2013-05-27 2014-12-04 Novartis Ag Imidazopyrrolidinone derivatives and their use in the treatment of disease
EP3412675A1 (en) 2013-05-27 2018-12-12 Novartis AG Imidazopyrrolidinone derivatives and their use in the treatment of disease
WO2014191896A1 (en) 2013-05-27 2014-12-04 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
WO2014191911A1 (en) 2013-05-28 2014-12-04 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
WO2014191906A1 (en) 2013-05-28 2014-12-04 Novartis Ag Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
EP3299365A1 (en) 2013-07-15 2018-03-28 Novartis AG Piperidinyl-indole derivatives complement factor b inhibitors and uses thereof
WO2015009616A1 (en) 2013-07-15 2015-01-22 Novartis Ag Piperidinyl indole derivatives and their use as complement factor b inhibitors
WO2015008229A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors
WO2015008230A1 (en) 2013-07-18 2015-01-22 Novartis Ag Autotaxin inhibitors comprising a heteroaromatic ring-benzyl-amide-cycle core
EP4105208A1 (en) 2013-07-31 2022-12-21 Novartis AG 1,4-disubstituted pyridazine derivatives and their use for treating smn-deficiency-related conditions
WO2015059668A1 (en) 2013-10-25 2015-04-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
WO2015066241A1 (en) 2013-10-30 2015-05-07 Novartis Ag 2-benzyl-benzimidazole complement factor b inhibitors and uses thereof
WO2015066188A1 (en) 2013-11-01 2015-05-07 Novartis Ag Aminoheteroaryl benzamides as kinase inhibitors
WO2015066413A1 (en) 2013-11-01 2015-05-07 Novartis Ag Oxazolidinone hydroxamic acid compounds for the treatment of bacterial infections
WO2015070177A3 (en) * 2013-11-11 2015-10-08 Collaborative Medicinal Development, Llc Metal complexes and methods of treatment
CN105899519B (en) * 2013-11-11 2021-11-09 协同医药发展有限公司 Metal complexes and methods of treatment
CN105899519A (en) * 2013-11-11 2016-08-24 协同医药发展有限公司 Metal complexes and methods of treatment
US11266686B2 (en) 2013-11-11 2022-03-08 Procypra Therapeutics Llc Metal complexes and methods of treatment
WO2015075665A1 (en) 2013-11-21 2015-05-28 Novartis Ag Pyrrolopyrrolone derivatives and their use as bet inhibitors
EP3299368A1 (en) 2013-11-29 2018-03-28 Novartis AG Novel amino pyrimidine derivatives
EP3689865A1 (en) 2013-11-29 2020-08-05 Novartis AG Novel amino pyrimidine derivatives
WO2015079417A1 (en) 2013-11-29 2015-06-04 Novartis Ag Novel amino pyrimidine derivatives
EP4219478A1 (en) 2013-11-29 2023-08-02 Novartis AG Method of preparing amino pyrimidine derivatives
WO2015095301A2 (en) 2013-12-17 2015-06-25 Irm Llc Cytotoxic peptides and conjugates thereof
WO2015095477A1 (en) 2013-12-19 2015-06-25 Irm Llc [1,2,4]triazolo[1,5-a]pyrimidine derivatives as protozoan proteasome inhibitors for the treatment of parasitic diseases such as leishmaniasis
WO2015092740A1 (en) 2013-12-20 2015-06-25 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2015102929A1 (en) 2013-12-30 2015-07-09 Novartis Ag Tricyclic sulfonamide derivatives
WO2015148379A1 (en) 2014-03-24 2015-10-01 Novartis Ag Monobactam organic compounds for the treatment of bacterial infections
EP3511328A1 (en) 2014-03-24 2019-07-17 Novartis AG Monobactam organic compounds for the treatment of bacterial infections
WO2015161052A1 (en) 2014-04-17 2015-10-22 Novartis Ag Polycyclic herg activators
WO2015164458A1 (en) 2014-04-22 2015-10-29 Novartis Ag Isoxazoline hydroxamic acid derivatives as lpxc inhibitors
WO2015162456A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162459A1 (en) 2014-04-24 2015-10-29 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162461A1 (en) 2014-04-24 2015-10-29 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
WO2015162558A1 (en) 2014-04-24 2015-10-29 Novartis Ag Autotaxin inhibitors
WO2015175487A1 (en) 2014-05-13 2015-11-19 Novartis Ag Compounds and compositions for inducing chondrogenesis
WO2015175796A1 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015177646A1 (en) 2014-05-14 2015-11-26 Novartis Ag Carboxamide derivatives
WO2015173659A2 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015173656A1 (en) 2014-05-14 2015-11-19 Novartis Ag Carboxamide derivatives
WO2015181747A1 (en) 2014-05-28 2015-12-03 Novartis Ag Novel pyrazolo pyrimidine derivatives and their use as malt1 inhibitors
WO2015186062A1 (en) 2014-06-03 2015-12-10 Novartis Ag PYRIMIDO[4,5-b]QUINOLINE-4,5(3H,10H)-DIONE DERIVATIVES
WO2015186063A1 (en) 2014-06-03 2015-12-10 Novartis Ag Naphthyridinedione derivatives
WO2015186061A1 (en) 2014-06-03 2015-12-10 Novartis Ag Pyridopyrimidinedione derivatives
WO2015189791A1 (en) 2014-06-13 2015-12-17 Novartis Ag Auristatin derivatives and conjugates thereof
WO2016001878A1 (en) 2014-07-02 2016-01-07 Novartis Ag Cyclohexen-1-yl-pyridin-2-yl-1h-pyrazole-4-carboxylic acid derivatives and the use thereof as soluble guanylate cyclase activators
WO2016001876A1 (en) 2014-07-02 2016-01-07 Novartis Ag Thiophen-2-yl-pyridin-2-yl-1h-pyrazole-4-carboxylic acid derivatives and the use thereof as soluble guanylate cyclase activators
WO2016001875A1 (en) 2014-07-02 2016-01-07 Novartis Ag Indane and indoline derivatives and the use thereof as soluble guanylate cyclase activators
WO2016020836A1 (en) 2014-08-06 2016-02-11 Novartis Ag Quinolone derivatives as antibacterials
US9573969B2 (en) 2014-09-12 2017-02-21 Novartis Ag Compounds and compositions as kinase inhibitors
WO2016038581A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as raf kinase inhibitors
WO2016038582A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as raf kinase inhibitors
WO2016038583A1 (en) 2014-09-12 2016-03-17 Novartis Ag Compounds and compositions as kinase inhibitors
US9809610B2 (en) 2014-09-12 2017-11-07 Novartis Ag Compounds and compositions as kinase inhibitors
US9623017B2 (en) 2014-09-24 2017-04-18 Pain Therapeutics, Inc. 4:3 naltrexone: 5-methyl-2-furaldehyde cocrystal
WO2016049266A1 (en) * 2014-09-24 2016-03-31 Pain Therapeutics, Inc. 4:3 naltrexone: 5-methyl-2-furaldehyde cocrystal
US10463653B2 (en) 2014-10-03 2019-11-05 Novartis Ag Use of ring-fused bicyclic pyridyl derivatives as FGFR4 inhibitors
US10507201B2 (en) 2014-10-03 2019-12-17 Novartis Ag Use of ring-fused bicyclic pyridyl derivatives as FGFR4 inhibitors
WO2016079669A1 (en) 2014-11-19 2016-05-26 Novartis Ag Labeled amino pyrimidine derivatives
WO2016088082A1 (en) 2014-12-05 2016-06-09 Novartis Ag Amidomethyl-biaryl derivatives complement factor d inhibitors and uses thereof
WO2016097995A1 (en) 2014-12-16 2016-06-23 Novartis Ag Isoxazole hydroxamic acid compounds as lpxc inhibitors
WO2016103155A1 (en) 2014-12-23 2016-06-30 Novartis Ag Triazolopyrimidine compounds and uses thereof
WO2016151499A1 (en) 2015-03-25 2016-09-29 Novartis Ag Formylated n-heterocyclic derivatives as fgfr4 inhibitors
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
WO2016174616A1 (en) 2015-04-30 2016-11-03 Novartis Ag Fused tricyclic pyrazole derivatives useful for modulating farnesoid x receptors
WO2016203432A1 (en) 2015-06-17 2016-12-22 Novartis Ag Antibody drug conjugates
US11096936B2 (en) 2015-09-24 2021-08-24 Cassava Sciences, Inc. Cocrystals of naloxone and naltrexone
WO2017053936A1 (en) 2015-09-24 2017-03-30 Pain Therapeutic, Inc. Cocrystals of naloxone and naltrexone
EP3610883A1 (en) 2015-10-21 2020-02-19 Hox Therapeutics Limited Peptides
WO2017081641A1 (en) 2015-11-13 2017-05-18 Novartis Ag Novel pyrazolo pyrimidine derivatives
EP3757097A1 (en) 2015-11-26 2020-12-30 Novartis AG Diamino pyridine derivatives
WO2017089985A1 (en) 2015-11-26 2017-06-01 Novartis Ag Diamino pyridine derivatives
CN105541701A (en) * 2015-12-11 2016-05-04 吉林大学珠海学院 Pharmaceutical cocrystal of deferiprone with maleic acid as precursor, and preparation method thereof
CN105399664A (en) * 2015-12-11 2016-03-16 吉林大学珠海学院 Deferiprone pharmaceutical cocrystal with 2,5-dihydroxybenzoic acid as precursor and preparation method thereof
CN105399665A (en) * 2015-12-11 2016-03-16 吉林大学珠海学院 Deferiprone pharmaceutical cocrystal with p-hydroxybenzoic acid as precursor and preparation method thereof
US10307429B2 (en) 2015-12-16 2019-06-04 Druggability Technologies Ip Holdco Limited Complexes of celecoxib and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
WO2017103677A1 (en) 2015-12-16 2017-06-22 Druggability Technologies Ip Holdco Limited Complexes of celecoxib and its salts and derivatives process for the preparation thereof and pharmaceutical compositions containing them
US10688110B2 (en) 2015-12-16 2020-06-23 Nangenex Nanotechnology Incorporated Complexes of Celecoxib and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
WO2017103888A1 (en) 2015-12-18 2017-06-22 Novartis Ag Indane derivatives and the use thereof as soluble guanylate cyclase activators
WO2017103824A1 (en) 2015-12-18 2017-06-22 Novartis Ag Tricyclic compounds and compositions as kinase inhibitors
WO2017125898A1 (en) 2016-01-21 2017-07-27 Novartis Ag Compounds and compositions for the treatment of cryptosporidiosis
WO2017140821A1 (en) 2016-02-19 2017-08-24 Novartis Ag Tetracyclic pyridone compounds as antivirals
US10590079B2 (en) 2016-03-01 2020-03-17 Novartis Ag Cyano-substituted indoles as LSD1 inhibitors
WO2017149463A1 (en) 2016-03-01 2017-09-08 Novartis Ag Cyano-substituted indole compounds and uses thereof as lsd1 inhibitors
US10858366B2 (en) 2016-03-08 2020-12-08 Novartis Ag Tricyclic compounds useful to treat orthomyxovirus infections
WO2017153919A1 (en) 2016-03-08 2017-09-14 Novartis Ag Tricyclic compounds useful to treat orthomyxovirus infections
US11453677B2 (en) 2016-03-08 2022-09-27 Novartis Ag Tricyclic compounds useful to treat orthomyxovirus infections
EP4292658A2 (en) 2016-03-24 2023-12-20 Novartis AG Alkynyl nucleoside analogs as inhibitors of human rhinovirus
WO2017163186A1 (en) 2016-03-24 2017-09-28 Novartis Ag Alkynyl nucleoside analogs as inhibitors of human rhinovirus
WO2017175185A1 (en) 2016-04-08 2017-10-12 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
US10195218B2 (en) 2016-05-31 2019-02-05 Grunenthal Gmbh Crystallization method and bioavailability
WO2017216686A1 (en) 2016-06-16 2017-12-21 Novartis Ag 8,9-fused 2-oxo-6,7-dihydropyrido-isoquinoline compounds as antivirals
WO2017216685A1 (en) 2016-06-16 2017-12-21 Novartis Ag Pentacyclic pyridone compounds as antivirals
WO2017221100A1 (en) 2016-06-20 2017-12-28 Novartis Ag Imidazopyrimidine compounds useful for the treatment of cancer
US10689378B2 (en) 2016-06-20 2020-06-23 Novartis Ag Triazolopyridine compounds and uses thereof
US11548897B2 (en) 2016-06-20 2023-01-10 Novartis Ag Crystalline forms of a triazolopyrimidine compound
US10676479B2 (en) 2016-06-20 2020-06-09 Novartis Ag Imidazolepyridine compounds and uses thereof
US11091489B2 (en) 2016-06-20 2021-08-17 Novartis Ag Crystalline forms of a triazolopyrimidine compound
WO2017221092A1 (en) 2016-06-20 2017-12-28 Novartis Ag Triazolopyridine compounds and uses thereof
CN106432136B (en) * 2016-06-30 2018-12-25 中国药科大学 A kind of Hydrochioro and atenolol are total to unformed system and preparation method thereof
CN106432136A (en) * 2016-06-30 2017-02-22 中国药科大学 Hydrochlorothiazide and atenolol co-amorphous system and preparation method thereof
WO2018014829A1 (en) 2016-07-20 2018-01-25 Novartis Ag Aminopyridine derivatives and their use as selective alk-2 inhibitors
EP3971177A1 (en) 2016-07-20 2022-03-23 Novartis AG Aminopyridine derivatives and their use as selective alk-2 inhibitors
US11912715B2 (en) 2016-08-29 2024-02-27 Novartis Ag Fused tricyclic pyridazinone compounds useful to treat orthomyxovirus infections
US11098051B2 (en) 2016-08-29 2021-08-24 Novartis Ag Fused tricyclic pyridazinone compounds useful to treat orthomyxovirus infections
WO2018042303A1 (en) 2016-08-29 2018-03-08 Novartis Ag Fused tricyclic pyridazinone compounds useful to treat orthomyxovirus infections
WO2018042316A1 (en) 2016-08-29 2018-03-08 Novartis Ag N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
EP3848372A1 (en) 2016-08-29 2021-07-14 Novartis AG Fused tricyclic pyridazinone compounds useful to treat orthomyxovirus infections
EP4059934A1 (en) 2016-09-09 2022-09-21 Novartis AG Compounds and compositions as inhibitors of endosomal toll-like receptors
WO2018047081A1 (en) 2016-09-09 2018-03-15 Novartis Ag Compounds and compositions as inhibitors of endosomal toll-like receptors
WO2018047109A1 (en) 2016-09-09 2018-03-15 Novartis Ag Polycyclic pyridone compounds as antivirals
WO2018055551A1 (en) 2016-09-23 2018-03-29 Novartis Ag Aza-indazole compounds for use in tendon and/or ligament injuries
WO2018055550A1 (en) 2016-09-23 2018-03-29 Novartis Ag Indazole compounds for use in tendon and/or ligament injuries
EP3698796A1 (en) 2016-09-28 2020-08-26 Novartis AG Pharmaceutical combination of a tricyclic beta-lactamase inhibitor with specific beta-lactam antibiotics
WO2018060926A1 (en) 2016-09-28 2018-04-05 Novartis Ag Beta-lactamase inhibitors
WO2018073753A1 (en) 2016-10-18 2018-04-26 Novartis Ag Fused tetracyclic pyridone compounds as antivirals
WO2018087677A1 (en) 2016-11-10 2018-05-17 Novartis Ag Bmp potentiators
WO2018141749A1 (en) 2017-02-01 2018-08-09 Medivir Ab Therapeutic applications of malt1 inhibitors
WO2018172997A1 (en) 2017-03-24 2018-09-27 Novartis Ag Isoxazole carboxamide compounds and uses thereof
WO2018198079A1 (en) 2017-04-27 2018-11-01 Novartis Ag Fused indazole pyridone compounds as antivirals
EP3998269A1 (en) 2017-04-27 2022-05-18 Novartis AG Fused indazole pyridone compounds as antivirals
US10301312B2 (en) 2017-04-27 2019-05-28 Novartis Ag Fused indazole pyridone compounds as antivirals
US10975078B2 (en) 2017-04-27 2021-04-13 Novartis Ag Fused indazole pyridone compounds as antivirals
WO2018203302A1 (en) 2017-05-05 2018-11-08 Novartis Ag Tricyclic 2-quinolinones as antibacterials
WO2018234978A1 (en) 2017-06-19 2018-12-27 Novartis Ag Substituted 5-cyanoindole compounds and uses thereof
US10604502B2 (en) 2017-06-19 2020-03-31 Novartis Ag Substituted 5-cyanoindole compounds and uses thereof
WO2019087162A1 (en) 2017-11-06 2019-05-09 Novartis Ag Polycyclic herg activators
WO2019087163A1 (en) 2017-11-06 2019-05-09 Novartis Ag Polycyclic herg activators
WO2019097479A1 (en) 2017-11-17 2019-05-23 Novartis Ag Novel dihydroisoxazole compounds and their use for the treatment of hepatitis b
WO2019102256A1 (en) 2017-11-24 2019-05-31 Novartis Ag Pyridinone derivatives and their use as selective alk-2 inhibitors
WO2019123285A1 (en) 2017-12-20 2019-06-27 Novartis Ag Fused tricyclic pyrazolo-dihydropyrazinyl-pyridone compounds as antivirals
US11234977B2 (en) 2017-12-20 2022-02-01 Novartis Ag Fused tricyclic pyrazolo-dihydropyrazinyl-pyridone compounds as antivirals
WO2019124507A1 (en) 2017-12-21 2019-06-27 杏林製薬株式会社 Therapeutic agent for nocturnal pollakiuria
EP3778557A1 (en) 2018-02-07 2021-02-17 Novartis AG Combinations comprising a substituted biphenylbutanoic ester derivative as nep inhibitor and valsartan
WO2019154416A1 (en) 2018-02-07 2019-08-15 Novartis Ag Substituted bisphenyl butanoic ester derivatives as nep inhibitors
WO2019166951A1 (en) 2018-02-28 2019-09-06 Novartis Ag Indole-2-carbonyl compounds and their use for the treatment of hepatitis b
US11629149B2 (en) 2018-02-28 2023-04-18 Novartis Ag 10-(di(phenyl)methyl)-4-hydroxy-8,9,9A,10-tetrahydro-7H-pyrrolo[1′,2′:4,5]pyrazino[1,2-b]pyridazine-3,5-dione derivatives and related compounds as inhibitors of the orthomyxovirus replication for treating influenza
WO2019166950A1 (en) 2018-02-28 2019-09-06 Novartis Ag 10-(di(phenyl)methyl)-4-hydroxy-8,9,9a,10-tetrahydro-7h-pyrrolo[1 ',2':4,5]pyrazino[1,2-b]pyridazine-3,5-dione derivatives and related compounds as inhibitors of the orthomyxovirus replication for treating influenza
WO2019223632A1 (en) 2018-05-22 2019-11-28 Js Innomed Holdings Ltd. Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
WO2019244049A1 (en) 2018-06-19 2019-12-26 Novartis Ag Cyanotriazole compounds and uses thereof
WO2020016594A1 (en) 2018-07-19 2020-01-23 Benevolentai Bio Limited Imidazo[1,2-b]pyridazine derivatives as trk inhibitors
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
WO2020035779A1 (en) 2018-08-17 2020-02-20 Novartis Ag Urea compounds and compositions as smarca2/brm atpase inhibitors
EP4219488A1 (en) 2018-08-17 2023-08-02 Novartis AG Method for the preparation of urea compounds as smarca2/brm-atpase inhibitors
WO2020039209A1 (en) 2018-08-23 2020-02-27 Benevolentai Bio Limited Imidazo[1,2-b]pyridazines as trk inhibitors
US11072610B2 (en) 2018-09-12 2021-07-27 Novartis Ag Antiviral pyridopyrazinedione compounds
WO2020053654A1 (en) 2018-09-12 2020-03-19 Novartis Ag Antiviral pyridopyrazinedione compounds
WO2020058913A1 (en) 2018-09-21 2020-03-26 Novartis Ag Isoxazole carboxamide compounds and uses thereof
US11891401B2 (en) 2018-10-09 2024-02-06 Novartis Ag Solid forms of N-(4-fluoro-3-(6-(3-methylpyridin-2-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-2-yl)phenyl)-2,4-dimethyloxazole-5-carboxamide
JP2022504259A (en) * 2018-10-09 2022-01-13 ノバルティス アーゲー N- (4-fluoro-3- (6- (3-methylpyridine-2-yl)-[1,2,4] triazolo [1,5-A] pyrimidin-2-yl) phenyl) -2,4 -Solid form of dimethyloxazole-5-carboxamide
WO2020075046A1 (en) * 2018-10-09 2020-04-16 Novartis Ag Solid forms of n-(4-fluoro-3-(6-(3-methylpyridin-2-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-2-yl)phenyl)-2,4-dimethyloxazole-5-carboxamide
WO2020128768A1 (en) 2018-12-18 2020-06-25 Novartis Ag N-(pyridin-2-ylsulfonyl)cyclopropanecarboxamide derivatives and their use in the treatment of a cftr mediated disease
WO2020144604A1 (en) 2019-01-11 2020-07-16 Novartis Ag Lta4h inhibitors for the treatment of hidradenitis suppurativa
WO2020150626A1 (en) 2019-01-18 2020-07-23 Biogen Ma Inc. Imidazo[1,2-a]pyridinyl derivatives as irak4 inhibitors
WO2020154581A1 (en) * 2019-01-24 2020-07-30 Assia Chemical Industries Ltd Solid state forms of fedovapagon-salicyclic acid co-crystal
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
WO2020198077A1 (en) 2019-03-22 2020-10-01 Sumitomo Dainippon Pharma Oncology, Inc. Compositions comprising pkm2 modulators and methods of treatment using the same
WO2020250116A1 (en) 2019-06-10 2020-12-17 Novartis Ag Pyridine and pyrazine derivative for the treatment of cf, copd, and bronchiectasis
WO2020263967A1 (en) 2019-06-27 2020-12-30 Biogen Ma Inc. 2h-indazole derivatives and their use in the treatment of disease
WO2020263980A1 (en) 2019-06-27 2020-12-30 Biogen Ma Inc. Imidazo[1,2-a]pyridinyl derivatives and their use in the treatment of disease
US11529350B2 (en) 2019-07-03 2022-12-20 Sumitomo Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (TNK1) inhibitors and uses thereof
WO2021003417A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
WO2021038426A1 (en) 2019-08-28 2021-03-04 Novartis Ag Substituted 1,3-phenyl heteroaryl derivatives and their use in the treatment of disease
WO2021044351A1 (en) 2019-09-06 2021-03-11 Novartis Ag Methods of treating liver disease using lta4h inhibitors
WO2021060949A1 (en) * 2019-09-26 2021-04-01 대봉엘에스 주식회사 Co-crystalline efinaconazole, and method for producing same
US11667613B2 (en) 2019-09-26 2023-06-06 Novartis Ag Antiviral pyrazolopyridinone compounds
CN113795484A (en) * 2019-09-26 2021-12-14 大峰Ls株式会社 Eutectic-type efinaconazole and preparation method thereof
WO2021124172A1 (en) 2019-12-18 2021-06-24 Novartis Ag 3-(5-methoxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2021148807A1 (en) 2020-01-22 2021-07-29 Benevolentai Bio Limited Pharmaceutical compositions and their uses
WO2021148805A1 (en) 2020-01-22 2021-07-29 Benevolentai Bio Limited Topical pharmaceutical compositions comprising imidazo[1,2-b]pyridazine compounds
WO2021191169A1 (en) 2020-03-23 2021-09-30 Artemiflow GmbH 1,2,4-trioxane compounds and compositions comprising the same for use in the treatment of covid-19
EP3884938A1 (en) 2020-03-25 2021-09-29 ArtemiFlow GmbH 1,2,4-trioxane compounds and compositions comprising the same for use in the treatment of covid-19
WO2021214073A1 (en) 2020-04-20 2021-10-28 Novartis Ag Antiviral 1,3-di-oxo-indene compounds
WO2021214080A1 (en) 2020-04-20 2021-10-28 Novartis Ag Antiviral 1,3-di-oxo-indene compounds
WO2021218930A1 (en) * 2020-04-30 2021-11-04 江苏恩华药业股份有限公司 Co-amorphous substance of celecoxib and pregabalin and preparation method therefor
CN115551834B (en) * 2020-04-30 2023-11-03 苏州恩华生物医药科技有限公司 Celecoxib and pregabalin co-amorphous substance and preparation method thereof
CN113582927B (en) * 2020-04-30 2023-07-04 苏州恩华生物医药科技有限公司 Celecoxib and pregabalin co-amorphous substance and preparation method thereof
CN115551834A (en) * 2020-04-30 2022-12-30 苏州恩华生物医药科技有限公司 Celecoxib and pregabalin co-amorphous compound and preparation method thereof
CN113582927A (en) * 2020-04-30 2021-11-02 苏州恩华生物医药科技有限公司 Celecoxib and pregabalin co-amorphous compound and preparation method thereof
US11806346B2 (en) 2020-05-13 2023-11-07 Chdi Foundation, Inc. HTT modulators for treating Huntington's disease
WO2021255607A1 (en) 2020-06-15 2021-12-23 Novartis Ag Methyl 2-(fluoromethyl)-5-oxo-4-phenyl-4,5,6,7-tetrahydro-1h-cyclopenta[b]pyridine-3-carboxylates and methyl 2-(fluoromethyl)-5-oxo-4-phenyl-1,4,5,7-tetrahydrofuro[3,4-b]pyridine-3-carboxylates as cav1.2 activators
WO2021255608A1 (en) 2020-06-16 2021-12-23 Novartis Ag Methyl 2-methyl-5-oxo-1,4,5,7-tetradhydrofuro[3,4- b]pyridine-3-carboxylate compounds as cav1.2 activators
WO2022058902A1 (en) 2020-09-17 2022-03-24 Novartis Ag Compounds and compositions as sppl2a inhibitors
CN112552189A (en) * 2020-11-10 2021-03-26 中国海洋大学 Pharmaceutical co-crystal of amantadine hydrochloride and resveratrol and preparation method thereof
CN112521292A (en) * 2020-12-18 2021-03-19 深圳市萱嘉生物科技有限公司 Eutectic crystal of betaine and organic acid and preparation method and application thereof
WO2022140415A1 (en) 2020-12-22 2022-06-30 Biogen Ma Inc. 2h-indazole derivatives as irak4 inhibitors and their use in the treatment of disease
WO2022140425A1 (en) 2020-12-22 2022-06-30 Biogen Ma Inc. Imidazo[1,2-a]pyridine derivatives as irak4 inhibitors and their use in the treatment of disease
WO2022150446A1 (en) 2021-01-07 2022-07-14 Biogen Ma Inc. Tyk2 inhibitors
WO2022195454A1 (en) 2021-03-15 2022-09-22 Novartis Ag Pyrazolopyridine derivatives and uses thereof
WO2022195355A1 (en) 2021-03-15 2022-09-22 Novartis Ag Benzisoxazole derivatives and uses thereof
WO2022201097A1 (en) 2021-03-26 2022-09-29 Novartis Ag 1,3-substituted cyclobutyl derivatives and uses thereof
WO2022217232A1 (en) 2021-04-10 2022-10-13 Sunovion Pharmaceuticals Inc. Chromans and benzofurans as 5-ht1a and taar1 agonists
WO2022217248A1 (en) 2021-04-10 2022-10-13 Sunovion Pharmaceuticals Inc. Substituted sulfonamide-chroman compounds, and pharmaceutical compositions, and methods of use thereof
WO2022219546A1 (en) 2021-04-16 2022-10-20 Novartis Ag Heteroaryl aminopropanol derivatives as inhibitors of lta4h
WO2022234287A1 (en) 2021-05-06 2022-11-10 Benevolentai Bio Limited Imidazopyridazine derivatives useful as trk inhibitors
WO2022249060A1 (en) 2021-05-26 2022-12-01 Novartis Ag Triazolo-pyrimidine analogues for treating diseases connected to the inhibiton of werner syndrome recq helicase (wrn)
WO2022254362A1 (en) 2021-06-03 2022-12-08 Novartis Ag 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and medical uses thereof
WO2023275796A1 (en) 2021-07-01 2023-01-05 Novartis Ag Heterocyclic derivatives as sphingosine-1-phosphate 3 inhibitors
WO2023002443A1 (en) 2021-07-22 2023-01-26 Novartis Ag Substituted pyridone compounds useful to treat orthomyxovirus infections
WO2023049721A1 (en) 2021-09-23 2023-03-30 Sunovion Pharmaceuticals Inc. Methods of treating metabolic disorders
CN113845438A (en) * 2021-10-12 2021-12-28 河北大学 Acetaminophen-piracetam pharmaceutical co-crystal and preparation method thereof
CN113845438B (en) * 2021-10-12 2023-02-21 河北大学 Acetaminophen-piracetam pharmaceutical co-crystal and preparation method thereof
WO2023094965A1 (en) 2021-11-23 2023-06-01 Novartis Ag Naphthyridinone derivatives for the treatment of a disease or disorder
CN114031515A (en) * 2021-11-29 2022-02-11 河北大学 Paracetamol-ibuprofen pharmaceutical co-crystal and preparation method thereof
CN114031515B (en) * 2021-11-29 2022-10-21 河北大学 Acetaminophen-ibuprofen pharmaceutical co-crystal and preparation method thereof
WO2023111799A1 (en) 2021-12-13 2023-06-22 Novartis Ag PYRIDINE-3-CARBOXYLATE COMPOUNDS AS CaV1.2 ACTIVATORS
WO2023139534A1 (en) 2022-01-24 2023-07-27 Novartis Ag Spirocyclic piperidinyl derivatives as complement factor b inhibitors and uses thereof
CN114644669A (en) * 2022-03-10 2022-06-21 华润紫竹药业有限公司 Preparation method and application of progesterone eutectic
WO2023187715A1 (en) 2022-04-01 2023-10-05 Novartis Ag Complement factor b inhibitors and uses thereof
CN114835657A (en) * 2022-06-10 2022-08-02 大连工业大学 Method for regulating and controlling ethenzamide-saccharin eutectic crystal polycrystalline type based on ionic liquid
WO2024006493A1 (en) 2022-07-01 2024-01-04 Biogen Ma Inc. Tyk2 inhibitors
WO2024028782A1 (en) 2022-08-03 2024-02-08 Novartis Ag Nlrp3 inflammasome inhibitors

Also Published As

Publication number Publication date
CA2514733A1 (en) 2004-09-16
EP1631260A2 (en) 2006-03-08
WO2004078163A3 (en) 2005-01-20
JP2007524596A (en) 2007-08-30

Similar Documents

Publication Publication Date Title
WO2004078163A2 (en) Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
JP4923182B2 (en) Celecoxib and nicotinamide co-crystal and pharmaceutical composition containing the co-crystal
US20070026078A1 (en) Pharmaceutical co-crystal compositions
US20070059356A1 (en) Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US20100311701A1 (en) Pharmaceutical Co-Crystal Compositions
US7803786B2 (en) Pharmaceutical co-crystal compositions and related methods of use
EP1755388B1 (en) Mixed co-crystals and pharmaceutical compositions comprising the same
US8436029B2 (en) Pharmaceutical forms, and methods of making and using the same
AU2005212229B2 (en) Modafinil compositions
US7566805B2 (en) Modafinil compositions
CA2534664C (en) Modafinil compositions
US8809586B2 (en) Modafinil compositions
US20060287392A1 (en) Gabapentin compositions
EP2292213A1 (en) Compositions comprising a polymorphic form of armodafinil
IL199140A (en) Modafinil compositions
ALMARSSON et al. Patent 2514733 Summary
ZA200602736B (en) Modafinil compositions
NZ548656A (en) A polymorph of R-(-)-modafinil, for use in treating sleep and other disorders

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2514733

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 10546963

Country of ref document: US

Ref document number: 2006508979

Country of ref document: JP

Ref document number: 2007059356

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2004715190

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004715190

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10546963

Country of ref document: US