WO2016054240A1 - Fixed dose combinations for the treatment of viral diseases - Google Patents

Fixed dose combinations for the treatment of viral diseases Download PDF

Info

Publication number
WO2016054240A1
WO2016054240A1 PCT/US2015/053268 US2015053268W WO2016054240A1 WO 2016054240 A1 WO2016054240 A1 WO 2016054240A1 US 2015053268 W US2015053268 W US 2015053268W WO 2016054240 A1 WO2016054240 A1 WO 2016054240A1
Authority
WO
WIPO (PCT)
Prior art keywords
salt
inhibitor
pharmaceutical composition
composition
drugs
Prior art date
Application number
PCT/US2015/053268
Other languages
French (fr)
Inventor
Sean Dalziel
Mark Menning
Original Assignee
Sean Dalziel
Mark Menning
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sean Dalziel, Mark Menning filed Critical Sean Dalziel
Publication of WO2016054240A1 publication Critical patent/WO2016054240A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • HCV Hepatitis C virus
  • HCV infection can often be medically managed. Early medical intervention is helpful, but people who are infected often experience mild symptoms, and thus do not seek treatment. Indeed, some people infected with the virus can be cleared of it with therapies that involve treatment with the appropriate anti-viral medicines.
  • HCV is an enveloped virion that contains a positive stranded RNA genome encoding all known virus-specific proteins in one single, uninterrupted, open reading frame.
  • the virus has several genotypes, 1-4, with genotype 1 being the most prevalent.
  • the open reading frame comprises approximately 9500 nucleotides encoding a single large polyprotein of about 3000 amino acids.
  • the polyprotein comprises a core protein, envelope proteins El and E2, a membrane bound protein p7, and the non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • a cellular protease cleaves the viral protein at the NS2- NS3 junction allowing a viral protease (NS3 protease) to mediate subsequent cleavages.
  • the NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. It is thought that NS2 and NS4A are involved in proteolytic activity.
  • NS5A is a phosphoprotein involved in viral replication.
  • NS5B is a RNA-dependent RNA polymerase.
  • the inhibition of the nonstructural proteins inhibits HCV replication.
  • these non-structural proteins have been targets for drug development to treat HCV infection, alone, or in combination with existing therapies. Inhibitors of these viral proteins are often referred to as "direct acting antiviral, or HCV, inhibitors.”
  • Combination drugs such as fixed-dose combinations have become increasingly prevalent in certain therapeutic areas (including cancer, cardiovascular, and infectious disease) and that these products play an important role in optimizing adherence to dosing regimens and improving patient outcomes.
  • Combination drugs such as fixed-dose combinations may be particularly beneficial for the treatment of certain viral diseases, including HCV, where a combination may comprise two or more drugs. Nevertheless, such combinations are just now being developed.
  • compositions comprising a nonstructural protein 5A (NS5A) inhibitor or salt thereof; a nonstructural protein 3/4A (NS3/4A) inhibitor or salt thereof; and a nonstructural protein 5B (NS5B) inhibitor or salt thereof.
  • NS5A nonstructural protein 5A
  • NS3/4A nonstructural protein 3/4A
  • NS5B nonstructural protein 5B
  • the NS5A inhibitor or salt thereof can comprise ACH-3102 or salt thereof.
  • the NS3/4A inhibitor or salt thereof can comprise simeprevir or salt thereof.
  • the composition can comprise further interferon.
  • the pharmaceutical composition can further comprise ribavirin or salt thereof.
  • NS5B inhibitor or salt thereof can also comprise a non-nucleoside inhibitor or salt thereof.
  • the NS5B inhibitor or salt thereof can also comprise a nucleoside inhibitor or salt thereof.
  • the pharmaceutical composition can further comprise one or more additional drugs or salts thereof.
  • the one or more additional drugs or salts thereof can be daclatasvir or salt thereof, an NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, or any combination thereof.
  • the pharmaceutical composition can be a tablet.
  • the pharmaceutical composition can comprise a sustained release composition.
  • the sustained release composition can release at least or at least about 50, 60, 70, 80, 90, 95, or 99% by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, before or before about 24 hours based on the weight of the drug of the composition.
  • the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
  • the pharmaceutical composition can comprise an extended release composition.
  • the extended release composition can release not more than or more than about 5, 10, 20, 25, or 30% after 90 minutes, from or from about 35, 40, or 45% to 50, 55, 60, or 65% after 4 hours, from or from about 50, 55, 60, or 65% to 70, 75, 80 or 85% after 8 hours; from or from about 70, 75, 80, or 85 to 90, 95, or 99% after 14 hours; and not less than or not less than about 90, 95, 97, or 99% after 24 hours by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition.
  • the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
  • the pharmaceutical composition can comprise a delayed release composition.
  • the delayed release composition can release from or from about 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 15, 20, 25, or 30% after 90 minutes by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition.
  • the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
  • the NS5A inhibitor or salt thereof, said NS3/4A inhibitor or salt thereof, and said NS5B inhibitor or salt thereof can be in separate sections or layers.
  • the pharmaceutical composition can comprise at least one immediate release, at least one extended release, or at least one delayed release section or layer.
  • the pharmaceutical composition can comprise at least two or at least three or two or three layers or sections, wherein each section or layer comprises independently an immediate release, an extended release, or a delayed release section or layer.
  • the pharmaceutical composition can further comprise a coating.
  • the coating can comprise an enteric coating.
  • the pharmaceutical composition can comprise an immediate release composition.
  • one or more active ingredients or salt thereof of the pharmaceutical composition can inhibit HCV viral replication of one or more HCV genotypes.
  • the one or more HCV genotypes can comprise a type I or IV genotype.
  • the one or more HCV genotypes can comprise a type I genotype.
  • the one or more HCV genotypes can comprise a type IV genotype.
  • the one or more HCV genotypes can comprise a type II genotype.
  • the one or more HCV genotypes can comprise a type III genotype.
  • compositions comprising simeprevir or salt thereof loaded onto silica carrier particles.
  • the silica carrier particles can comprise fumed silica.
  • the pharmaceutical composition can comprise one or more additional drugs or salts thereof.
  • the one or more additional drugs or salts thereof can comprise an NS5A inhibitor or salt thereof.
  • the NS5A inhibitor or salt thereof can also comprise ACH-3102 or salt thereof.
  • the one or more additional drugs or salts thereof can comprise an NS5B inhibitor or salt thereof.
  • the NS5B inhibitor or salt thereof can comprise a non-nucleoside inhibitor or salt thereof.
  • the NS5B inhibitor or salt thereof can comprise a nucleoside inhibitor or salt thereof.
  • the pharmaceutical composition can comprise interferon.
  • the pharmaceutical composition can also further comprise ribavirin or salt thereof.
  • the pharmaceutical compositions can also comprise the dissolution profile of the sustained release, extended release, or delayed release composition.
  • the pharmaceutical composition can also be in the form of a tablet.
  • the tablet can exhibit a hardness of less than or less than about 40, 30, 20, 10, 5, or 1 kp.
  • the pharmaceutical composition can further comprise fumed silica.
  • the NS3/4A inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS3/4A inhibitor or salt thereof can comprise from or from about 50 mg to 1000 mg. In some cases, the NS5A inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS5A inhibitor or salt thereof can comprise from or from about 10 mg to 250 mg.
  • the NS5B inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS5B inhibitor or salt thereof can comprise from or from about 10 mg to 1000 mg.
  • the pharmaceutical composition after oral administration can comprise a Cmax from or from about 1.0, 1.5, 2.0, 2.5, or 3.0 pg/ml to 5.0, 10.0, 15.0, 20.0, or 25.0 ng/ml with plasma of patients with an average distribution volume of 50 L.
  • the pharmaceutical composition after oral administration, can comprise a Tmax from or from about 30, 60, 90, 120, 150, 180, 210, or 240 minutes.
  • the pharmaceutical composition after oral administration, can comprise a Tmax from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours.
  • a pharmaceutical composition comprising a therapeutically effective amount of an NS 5 A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, and an NS5B inhibitor or salt thereof.
  • methods of treating or preventing viral infections comprising administering to a patient in need thereof the pharmaceutical composition as described throughout.
  • the viral infection can be hepatitis.
  • the viral infection can also be hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
  • HAV hepatitis A
  • HBV hepatitis B
  • HCV hepatitis C
  • HDV hepatitis D
  • HEV hepatitis E
  • HV hepatitis F
  • HGV hepatitis G
  • Disclosed herein are also methods of making a pharmaceutical composition comprising loading simeprevir or a salt thereof onto silica carrier particles to form silica carrier particles loaded with simeprevir or salt thereof.
  • the method can further comprise adding to said composition one or more additional drugs or salts thereof.
  • the one or more additional drugs can comprise an NS5A inhibitor or salt thereof.
  • the NS5A inhibitor or salt thereof can comprise ACH-3102 or salt thereof.
  • the one or more additional drugs can comprise an NS5B inhibitor or salt thereof.
  • the NS5B inhibitor or salt thereof can comprise a non-nucleoside inhibitor or salt thereof.
  • the NS5B inhibitor or salt thereof can comprise a nucleoside inhibitor or salt thereof.
  • the method can further comprise adding to the composition interferon.
  • the method can further comprise adding to the composition ribavirin or salt thereof.
  • the composition can be compressed into a tablet. In some cases, the composition can be formed into a bilayer or multilayer tablet.
  • compositions comprising two or more compositions that inhibit two or more components of Figure 1.
  • the pharmaceutical composition can also comprise three or more compositions that inhibit three or more components of Figure 1.
  • the pharmaceutical composition can also comprise four or more compositions that inhibit four or more components of Figure 1.
  • the viral infection can be hepatitis.
  • the viral infection can be hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
  • Fig. 1 shows the different components of the Hepatitis C pathway.
  • the term "fixed-dose combination" and its grammatical equivalents as used herein can refer to a formulation that can comprise two or more active pharmaceutical ingredients (APIs) combined in a single dosage form.
  • the dosages can be any type of dosage form, including but not limited to dosages forms for oral administration (for example, a pill, tablet, capsule, or other delivery vehicle).
  • targeted drug and “direct targeted drug” and their grammatical equivalents as used herein can refer to and can encompass any compound that can inhibit the activity of viral proteins involved in viral replication.
  • targeted drug can refer to inhibitors of the activity of HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • targeted drug can encompass all proteins involved in viral replication for all the genotypes of HCV.
  • immunotherapy can refer to a drug or an approach for the treatment of a disease such as viral diseases, including HCV, cancer, or other immunological disorder where the drug or treatment acts on the immune system, to harness its ability to combat the disease.
  • immunotherapy can include an antibody that affects immune function, a cell cycle checkpoint inhibitor, and other small molecule immunomodulator drugs such as thalidomide, lenalidomide, and pomalidomide.
  • the term "about” as used herein and its grammatical equivalents, in relation to a reference numerical value can include a range of values plus or minus 10% from that value.
  • the amount "about 10 " can include amounts from 9 to 11.
  • the term “about " in relation to a reference numerical value can also include a range of values plus or minus 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% from that value.
  • treating can include achieving a therapeutic benefit and/or a prophylactic benefit.
  • Therapeutic benefit can be eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit can be achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement can be observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions can be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • 'Therapeutically effective amount as used herein and its grammatical equivalents can refer to the amount of an active ingredient, with or without additional active ingredients, which can be effective to achieve its intended purpose. While individual patient needs may vary, determination of optimal ranges for effective amounts of the compounds and compositions is within the skill of an ordinary practitioner of the art. Generally, the dosage required to provide an effective amount of the composition, and which can be adjusted by one of ordinary skill in the art, can vary depending on the age, health, physical condition, sex, weight, extent of the dysfunction of the recipient, frequency of treatment and the nature, and scope of the dysfunction.
  • patient or “subject” as used herein and its grammatical equivalents can include mammals, such as humans, including those in need of treatment thereof. Depending on the context, the terms “patient “ and “subject” can sometimes be used interchangeably.
  • COMPOSITIONS [0044] Disease, for example, viral diseases, including but not limited to HCV, can be treated with a variety of different compositions. Many of the compositions that are effective to treat disease, including viral disease, have varying degree of efficacy. Many of these compositions are insoluble in water, causing unpredictability when formulating the drugs. Putting aside these formulation complications, this section focuses on the compositions that can be used to treat disease, for example, viral diseases, including but not limited to HCV. The
  • compositions, and combinations of compositions presented in this section can be used for the different types of formulations presented thought this application.
  • Peg- interferon-alpha has the advantage over interferon in that it is much slower to be eliminated from the body (longer half-life), and thus more efficacious. Nevertheless, pegylation of interferon can impart side effects to the molecule, and despite its enhanced efficacy and the treatment is often not optimal in terms of viral elimination. Further, when combined with ribavirin, the combination of peg-interferon and ribavirin has additional and undesirable side effects, such as flu-like symptoms spanning months in duration.
  • NS3/NS4A protease inhibitors
  • NS5A inhibitors NS5B polymerase inhibitors
  • Compounds that target the NS5B polymerase can be analogs or derivatives of either nucleosides, nucleotides, or other compounds that are neither of these; so called, non-nucleoside/tide inhibitors, or non-nuc inhibitors.
  • these compounds have been approved, or are being approved, in combination with known therapies. More recently, they are being approved for use alone, or in combination, with other compounds that target the aforementioned viral proteins.
  • HCV drugs that inhibit one or more of HCV's proteins
  • examples of direct acting antiviral HCV drugs that inhibit one or more of HCV's proteins include Incivek ® (telaprevir) and Victrelis ® (boceprevir), which are used in combination with interferon and ribavirin. These are being developed by Vertex and Merck Pharmaceuticals, respectively. Both drugs target the viral proteases NS3/NS4A of genotype 1 (the most common type) of the virus.
  • Olysio ® simprevir
  • Olysio ® a hepatitis C virus
  • CHC chronic hepatitis C
  • Olysio ® also targets the NS3/NS4 proteases.
  • Newer drugs that don't have to be administered with interferon and ribavirin have been, or are being developed. For example, Bristol-Myers Squibb (BMS) ( See WO
  • Daklinza-Sovaldi all-oral regimen comprising 2 single drugs administered once daily each.
  • Sovaldi ® (sofosbuvir) is a nucleotide analogue inhibitor of NS5B.
  • Sovaldi ® is approved for people with genotypes 1 and 4, in combination with interferon and ribavirin. People with genotypes 2 and 3 can use Sovaldi ® with ribavirin alone (an all oral regimen); they no longer have to take injections of interferon.
  • Abb Vie, Inc. (Abbott Pharmaceuticals) is developing dasabuvir (ABT-333), aNS5B polymerase, non-nucleotide inhibitor. See, US Patent No. 8,466,159. Dasabuvir targets genotype 1 of the virus.
  • non-nucleotide inhibitors tend to have the disadvantage of requiring that the patient take a pill more than once a day.
  • Achillion Pharmaceuticals' ACH-3102 is a nucleoside NS5A inhibitor. See, U.S. Patent 8,809,313. Achillion also has another drug in phase 1 development, ACH-3422. It is a nucleotide pro-drug of a uridine analog designed to inhibit Hepatitis HCV NS5B polymerase.
  • the aforementioned viral targeted drugs can be administered individually, that is, as single drugs.
  • combination drugs can be used.
  • Gilead developed a single, once-daily pill which combines Sovaldi ® and ledipasvir (an NS5A inhibitor).
  • Harvoni ® the ledipasvir/sofosbuvir combination, has two direct-acting antiviral drugs that interfere with HCV replication that can be used to treat patients with genotypes la or lb without PEG-interferon.
  • Harvoni ® was approved for treating chronic (lasting a long time) hepatitis C genotype 1 infection in adults, but not children.
  • DAA Direct Acting Antivirals
  • Combination drugs such as fixed-dose combination products
  • Combination drugs e.g., fixed-dose combination products may have cost advantages due to reduced co-pays for patients. Where therapeutic effects of one drug depends on concurrent dosing of another drug, a combination drugs, e.g., a fixed-dose combination can enable additive,
  • combination drugs e.g., fixed-dose combinations, such as less flexibility in fixed dose ratios, and difficulty to separate timing of one drug from another in the combination where desired.
  • combination drugs e.g., fixed-dosed combination can be beneficial.
  • the drugs listed in Table 1 can be formulated in combination drugs, e.g. fixed-drug combinations. In some instances, the formulations can lead to unexpected synergy and greater efficacy for the treatment of viral disease, including but not limited to HCV.
  • One aspect of the present invention is a method for formulating a combination drug, e.g., a fixed-dose combination for treating viral diseases comprising two or more anti-viral drugs in a single delivery vehicle, where the method comprises using excipients such as fillers, disintegrants, lubricants, surfactants, and carriers in amounts which facilitate rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • Another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination products for treating viral diseases comprising two or more direct acting anti-viral drugs, where the drugs inhibit virally encoded proteins involved in viral replication, and where the method comprises using excipients and active substances in amounts which facilitate commercially useful tablet hardness characteristics and achieve rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • a combination drug e.g., a fixed-dose combination products for treating viral diseases comprising two or more direct acting anti-viral drugs, where the drugs inhibit virally encoded proteins involved in viral replication
  • the method comprises using excipients and active substances in amounts which facilitate commercially useful tablet hardness characteristics and achieve rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • compositions as described throughout can be formulated to achieve a hardness of less than 50 kilopond (kp).
  • Other effective hardness that can be useful can include less than 45, 40, 35, 30, 25, 20, 15, 10, 5, 4, 3, 2, 1, or less than 1 kp. Achieving this level of hardness can result in rapid disintegration and dissolution characteristics that are beneficial.
  • the formulation can include one or more of the drugs or any combination, e.g., fixed-dose combination as described herein.
  • a further aspect of the present invention are methods for formulating a combination drug, e.g., a fixed-dose combination for treating viral disease comprising two or more drugs, where the drugs inhibit HCV viral replication of one or more viral genotypes by inhibiting the activity of certain proteins encoded by the HCV genome and that are involved in its replication, where those proteins encoded by the HCV genome comprise non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the method comprises using fillers, disintegrants, or excipients, or other active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • a combination drug e.g., a fixed-dose combination for treating viral disease comprising two or more drugs, where the drugs inhibit HCV viral replication of one or more viral genotypes by inhibiting the activity of certain proteins encoded by the HCV genome and that are involved in its replication, where those proteins encoded by the H
  • the formulation can include one or more drugs that inhibit NS2, NS3, NS4A, NS4B, NS5A, NS5B, and/or any combinations thereof.
  • an NS2 inhibitor can be combined with an NS3 inhibitor.
  • any one or more of an NS4A, NS4B, NS5A, or NS5B inhibitor can be added to the NS2/NS3 combination.
  • the combination can be an NS2/NS3/NS4B combination, or an
  • an NS3 inhibitor can be combined with an NS4A and inhibitor. Further, any one or more of an NS2, NS4B, NS5A, or NS5B inhibitor can be added to the NS3/NS4A combination.
  • an NS4A inhibitor can be combined with an NS4B inhibitor. Further, any one or more of an NS2, NS3, NS5A, or NS5B inhibitor can be added to the NS4A/NS4B combination.
  • an NS4B inhibitor can be combined with an NS5A inhibitor. Further, any one or more of an NS2, NS3, NS4A, or NS5B inhibitor can be added to the NS4B/NS5A combination.
  • an NS5A inhibitor can be combined with an NS5B inhibitor. Further, any one or more of an NS2, NS3, NS4A, or NS4B inhibitor can be added to the NS5A/NS5B combination.
  • Yet another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination of daclatasvir (Daklinza ® ) comprising one or more other drugs that inhibit those direct acting viral proteins that are involved in viral replication, e.g., HCV replication, where the method comprises using fillers, disintegrants, or other excipients and active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • a combination drug e.g., a fixed-dose combination of daclatasvir (Daklinza ® ) comprising one or more other drugs that inhibit those direct acting viral proteins that are involved in viral replication, e.g., HCV replication
  • the method comprises using fillers, disintegrants, or other excipients and active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • Yet another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination of simeprevir (Olysio ® ), e.g., in a tablet, comprising one or more other drugs that target and inhibit direct acting viral proteins that are involved in viral replication, e.g., HCV replication, comprising contacting simeprevir with an inorganic porous silica carrier particle.
  • a combination drug e.g., a fixed-dose combination of simeprevir (Olysio ® ), e.g., in a tablet, comprising one or more other drugs that target and inhibit direct acting viral proteins that are involved in viral replication, e.g., HCV replication, comprising contacting simeprevir with an inorganic porous silica carrier particle.
  • a combination drug e.g., a fixed-dose combination of daclatasvir with other compounds that can inhibit one or more HCV non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B
  • the combinations can comprise one or more of: daclatasvir and simeprevir; daclatasvir and sofosbuvir; daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and sofosbuvir; daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the method comprises using excipients and the active substances in amounts which facilitate commercially useful hardness for packaging, storage, and transportation, and rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a
  • Such methods of making a combination drug may also comprise ribavirin.
  • An aspect of the invention are methods for treating viral diseases, including HCV, with a combination drug, e.g., a fixed-dose combination of daclatasvir where the
  • combinations can comprise inhibitors that can inhibit the activity of one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the inhibitors can be one or more of: daclatasvir and simeprevir; daclatasvir and sofosbuvir; daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and sofosbuvir;
  • daclatasvir and ABT-333 daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the method comprises using excipient and the active substances in amounts which facilitate commercially useful tablet size, hardness, and rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • Such methods may also comprise using ribavirin.
  • Such methods for treating viral diseases can also be used in combination with other therapies, including interferons, other antiviral classes, immunotherapy, chemotherapy, or liver disease treatments for fibrosis or cirrhosis.
  • compositions of a combination drug e.g., a fixed- dose combination for treating viral diseases comprising two or more direct acting anti-viral drugs that can be formulated in a device for oral delivery, where the device comprises a pill, tablet, capsule, or film comprising excipients and active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
  • the pills or tables can be small, e.g., less than 1cm in length, circumference, and/or girth.
  • a further aspect of the invention are compositions of a combination drug, e.g., a fixed-dose combination comprising two or more drugs in a device for oral delivery, where one of the drugs is daclatasvir, and the drugs inhibit one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and the device comprises a pill, tablet, or capsule comprising excipients and active substances in amounts which facilitate the disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof. If pills, capsules, or tablets are formulated, the pills or tables can be small.
  • compositions of a combination drug e.g., a fixed-dose combination in a device for oral delivery to a patient in need thereof, comprising two or more drugs, where the drugs inhibit HCV viral replication of one or more genotypes by inhibiting the activity of proteins encoded by the HCV genome that are involved in HCV replication, where said proteins comprise non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and the device comprises a pill, tablet, or capsule comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
  • the pills or tables can be small
  • a further aspect of the invention are compositions of a combination drug, e.g., a fixed-dose combination of daclatasvir with one or more other drugs in a device for oral delivery to a patient in need thereof that inhibit HCV's non-structural proteins, NS3, and NS4A, where one of said other drugs comprises simeprevir, and the device comprises a tablet, comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
  • a combination drug e.g., a fixed-dose combination of daclatasvir with one or more other drugs in a device for oral delivery to a patient in need thereof that inhibit HCV's non-structural proteins, NS3, and NS4A, where one of said other drugs comprises simeprevir, and the device comprises a tablet, comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-
  • compositions of a combination drug e.g., a fixed-dose combination of daclatasvir in a device for oral delivery to a patient in need thereof with at least one other drug that inhibit one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, where the drugs can be one or more of:
  • daclatasvir and sofosbuvir daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and ribavirin; daclatasvir and simeprevir and sofosbuvir; daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the device comprises a tablet comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
  • compositions of a combination drug e.g., a fixed- dose combination of daclatasvir with other drugs in a device for oral delivery to a patient in need thereof, where the combinations can inhibit one or more of HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the device comprises a tablet comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
  • the combinations may also comprise ribavirin.
  • Excipients Depending on the type of formulation, one or more excipients can be used.
  • the one or more excipients can include those found in the Handbook of
  • Chlorofluorocarbons CFC
  • Chloroxylenol Chloroxylenol, Cholesterol, Citric Acid Monohydrate, coconut Oil, Colloidal Silicon Dioxide, Coloring Drugs, Copovidone, Corn Oil, Corn Starchand Pregelatinized Starch, Cottonseed Oil, Cresol, Croscarmellose Sodium, Crospovidone, Cyclodextrins, Cyclomethicone, Denatonium Benzoate, Dextrates, Dextrin, Dextrose, Dibutyl Phthalate, Dibutyl Sebacate, Diethanolamine, Diethyl Phthalate, Difluoroethane (HFC), Dimethicone, Dimethyl Ether, Dimethyl Phthalate, Dimethyl Sulfoxide,
  • Hydrophobic Colloidal Silica Hydroxyethyl Cellulose, Hydroxyethylmethyl Cellulose, Hydroxypropyl Betadex, Hydroxypropyl Cellulose, Hydroxypropyl
  • Polymethacrylates Poly(methyl vinylether/maleic anhydride), Polyoxyethylene Alkyl Ethers, Polyoxyethylene Castor Oil Derivatives, Polyoxyethylene Sorbitan Fatty Acid Esters, Polyoxyethylene Stearates, Polyoxylglycerides, Polyvinyl Acetate Phthalate, Polyvinyl Alcohol, Potassium Alginate, Potassium Alum, Potassium Benzoate, Potassium Bicarbonate, Potassium Chloride, Potassium Citrate, Potassium Hydroxide, Potassium Metabisulfite, Potassium Sorbate, Povidone, Propionic Acid, Propyl Gallate, Propylene Carbonate, Propylene Glycol, Propylene Glycol Alginate, Propylparaben, Propylparaben Sodium, Pyrrolidone, Raffinose, Saccharin, Saccharin Sodium, Safflower Oil, Saponite, Sesame Oil, Shellac, Simethicone, Sodium Acetate
  • the combination drugs e.g., the fixed-dose combinations of the invention can be delivered orally.
  • the combination drugs e.g., the fixed-dose combinations that are suitable for oral administration can be formulated as discrete dosage forms, such as, but are not limited to, pills, tablets (e.g., chewable tablets), caplets, capsules, and films.
  • Such dosage forms can contain predetermined amounts of active ingredients, and may be generally, but not totally, prepared by any methods of pharmacy. See generally, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).
  • An aspect of the instant invention is a description of novel and inventive, and previously, unknown ways to formulate certain targeted drugs that inhibit HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • Typical oral dosage forms of the invention can prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients that can be suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring drugs, preservatives, and coloring drugs.
  • excipients that can be suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro- crystalline cellulose, diluents, granulating drugs, lubricants, binders, and disintegrating drugs.
  • pills, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or non-aqueous techniques. Such dosage forms can be prepared by methods of pharmacy by skilled practitioners of the art.
  • compositions and dosage forms can be prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then optionally shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention can be present in from or from about 50 to 99 (e.g., from 50 to 99 or from about 50 to about 99) weight percent of the pharmaceutical composition or dosage form.
  • the binder or filler can also be present from or from about 20 to 30, 30 to 40, or 40 to 50, 50 to 60, 60 to 70, 70 to 80, 80 to 90, or 90 to 98 weight percent of the pharmaceutical composition.
  • the binder or filler can also be present from or from about greater than 20, 30, 40, 50, 60, 70, 80, 90, or 95, weight percent of the pharmaceutical composition.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101.TM., AVICEL-PH- 103.TM., AVICEL RC-581.TM., AVICEL-PH-105.TM. (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa.), and mixtures thereof.
  • a specific binder can be a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.TM.
  • Suitable anhydrous or low moisture excipients or additives can include AVICEL-PH- 103.TM, and Starch 1500 LM.TM.
  • Silicified microcrystalline cellulose can also be used.
  • CSD5866 can be used.
  • microcrystalline cellulose can include any of the following in Table 2.
  • Disintegrants can be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. However, tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from or from about 0.5 to 15 weight percent of disintegrant.
  • disintegrants that can be used in the formulations described herein can be from or from about 1 to 5 weight percent of disintegrant.
  • the disintegrants can be from or from about 0 to 0.1; 0.05 to 0.15; 0.125 to 0.2; 0.15 to 0.30; 0.25 to 0.5; 0.4 to 0.7; 0.6 to 0.75; 0.725 to 0.9; 0.8 to 1.0; 0.9 to 1.1; 1.0 to 1.3; 1.2 to 1.5; 1.4 to 2.0; 1.9 to 2.5; 2.4 to 3.0; 2.9 to 3.5; 3.4 to 3.8; 3.7 to 3.9; 3.85; to 5.0; 5.0 to 6.0; 6.0 to 7.0; 7.0 to 8.0; 8.0 to 9.0; 9.0 to 10.0; 10.0 to 11.0; 11.0 to 12.0; 12.0 to 13.0; 13.0 to 14.0; 14.0 to 15.0; 15.0 to 16.0; 16.0 to 17.0; 17.0 to 18.0; 18.0 to 19.0; 19.0 to 20.0; 20.0 to 21.0
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • croscarmellose sodium can be used when formulating fixed dose combinations of daclatasvir.
  • Croscarmellose sodium can be used from or from about 0.1 to 25 weight percent, for example as any of the weight percentages described above.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • lubricants that can be used include, but are not limited to, for example, a solid silica gel (AE OSIL 200 ® , manufactured by W.R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SIL.TM. (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about one weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated. However, in some cases up to 15 weight percent can be used.
  • AE OSIL 200 ® manufactured by W.R. Grace Co. of Baltimore, Md.
  • CAB-O-SIL.TM. a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.
  • lubricants in the amount of less than 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5, 0.25, or 0.1 weight percent can be used.
  • Lubricants in the amount of from or from about 0.01 to 0.1; 0.1 to 0.2; 0.2 to 0.3; 0.3 to 0.4; 0.4 to 0.5; 0.5 to 0.6; 0.6 to 0.7; 0.7 to 0.8; 0.8 to 0.9; 0.9 to 1; 1 to 2; 2 to 3; 3 to 4; 4 to 5; 5 to 6; 6 to 7; 7 to 8; 8 to 9; 9 to 10; 10 to 11; 11 to 12; 12 to 13; 13 to 14; or 14 to 15 weight percent can be used.
  • One aspect of the present invention is concerned with a process for making rapidly dissolving and dispersing dosage forms, e.g., orally consumable formulations, such as films, for the delivery of pharmaceutically active drugs and with the dosage forms so obtained.
  • dosage forms e.g., orally consumable formulations, such as films
  • ribavirin can be used as the filler.
  • ribavirin possesses desired filler properties. Without intending to be bound to any particular theory, it is thought that use of an appropriate filler in this way avoids the stalling of disintegration and dissolution events of daclatasvir in the intestinal tract of a patient which occurs when daclatasvir is formulated using standard methods. If daclatasvir is formulated without the appropriate filler, it gels up, and does not disintegrate and dissolve, significantly decreasing or eliminating absorption rates.
  • simeprevir can be combined with any suitable solid carrier, provided the resulting combination has physical properties that allow it to be more easily formulated than the parent compound.
  • suitable solid carriers can comprise kaolin, bentonite, hectorite, colloidal magnesium-aluminum silicate, silicon dioxide, magnesium trisilicate, aluminum hydroxide, magnesium hydroxide, magnesium oxide and talc.
  • the solid carrier can comprise calcium silicate (such as Zeopharm), or magnesium aluminometasilicate (aka magnesium aluminum silicate; such as Neusilin).
  • Calcium silicate can be a material containing not less than 4% calcium oxide and not less than 35% silicon dioxide. Thus, a person of skill in the art can substitute calcium silicate with a material that contains calcium oxide and silicon dioxide in the correct amounts or add them to the composition individually.
  • Magnesium aluminometasilicate can be a material that contains magnesium, aluminum, silicon, oxygen, and water. For example, magnesium
  • aluminometasilicate can contain or can contain about silicon dioxide 61.1%, magnesium oxide 13.7%, aluminum oxide 9.3%, titanium dioxide 0.1%, ferric oxide 0.9%, calcium oxide 2.7%, sodium oxide 2.9%, potassium oxide 0,3%, carbon dioxide 1.8%, and/or water of combination 7.2%.
  • Suitable silica derivatives for use in the compositions of the invention and methods for preparing such silica derivatives include those that are described in international patent application publication number WO 03/037379 and the references cited therein.
  • These silica derivatives can comprise a granular hydrophilic fumed silica that has a mean particle diameter of or of about 10 to 120 micron (e.g., 5 to 25; 20 to 50; 40 to 80; 75 to 100; 85 to 110; 100 to 120; or 110 to 125 micron) and a BET surface area of or of about 40 to 400 m 2 /g (e.g., 30 to 70; 60 to 100; 90 to 150; 130 to 180; 170 to 200; 190 to 240; 230 to 290; 280 to 320; 300 to 350; 340 to 400; 380 to 450; or 410 to 500 m 2 /g) (determined according to DIN 66 131 with nitrogen).
  • the silica derivatives can also have a pore volume of or of about 0.5 to 2.5 mL/g (e.g., 0.4 to 0.8; 0.7 to 0.9; 0.8 to 1.2; 1.1. to 1.6; 1.4 to 1.9; 1.7 to 2.2; 2.0 to 2.5; 2.3 to 3.0 mL/g), where less than or less than about 5% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%) of the overall pore volume has a pore diameter of less than or less than about 5 nm (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nm), the remainder being mesopores and macropores.
  • a pore volume of or of about 0.5 to 2.5 mL/g e.g., 0.4 to 0.8; 0.7 to 0.9; 0.8 to 1.2; 1.1. to 1.6; 1.4 to 1.9; 1.7 to 2.2; 2.0 to 2.5; 2.3 to 3.0 mL/g
  • the silica derivatives typically have a pH in the range of or about 3.6 to 8.5 (e.g., 3.5 to 4.0; 4.0 to 5.0; 5.0 to 6.0; 6.0 to 7.0; 7.0 to 8.0; or 8.0 to 9.0) and a tamped density of or of about 220 to 700 g/L ( e.g., 200 to 300; 300 to 400; 400 to 500; 500 to 600; 600 to 700; or 700 to 770 g/L).
  • a pH in the range of or about 3.6 to 8.5 e.g., 3.5 to 4.0; 4.0 to 5.0; 5.0 to 6.0; 6.0 to 7.0; 7.0 to 8.0; or 8.0 to 9.0
  • a tamped density of or of about 220 to 700 g/L e.g., 200 to 300; 300 to 400; 400 to 500; 500 to 600; 600 to 700; or 700 to 770 g/L.
  • a specific silica material that can be useful in the compositions and methods of the invention is AEROPERL ® 300 (fumed silica), which is available from Evonik Degussa AG, Dusseldorf, Germany.
  • AEROPERL ® 300 fumed silica
  • Other collodial silicon dioxide can be used, e.g., Aerosil, Cab-O-Sil, and or Wacker HDK.
  • Other materials having physical and chemical properties similar to the silica materials described herein can also be used.
  • the silica particles can have a mean grain diameter of or of about 20-40 micron (e.g., 10 to 20; 15 to 25; 20 to 30; 25 to 35; 30 to 40; 35 to 45; 40 to 50 micron), in one
  • the silica particles have a BET surface area of at least or of at least about 150 m 2 /g (e.g., 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, or 350 m 2 /g).
  • the silica particles have a BET surface area of at least 200 m2/g.
  • the silica particles have a BET surface area of at least 250 m2/g.
  • the silica particles have a BET surface area of at least 275 m2/g.
  • composition comprise simeprevir
  • the composition can be coated in the pores and on the surface of the fumed silica particles. It has been determined that up to or up to about 60% (w/w) of simeprevir can be loaded on these silica particles. Other amounts of simeprevir can be loaded on these silica particles such as up to or up to about 10, 20, 30, 40, 50, 70, 80, or 90% (w/w). This high loading capacity can be beneficial for pharmaceutical applications.
  • the weight percentage of simeprevir to the silica particles is 20% ⁇ 15%.
  • the weight percentage of simeprevir to the silica particles is 60% ⁇ 15%.
  • the weight percentage of simeprevir to the silica particles is 56% ⁇ 15%. In other embodiments of the invention, the weight percentage of simeprevir can be 10, 20, 30, 40, 50, 70, 80, or 90% ⁇ 5, 10, 15, 20, 25, or 30% (or any combinations such as 10% ⁇ 25% or 70% ⁇ 10%.
  • Simeprevir can be loaded on the solid carrier using any suitable method.
  • the term "loaded" on a solid carrier and its grammatical equivalents includes, but is not limited to drugs, e.g., simeprevir, coated with the pores and on the surface of a solid carrier.
  • simeprevir can be loaded on the solid carrier by: a) spraying a solution of the compound (e.g. a solution of the compound in dichloromethane solvent) onto the solid carrier.
  • a solution of the compound e.g. a solution of the compound in dichloromethane solvent
  • Any solvent in which simeprevir is soluble can be used.
  • the solvent can comprise a volatile organic solvent.
  • the volatile solvent can then be evaporated resulting in a dry flowable powder containing simeprevir loaded on the carrier particles.
  • One or more pharmaceutically acceptable excipients can be combined with the mixture to provide a second mixture.
  • These pharmaceutically acceptable excipients can include fillers, binders, and disintegrants, including those that were disclosed above in the excipient section.
  • fillers and disintegrants that are compatible with this aqueous process.
  • microcrystalline cellulose (filler) and croscarmellose sodium (disintegrant) were found to be particularly compatible with the subsequent aqueous granulation process.
  • the weight percentage of microcrystalline cellulose to the total weight of the second mixture is or is about 50% ⁇ 25% (e.g., 10, 20, 30, 40, 60, 70, or 80% ⁇ 5, 10, 15, 20, 25, 30, 35, or 40%). In one embodiment of the invention the weight percentage of croscarmellose sodium is or is about 2% ⁇ 2% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% ⁇ 0.5, 1, 1.5, 2.5, 3, 3.5, 4, 4.5, or 5%).
  • the second mixture can be mixed, for example, using a mechanical mixer.
  • the mixture can be further processed for tableting or capsule filling. This further processing can entail unit operations such as blending, or roller compaction, wet granulation, and/or milling. It can also entail co- formulation with other active drugs.
  • a suitable pharmaceutically acceptable lubricant/glidant e.g. , magnesium stearate, stearic acid, calcium stearate, zinc stearate, or pregelatinized starch or any disclosed in the excipients section
  • the weight percentage of magnesium stearate to the total weight of the fourth mixture can be 1% ⁇ 0.5% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% ⁇ 0.5, 1, 1.5, 2.5, 3, 3.5, 4, 4.5, or 5%).
  • liquid form preparations including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions, or solid form preparations which are intended to be converted shortly before use to liquid form
  • Emulsions can be prepared in solutions, for example, in aqueous propylene glycol solutions or can contain emulsifying drugs, for example, such as lecithin, sorbitan monooleate, or acacia.
  • Aqueous solutions can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening drugs.
  • Aqueous suspensions can be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending drugs.
  • Suitable fillers or carriers with which the compositions can be administered include agar, alcohol, fats, lactose, starch, cellulose derivatives, polysaccharides, polyvinylpyrrolidone, silica, sterile saline and the like, or mixtures thereof used in suitable amounts.
  • Solid form preparations include solutions, suspensions, and emulsions, and can contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing drugs, and the like.
  • a syrup or suspension can be made by adding the active compound to a concentrated, aqueous solution of a sugar, e.g., sucrose, to which can also be added any accessory ingredients.
  • a sugar e.g., sucrose
  • accessory ingredients can include flavoring, an drug to retard crystallization of the sugar or an drug to increase the solubility of any other ingredient, e.g., as a polyhydric alcohol, for example, glycerol or sorbitol.
  • Pharmaceutically acceptable derivative It will also be appreciated that the drugs that are administered in a fixed dose combination in a single pill can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof.
  • a pharmaceutically acceptable derivative can include, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, free- base, acid, mixtures thereof, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • the term “pharmaceutically acceptable salt” and its grammatical equivalents can refer to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a pharmaceutically acceptable salt reasonable benefit/risk ratio.
  • a “pharmaceutically acceptable salt” can mean any non-toxic salt or salt of an ester of a compound of this invention.
  • the term “inhibitorily active metabolite or residue thereof and its grammatical equivalent can refer to a metabolite or residue of the parent drug is also an inhibitor.
  • a drug or composition is a salt, it can refer to a pharmaceutically acceptable salt, including but not limited to the salts found in the handbook of
  • the drugs can be formulated into, but not limited to, hydrochloride salts, hydrobromide salts, hydroiodide salts, fumaric acid salts, maleic acid salts, amino acid salts, mineral acid salts, addition salts, nitrate salts, phosphate salts, succinate salts, maleate salts, fumarate salts, citrate salts, tartrate salts, gluconate salts, lactate salts, lactobionate salts, lauryl sulfate salts, glutamate salts, acetamidobenzoate salts, potassium salts, sodium salts, calcium salts, tromethamine salts, 2-aminoethanol salts, lysine salts, and/or arginine salts.
  • Pharmaceutically acceptable salts of the compounds of this invention suitable for inclusion in a fixed dose combination in a single pill can include those derived from suitable inorganic and organic acids and bases.
  • suitable inorganic and organic acids and bases can include those derived from suitable inorganic and organic acids and bases.
  • pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts can include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pec
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N 4" (Ci -4 alkyl) 4 salts.
  • This invention also envisions the quatemization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products can be obtained by such quatemization.
  • Representative alkali or alkaline earth metal salts can include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts can include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
  • composition e.g., fixed-dose combinations
  • a pharmaceutically acceptable carrier, adjuvant, or vehicle which, as used herein, can include any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active drugs, isotonic drugs, thickening or emulsifying drugs, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • materials which can serve as pharmaceutically acceptable carriers can include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, mesoporous silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacan
  • the weight of the composition can vary and total weight can have an effect on the effectiveness of the composition, e.g., by affecting dissolution rates.
  • the weight of the composition in whatever form, e.g., tablet, capsule, powder, pill, etc.
  • the weight of the composition can be less than or less than about 10 grams (g).
  • the composition can be less than or less than about 9, 8, 7, 6, 5, 4, 3, 2, or 1 grams.
  • the composition can be less than or less than about 3 grams.
  • the composition can be less than or less than about 1.6 grams.
  • the composition can be less than or less than about 1.2 grams.
  • the composition can be less than or less than about 0.9 grams.
  • the composition can also be from or from about 0.1 to 0.5; 0.4 to 0.9; 0.8 to 1.3; 1.2 to 1.8; 1.7 to 2.3; 2.2 to 2.8; 2.7 to 3.3; or 3.2 to 4.0 grams.
  • the volume of the composition can vary and total volume can have an effect on the effectiveness of the composition, e.g., by affecting dissolution rates.
  • the volume of the composition in whatever form, e.g., tablet, capsule, powder, pill, etc.
  • the volume of the composition can be less than or less than about 100ml.
  • the composition can be less than or less than about 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 mis.
  • the composition can be less than or less than about 3 mis.
  • the composition can be less than or less than about 1.6 mis.
  • the composition can be less than or less than about 1.2 mis.
  • the composition can be less than or less than about 0.9 mis.
  • the composition can also be from or from about 0.1 to 0.5; 0.4 to 0.9; 0.8 to 1.3; 1.2 to 1.8; 1.7 to 2.3; 2.2 to 2.8; 2.7 to 3.3; or 3.2 to 4.0 mis.
  • the weight/volume percent (mass/volume) can be important.
  • Weight volume percent can be calculated by:
  • Weight Volume percent (weight of solute (in g)/volume of solution (in mL)) X 100
  • the weight/volume can be less than or less than about 50%.
  • the compositions disclosed herein can contain a weight/volume percent of less than or less than about 45, 40, 35, 30, 25, 20, 15, 10, 5, or 1%.
  • compositions can also have a weight/volume percent of from or from about 0.1 to 0.2; 0.2 to 0.3; 0.3 to 0.4; 0.4 to 0.5; 0.5 to 0.6; 0.6 to 0.7; 0.7 to 0.8; 0.8 to 0.9; 0.9 to 1.0; 1.0 to 1.3; 1.2 to 1.4; 1.3 to 1.5; 1.4 to 2.1; 2 to 8; 5 to 10; 6 to 14; 13 to 16; 15 to 20; 19 to 30; 22 to 38; 35 to 45; or 40 to 50%.
  • compositions comprising an drug or combination of drugs of the instant disclosure.
  • Such pharmaceutical compositions can be used to treat disease such as viral disease, including hepatitis, e.g., HCV.
  • Compounds of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, transdermal patch, pulmonary, vaginal, suppository, or parenteral (including intramuscular, intraarterial, intrathecal, intradermal, intraperitoneal, subcutaneous and intravenous) administration or in a form suitable for administration by aerosolization, inhalation or insufflation.
  • oral including buccal and sub-lingual
  • parenteral including intramuscular, intraarterial, intrathecal, intradermal, intraperitoneal, subcutaneous and intravenous
  • aerosolization inhalation or insufflation
  • compositions of this disclosure are disclosed, for example, in U.S. Pat. Nos. 4,897,268, 5,075,109, 5,928,647, 5,811,128, 5,820,883, 5,853,763, 5,814,344 and 5,942,252.
  • the compound can be administered in liposomes or microspheres (or microparticles).
  • Methods for preparing liposomes and microspheres for administration to a subject are well known to those of skill in the art.
  • U.S. Pat. No. 4,789,734 the contents of which are hereby incorporated by reference, describes methods for encapsulating biological materials in liposomes. Essentially, the material is dissolved in an aqueous solution, the appropriate phospholipids and lipids added, and along with surfactants if required, and the material dialyzed or sonicated, as necessary.
  • a review of known methods is provided by G. Gregoriadis, Chapter 14, “Liposomes,” Drug Carriers in Biology and Medicine, pp. 2.sup.87- 341 (Academic Press, 1979).
  • Microspheres formed of polymers or polypeptides are well known to those skilled in the art, and can be tailored for passage through the gastrointestinal tract directly into the blood stream. Alternatively, the compound can be incorporated and the microspheres, or composite of microspheres, implanted for slow release over a period of time ranging from days to months. See, for example, U.S. Pat. Nos. 4,906,474, 4,925,673 and 3,625,214, and Jein, TIPS 19:155-157 (1998).
  • the concentration of drug can be adjusted, the pH of the solution buffered and the isotonicity adjusted to be compatible with intravenous injection, as is well known in the art.
  • the compounds of the disclosure can be formulated as a sterile solution or suspension, in suitable vehicles, well known in the art.
  • the pharmaceutical compositions can be sterilized by conventional, well-known sterilization techniques, or can be sterile filtered.
  • the resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • Suitable formulations and additional carriers are described in Remington "The Science and Practice of Pharmacy” (20th Ed., Lippincott Williams & Wilkins, Baltimore MD).
  • the drugs or their pharmaceutically acceptable salts can be provided alone or in combination with one or more other drugs or with one or more other forms.
  • a formulation can comprise one or more drugs in particular proportions, depending on the relative potencies of each drug and the intended indication. For example, in compositions for targeting two different host targets, and where potencies are similar, about a 1:1 ratio of drugs can be used.
  • the two forms can be formulated together, in the same dosage unit, e.g., in one cream, suppository, tablet, capsule, aerosol spray, or packet of powder to be dissolved in a beverage; or each form can be formulated in a separate unit, e.g., two creams, two suppositories, two tablets, two capsules, a tablet and a liquid for dissolving the tablet, two aerosol sprays, or a packet of powder and a liquid for dissolving the powder, etc.
  • a drug can be administered in combination with one or more other compounds, forms, and/or drugs, e.g., as described above.
  • Pharmaceutical compositions comprising combinations a first viral drugs, e.g., HCV drugs, with one or more other second viral drug can be formulated to comprise certain molar ratios. For example, molar ratios of about 99: 1 to about 1 : 99 of a first (viral) drug to the second (viral) drug can be used.
  • the range of molar ratios of a first (viral) drug to the second (viral) drug can be from about 80:20 to about 20:80; about 75:25 to about 25:75, about 70:30 to about 30:70, about 66:33 to about 33:66, about 60:40 to about 40:60; about 50:50; and about 90: 10 to about 10:90.
  • the molar ratio of a first (viral) drug to the second (viral) drug can be used can be about 1 :9, and in some embodiments can be about 1:1.
  • the two drugs, forms and/or compounds can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, or packet of powder to be dissolved in a beverage; or each drug, form, and/or compound can be formulated in separate units, e.g., two creams, suppositories, tablets, two capsules, a tablet and a liquid for dissolving the tablet, an aerosol spray a packet of powder and a liquid for dissolving the powder, etc.
  • the drugs and/or combinations of drugs can be administered with still other drugs.
  • the choice of drugs that can be co-administered with the drugs and/or combinations of drugs of the instant disclosure can depend, at least in part, on the condition being treated.
  • Drugs of particular use in the formulations of the present disclosure can include, for example, any drug having a therapeutic effect for a viral infection, including, e.g., drugs used to treat inflammatory conditions.
  • formulations of the instant disclosure can additionally contain one or more conventional anti-inflammatory drugs, such as an NSAID, e.g., ibuprofen, naproxen, acetaminophen, ketoprofen, or aspirin.
  • an NSAID e.g., ibuprofen, naproxen, acetaminophen, ketoprofen, or aspirin.
  • formulations of the instant disclosure can additionally contain one or more conventional hepatitis antiviral drugs as disclosed throughout.
  • formulations can additionally contain one or more supplements, such as vitamin C, E or other anti-oxidants.
  • gastroretentive formulations When formulating compounds of the disclosure for oral administration, it can be desirable to utilize gastroretentive formulations to enhance absorption from the gastrointestinal (GI) tract.
  • a formulation which is retained in the stomach for several hours can release compounds of the disclosure slowly and provide a sustained release that can be preferred in some embodiments of the disclosure. Disclosure of such gastro-retentive formulations are found in Klausner, E.A.; Lavy, E.; Barta, M.; Cserepes, E.; Friedman, M.; Hoffman, A. 2003 "Novel gastroretentive dosage forms: evaluation of gastroretentivity and its effect on levodopa in humans.” Pharm. Res.
  • compositions contain multiple sections or layers, each layer or section can independently can be designed to release the drug at different times.
  • the compounds of the disclosure can be formulated for parenteral
  • compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example, solutions in aqueous polyethylene glycol.
  • the vehicle can be chosen from those known in art to be suitable, including aqueous solutions or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • the formulation can also comprise polymer compositions which are biocompatible, biodegradable, such as poly(lactic-co- glycolic)acid. These materials can be made into micro or nanospheres, loaded with drug and further coated or derivatized to provide superior sustained release performance.
  • Vehicles suitable for periocular or intraocular injection include, for example, suspensions of therapeutic drug in injection grade water, liposomes and vehicles suitable for lipophilic substances. Other vehicles for periocular or intraocular injection are well known in the art.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition can also include a solubilizing drug and a local anesthetic such as lidocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active drug.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active drug.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • the active compound can be formulated in aqueous solutions, specifically in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer.
  • the solution can contain formulatory drugs such as suspending, stabilizing and/or dispersing drugs.
  • the active compound can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the pharmaceutical composition does not comprise an adjuvant or any other substance added to enhance the immune response stimulated by the peptide.
  • the pharmaceutical composition comprises a substance that inhibits an immune response to the peptide.
  • the drugs can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation or transcutaneous delivery (for example, subcutaneously or intramuscularly), intramuscular injection or use of a transdermal patch.
  • the drugs can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions comprising one or more drugs of the present disclosure exert local and regional effects when administered topically or injected at or near particular sites of infection.
  • Direct topical application e.g., of a viscous liquid, solution, suspension, dimethylsulfoxide (DMSO)-based solutions, liposomal formulations, gel, jelly, cream, lotion, ointment, suppository, foam, or aerosol spray, can be used for local administration, to produce for example, local and/or regional effects.
  • DMSO dimethylsulfoxide
  • compositions for such formulation include, for example, lower aliphatic alcohols, polyglycols (e.g., glycerol or polyethylene glycol), esters of fatty acids, oils, fats, silicones, and the like.
  • Such preparations can also include preservatives (e.g., p- hydroxybenzoic acid esters) and/or antioxidants (e.g., ascorbic acid and tocopherol). See also Dermatological Formulations: Percutaneous absorption, Barry (Ed.), Marcel Dekker Incl, 1983.
  • the solubility of the components of the present compositions can be enhanced by a surfactant or other appropriate co-solvent in the composition.
  • cosolvents include polysorbate 20, 60, and 80, Pluronic F68, F-84 and P-103, cyclodextrin, or other drugs known to those skilled in the art.
  • co-solvents can be employed at a level of from about 0.01% to 2% by weight.
  • the pharmaceutical dosage forms can be formulated to provide a controlled release of one or more drugs.
  • controlled release and its grammatical equivalents can refer to the release of the compound from a dosage form in which it is incorporated according to a desired profile over a period of time, e.g., an extended period of time.
  • Controlled release profiles include, for example, extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled.
  • controlled release compositions can allow delivery of an agent to a subject over an extended period of time.
  • Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response while minimizing side effects as compared to conventional rapid release dosage forms. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding short acting, immediate release preparations.
  • compositions herein can be formulated into an immediate release composition.
  • the compositions as disclosed herein can also be formulated into extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled formulations.
  • each section can be independently formulated into an extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled formulations. This can be desirable, if a first drug is required as precursor to increase the effectiveness of the second drug. Or in another case, if the same drug is formulated to be released at different areas of the gastrointestinal tract.
  • a sustained release composition e.g., in beads, can release drugs from or from about 20 to 80%, 20 to 70%, 25 to 60%, and 30 to 50%, of the drug within 2 hours.
  • a sustained release composition can release at least or at least about 50, 60, 70, 80, 90, 95, or 99% by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, before or before about 24 hours based on the weight of the drug of the composition.
  • the release profile of the sustained release compositions can often include achieving 90% after or after about 4 hours (i.e., beyond the 4 hour point of the dissolution test), 6 hours, or 8 hours. Some preferred sustained release compositions are so prolonged that they do not release 90% of the drug before or before about 10 hours, 12 hours, 20 hours, or 24 hours.
  • release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1N HC1 followed by pH 6.8 phosphate buffer, 900 ml, and 50 rptn.
  • An extended released composition can release drugs not more than or more than about 30% after 90 minutes, from or from about 45% to 60% after 4 hours, from or from about 65% to 80% after 8 hours; from or from about 80 to 90% after 14 hours; and not less than or not less than about 90% after 24 hours.
  • an extended release composition can releases not more than or more than about 5, 10, 20, 25, or 30% after 90 minutes, from or from about 35, 40, or 45% to 50, 55, 60, or 65% after 4 hours, from or from about 50, 55, 60, or 65% to 70, 75, 80 or 85% after 8 hours; from or from about 70, 75, 80, or 85 to 90, 95, or 99% after 14 hours; and not less than or not less than about 90, 95, 97, or 99% after 24 hours by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition.
  • These release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1 N HC1 followed by pH 6.8
  • a delayed released composition e.g., in beads, can release drugs from or from about 0 to 25% of the drug within 90 minutes (e.g., when inside the gastrointestinal tract).
  • a delayed release composition can release from or from about 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 15, 20, 25, or 30% after 90 minutes by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition.
  • the release profile of the delayed release compositions can often include and immediate release or sustained release after the initial hour period.
  • a composition can be coated with an enteric coating which is designed to release drug as it enters the small intestine, but not within the stomach. Any enteric coating can be used.
  • materials used for enteric coatings can include fatty acids, waxes, shellac, plastics, and plant fibers.
  • the coating is typically breached and the drug can be immediately released. This can be good for drugs that are pH sensitive. Further, the delay in drug release can be even later, e.g., within the large intestine. This delayed release can be good for those drugs that have an effect within the large intestine, in which absorption prior to the large intestine is undesirable.
  • release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1N HC1 followed by pH 6.8 phosphate buffer, 900 ml, and 50 rpm.
  • pharmacokinetics In standard pharmacokinetic studies, a sufficient number of data collection points should be included, and pharmacokinetic parameters , such as area under the blood concentration-time curve (AUC), clearance, maximum blood concentration (C max ), minimum blood concentration (C min ), time to reach maximum blood concentration (T max ), volume of distribution at steady state (Vd ss ), mean residence time (MRT), and half-life (t 1/2 ), should be determined by using model independent analytical methods.
  • the rate constant and information about the volume of distribution V 1 , Vd % , and Vd ss ) can be obtained using pharmacokinetic models like a compartment model. Models that can describe drug concentrations in a blood-versus-time profile are useful to estimate changes in blood concentrations caused by differences in dosage and dose regimen, and may be used to tailor individual dosing plans.
  • the analysis should also be extended to include a PK/PD analysis.
  • Cmax The pharmaceutical compositions described throughout can possess certain unique characteristics including Cmax.
  • the pharmaceutical compositions as described throughout can exhibit a Cmax from or from about 1.0, 1.5, 2.0, 2.5, or 3.0 pg/ml to 5.0, 10.0, 15.0, 20.0, or 25.0 ng/ml ⁇ e.g., 1.0 pg/ml to 25.0 ng/ml) with plasma of patients with an average distribution volume of 50 L, after oral administration.
  • the Cmax value can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or lOOpg/ml.
  • Cmax can also be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 ng/ml.
  • Cmax can be calculated by any standard method, e.g., the USP.
  • Cmax is the peak plasma concentration of a drug after administration.
  • Tmax The pharmaceutical compositions described throughout can possess certain unique characteristics including Tmax.
  • the pharmaceutical compositions as described throughout, can exhibit a Tmax from or from about 30, 60, 90, 120, 150, 180, 210, or 240 minutes with plasma of patients with an average distribution volume of 50 L after oral administration.
  • Tmax can also be from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, in some cases, e.g., in certain controlled release formulations.
  • Tmax can be calculated by any standard method. Tmax is the time it takes to reach Cmax.
  • the mathematical formula to calculate Tmax (ln(ka) - ln(k)) / (ka - k).
  • the compositions described herein can be used to treat disease, for example, viral disease.
  • the viral disease can be a virus that infects the liver such as the hepatitis virus.
  • Some examples of the hepatitis virus can include the virus that causes hepatitis A (HAV); hepatitis B (HBV); hepatitis C (HCV); hepatitis D (HDV); hepatitis E (HEV); hepatitis F (HFV); and/or hepatitis G (HGV).
  • Some embodiments of the present disclosure relates to methods of using pharmaceutical compositions to treat disease, such as viral disease, including hepatitis, e.g., HCV.
  • Another embodiment of the present disclosure provides methods, pharmaceutical compositions, and kits for the treatment of animal subjects.
  • the term "animal subject” and its grammatical equivalents as used herein includes humans as well as other mammals.
  • a pharmaceutical composition of the disclosure can be administered to a subject at risk of contracting a disease, such as viral disease, including hepatitis, e.g., HCV, or to a subject reporting one or more of the physiological symptoms of a disease, such as viral disease, including hepatitis, e.g., HCV, even though in some cases, the screening of the condition cannot have been made.
  • a disease such as viral disease, including hepatitis, e.g., HCV
  • a subject reporting one or more of the physiological symptoms of a disease such as viral disease, including hepatitis, e.g., HCV
  • Administration can prevent a disease, such as viral disease, including hepatitis, e.g., HCV from developing, or it can reduce, lessen, shorten and/or otherwise ameliorate the progression of a disease, such as viral disease, including hepatitis, e.g, HCV, or symptoms that develop.
  • the pharmaceutical composition can modulate or target a disease, such as viral disease, including hepatitis, e.g., HCV, associated biomarker.
  • the term modulate includes inhibition of a developmental disorder associated biomarkers or alternatively activation of a developmental disorder associated biomarkers.
  • the drugs or the combination of drugs have the ability to modify enzyme activity (including proteases).
  • the ability to reduce enzyme activity can be a measure of the potency or the activity of an drug, or combination of drugs, towards or against the enzyme or other biologically important molecular process.
  • Potency can be measured by cell free, whole cell and/or in vivo assays in terms of IC50, Ki and/or ED50 values.
  • An IC50 value represents the concentration of an drug required to inhibit enzyme activity by half (50%) under a given set of conditions.
  • a Ki value represents the equilibrium affinity constant for the binding of an inhibiting drug to the enzyme or other relevant biomolecule.
  • An ED50 value represents the dose of an drug required to affect a half-maximal response in a biological assay. Further details of these measures will be appreciated by those of ordinary skill in the art, and can be found in standard texts on biochemistry, enzymology, and the like.
  • kits that can be used to disease, such as viral disease, including hepatitis, e.g., HCV.
  • kits comprise an drug or combination of drugs that inhibits the causation agent of a disease, such as viral disease, including hepatitis, e.g., HCV and in some embodiments instructions teaching the use of the kit according to the various methods and approaches described herein.
  • kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the drug. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like.
  • Dry granulation can be used to granulate one or more drugs (a first drug) (as disclosed throughout) with intragranular excipients.
  • a first drug as disclosed throughout
  • intragranular excipients any of the excipients listed throughout can be used.
  • Exemplary excipients that can be used are fillers (e.g.,
  • microcrystalline cellulose and/or lactose
  • disintegrants e.g., croscarmellose sodium
  • lubricants e.g., magnesium stearate
  • the second drug can be blended for 3 minutes in a tumbling container, e.g., at approximately 25 rpm.
  • the blend can then be rolled and compacted on a formulation prototyping machine with counter rotating stainless steel rollers.
  • the stainless steel rollers can have or have about 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness, leading the formation of a compact.
  • These compacts can then be milled to produce granules through a screen, e.g., a stainless steel rotary screen mill of 16 mesh size (1.2 mm).
  • Other mesh sizes can be used, for example, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, 19, or 20.
  • the granules are milled until they are free flowing.
  • the free base of the drugs can also be dissolved and crystallized. These solids can then be dried and the resulting powdered blended with excipients for formulation, such as tableting.
  • compositions are tableted, they can be tableted into a bilayer drug.
  • the drugs can be configured into a bilayer using a tablet tooling (e.g., a round 1 ⁇ 4 inch tableting tool) at a compression force of or of about 2,000 lbs (e.g., 1000, 1500, 2500, or 3000 lbs).
  • the compression force can also be or be about 5,000 lbs (e.g., 4000, 4500, 5500, or 6000 lbs).
  • Tablets can have a certain thickness of for example, of or of about 0.5 to 5 mm.
  • the tablet can have a thickness of or of about 0.5, 1.0, 2.0, 3.0, 4.0, or 5.0 mm.
  • the table can also have a thickness of or of about 2.58 mm.
  • the table can also have a thickness of or of about 3.13 mm.
  • the tablet can also exhibit different hardness.
  • the hardness can be hardness of or of about less than 20 kp (e.g., less than 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 kp).
  • the hardness can also be or be about 11.8 kp.
  • the hardness can also be or be about 8.9 kp.
  • the hardness can also be or be about 2.9 kp.
  • Tablets can be tested for disintegration time in deionized water at 20 °C, to determine acceptable disintegration characteristics breaking up into dispersed granules and powder within a certain amount of time.
  • the disintegration from can vary. However, some disintegration times can be or can be about 3 minutes, 2 minutes 30 seconds, 1 minute 40 seconds, or 40 seconds.
  • two or more of the drugs can be directly bending with excipients at the same time. If there is a third drug, then this can be either directly blending with the first two drugs, or later added after it has been processed with its own excipients (e.g., in bilayer tableting).
  • one or more drugs can be formulated into an amorphous solid form.
  • amorphous solids can be produced via spray drying after dissolving in a solvent.
  • the amorphous form can improve dissolution and bioavailability.
  • amorphous drugs can be loaded onto hard readily flowable porous silica carrier particles, e.g., AeroPerl 300, by Evonik. If the silica carrier particles are used, then the drug can be dissolved with a solvent and then missed thoroughly until homogenous with the silica carrier particle.
  • AeroPerl loaded particles and then be blended with other drugs (that were processed with excipients) and further process into a desired formulation, such as a tablet.
  • Surfactants can be used as well to improve dissolution characteristics.
  • SLS sodium lauryl sulfate
  • ionic Tween 80 non-ionic lecithin types of surfactants
  • any sorbitan esters e.g., sorbitan diisostearate, sorbitan dioleate, sorbitan monoisostearate, sorbitan monolaurate, sorbitan monooleate, sorbitan monopalmiiate, sorbitan monostearate, sorbitan sesquiisostearate, sorbitan sesquioleate, sorbitan sesquistearate, sorbitan
  • the amount of surfactants used is typically less than or less than about 20% w/w. For example, less than or less than about 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1% surfactant can be used.
  • a fixed dose combination HCV treatment product was formulated and manufactured with two active drugs in a single tablet. This comprised a nucleotide NS5B class RNA polymerase inhibitor, and a NS5A class viral replication complex inhibitor. This product contained sofosbuvir (400 mg) as the NS5B inhibitor, and daclatasvir dihydrochloride (60 mg) as the NS5A inhibitor.
  • the manufacturing procedure was broken down into multiple segments: dry granulation of sofosbuvir with intragranular excipients, direct blending of daclatasvir dihydrochloride with excipients, bilayer tableting. Further unit processes such as film coating, and bottling and blister packaging are also added.
  • the stepwise procedure is detailed below.
  • Step 1 Dry Granulation, Milling, and Blending of Sofosbuvir
  • Lactose (filler) 30.0 % (w/w) • Croscarmellose sodium (disintegrant) 0.73 % (w/w)
  • sofosbuvir and dry granulation excipients were blended for 3 minutes in a tumbling container at approximately 25 rpm.
  • the blend was then roller compacted on a formulation prototyping machine with counter rotating stainless steel rollers of 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness.
  • the compacts formed were ribbon-like of 25 mm width, and had processing and mechanical properties desirable for larger-scale roller compaction processing.
  • the compacts were milled to produce granules through a stainless steel rotary screen mill of 16 mesh size (1.2 mm). The granules were readily free flowing, which would be useful for reliable processing on high-speed commercial scale tablet manufacturing equipment.
  • Step 2 Direct Blending of Daclatasvir dihydrochloride
  • Daclatasvir free base was dissolved and crystallized as the dihydrochloride salt following the procedure recorded for seed crystal preparation in the patent with US publication number: US 20090041716 Al.
  • the harvested solids were dried and the resulting powder was blended with excipients for tableting in the following mass proportions:
  • Croscarmellose sodium (disintegrant) 11.70 % (w/w)
  • Step 2 Direct Blending of Daclatasvir dihydrochloride
  • Sofosbuvir granules from #1 and daclatasvir powder from #2 above were compressed in a bilayer tableting configuration using 1 ⁇ 4-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 83 mg. Tablets had a measured thickness of 2.58 mm, and a tablet hardness of 8.9 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 3 minutes.
  • the tableting procedure of Step 3 was scaled-up in proportion to yield total individual tablet mass of 1,330 mg, 21 mm x 10 mm (length and width) for a caplet shaped tablet tooling type, and a compression pressure of 5,000 lb.
  • the tablet thickness was 7.0 mm, and was therefore comparable in dimensions to Atripla®, which is a commercial once daily oral antiviral fixed-dose combination product.
  • the active substance strengths of the bilayers corresponded to 400mg sofosbuvir and 60 mg daclatasvir parent, which are respectively the recommended daily doses for these active substances for purposes of HCV infection treatment.
  • a fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained viralogic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • a fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks.
  • the patient concurrently is administered 500 mg ribavirin twice daily.
  • the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • Step 1 Direct Blending of sofosbuvir and ribavirin
  • Sofosbuvir nucleotide NS5B active substance 20.0 % (w/w)
  • Ribavirin nucleoside active substance 49.60 % (w/w)
  • Microcrystalline cellulose 29.44 % (w/w)
  • Croscarmellose sodium (disintegrant) 0.74 % (w/w)
  • Step 2 Direct Blending of Daclatasvir dihydrochloride
  • Daclatasvir free base was dissolved and crystallized as the dihydrochloride salt following the procedure recorded for seed crystal preparation in the patent with US publication number: US 20090041716 Al.
  • the harvested solids were dried and the resulting powder was blended with excipients for tableting in the following mass proportions:
  • Sofosbuvir/ribavirin excipient blend from Step 1 and daclatasvir powder from Step 2 above were compressed in a bilayer tableting configuration using 1 ⁇ 4-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 73 mg. Tablets had a measured thickness of 2.69 mm, and a tablet hardness of 2.9 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 40 seconds.
  • the tableting procedure of Step 3 is scaled-up in proportion to yield total individual tablet mass of 1,330 mg, 21 mm x 10 mm (length and width) for a caplet shaped tablet tooling type, and a compression pressure of 5,000 lb.
  • the active substance strengths of the bilayers correspond to 200mg sofosbuvir, 500 mg ribavirin, and 30 mg daclatasvir parent, which the appropriate doses for this product to be taken twice daily for HCV infection treatment.
  • sofosbuvir/daclatasvir/ribavirin (200mg / 30mg / 250 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • Example 2a NS5B/NS3/4A Tableting
  • a fixed dose combination HCV treatment product was formulated and manufactured with two active drugs in a single tablet. This comprised a nucleotide NS5B class RNA polymerase inhibitor, and a NS3/4A class serine protease inhibitor. This product contained sofosbuvir (400 mg) as the NS5B inhibitor, and simeprevir sodium (150 mg) as the NS3/4A inhibitor.
  • the manufacturing procedure was broken down into multiple segments: dry granulation of sofosbuvir with intragranular excipients, loading of amorphous simeprevir sodium onto AeroPerl 300 porous silica carrier particles, direct blending of simeprevir- AeroPerl particles with excipients, bilayer tableting. Further unit processes such as film coating, and bottling and blister packaging can be conceived. The stepwise procedure is detailed below.
  • Step 1 Dry Granulation, Milling, and Blending of Sofosbuvir
  • Lactose (filler) 30.0 % (w/w)
  • Croscarmellose sodium (disintegrant) 0.73 % (w/w)
  • sofosbuvir and dry granulation excipients were blended for 3 minutes in a tumbling container at approximately 25 rpm.
  • the blend was then roller compacted on a formulation prototyping machine with counter rotating stainless steel rollers of 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness.
  • the compacts formed were ribbon-like of 25 mm width, and had processing and mechanical properties desirable for larger-scale roller compaction processing.
  • the compacts were milled to produce granules through a stainless steel rotary screen mill of 16 mesh size (1.2 mm). The granules were readily free flowing, which would be useful for reliable processing on high-speed commercial scale tablet manufacturing equipment.
  • Step 2 Loading Simeprevir on Silica AeroPerl Carrier Particles
  • simeprevir sodium exists as capsules (rather than tablets) and uses the amorphous solid form of simeprevir sodium produced via spray drying from dichloromethane solvent.
  • the amorphous form improves dissolution and bioavailability.
  • Our attempts to prepare tablets with amorphous simeprevir were hindered by a material property where the amorphous solid form of simeprevir tended to compress in a plastic manner under tablet compression forces, yielding tablets that were highly resilient against disintegration, dispersion, and dissolution in aqueous media.
  • amorphous simeprevir sodium was loaded onto hard readily flowable porous silica carrier particles.
  • AeroPerl 300 by Evonik, is an example carrier of this type and available for pharmaceutical use.
  • Simeprevir sodium was dissolved dichloromethane (the same solvent as used in commercial amorphous simeprevir manufacture, except there by a spray drying process). The concentration of simeprevir sodium in solution was 49.5 mg/mL.
  • the simeprevir solution was added to unloaded silica AeroPerl 300 particles, thoroughly mixed until homogeneous, and the solvent evaporated to dryness.
  • the resulting dry powder contained amorphous simeprevir sodium loaded onto AeroPerl silica carrier particles.
  • the material properties included a dense highly free flowing powder containing hard, small particles that were amenable to tableting with desirable disintegration and dispersion characteristics.
  • the particles contained 0.56 g simeprevir sodium per gram of powder. Other loading levels (both higher and lower) are conceivable.
  • Step 3 Direct Blending of Simeprevir AeroPerl Particles with Excipients
  • Sofosbuvir granules from Step 1 and simeprevir sodium powder from Step 3 above were compressed in a bilayer tableting configuration using 1 ⁇ 4-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 109 mg. Tablets had a measured thickness of 3.13 mm, and a tablet hardness of 11.8 kp. Tablets were tested for disintegration time in deionized water at 20 °C and 37 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 2 minutes 30 seconds, and within 1 minute 40 seconds respectively.
  • Microcrystalline cellulose (filler) 60.46 % (w/w)
  • the tablet thickness was 8.6 mm, and was therefore comparable in dimensions to Atripla®, which is a commercial once daily oral antiviral fixed-dose combination product.
  • the active substance strengths of the bilayers corresponded to 400mg sofosbuvir and 150 mg simeprevir parent, which are respectively the recommended daily doses for these active substances for purposes of HCV infection treatment.
  • a fixed-dose combination of sofosbuvir/Simeprevir (400mg / 150mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained viralogic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • a fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks.
  • the patient is also administered 500 mg ribavirin twice daily.
  • the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • a fixed dose combination tablet containing sofosbuvir 200mg, simeprevir 75mg, and ribavirin 500 mg for twice daily administration in treatment of HCV infection is prepared as follows:
  • Step 1 Blending of Ribavirin with Sofosbuvir
  • ribavirin active substance is added in a 1 : 1 mass ratio and further blended.
  • sofosbuvir/simeprevir/ribavirin (200mg / 75mg / 250 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • Example 3a NS5B /NS5A /NS3/4A Tableting
  • a fixed dose combination HCV treatment product was developed with three active drugs in a single tablet. This comprised a non-nucleoside/tide NS5B class RNA polymerase inhibitor, a NS5A class viral replication complex inhibitor, and a NS3/4A class serine protease inhibitor.
  • the active substances include dasabuvir (ABT-333) as the NS5B inhibitor, daclatasvir dihydrochloride (60 mg) as the NS5A inhibitor, and simeprevir sodium as the NS3/4A inhibitor.
  • Step 1 Blending of dasabuvir, simeprevir loaded AeroPerl, and excipients
  • Dasabuvir was crystallized as the sodium salt according to the procedure of patent US 20120014913. The salt was isolated and dried. The resulting powder of dasabuvir active substance was used for further processing.
  • Microcrystalline cellulose 29.42 % (w/w)
  • Croscarmellose sodium (disintegrant) 3.27 % (w/w)
  • Step 2 Direct Blending of Daclatasvir dihydrochloride
  • Daclatasvir dihydrochloride was blended with excipients in preparation for tableting according to Example la, Step 2.
  • Step 3 Bilayer Tableting
  • Dasabuvir sodium / simeprevir sodium on AeroPerl blend from Step 1, and daclatasvir powder from Step 2 above were compressed in a bilayer tableting configuration using 1 ⁇ 4-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 99 mg. Tablets had a measured thickness of 3.23 mm, and a tablet hardness of 8.0 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 2 minutes.
  • Step 4 Bilayer Tableting Scale-up
  • the materials from Steps 1 and 2 are roller compacted and granulated separately, then fed to a tableting press to manufacture fixed-dose combination tablets in a bilayer configuration where the dasabuvir/simeprevir comprise one layer, and daclatasvir comprises the second layer.
  • the mass ratios are 758mg : 152mg respectively.
  • the tablets are
  • the tablets are coated with 36mg (dry weight) of a polymeric film comprising hypromellose, titanium dioxide, and polyethylene glycol 400, in a pan coater.
  • Example 3b Dasabuvir/Simeprevir/Daclatasvir Fixed Dose Combination Tablet Use
  • dasabuvir/simeprevir/daclatasvir (250mg / 75mg / 30 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure).
  • the fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
  • Sofosbuvir active substance was weighed and then blended in specific proportions of the base formula from #1 in this procedure.
  • the tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
  • tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of sofosbuvir single drug formulation examples are tabulated and described below.
  • the disintegrant level was increased significantly to 6.70%. Surprisingly, this large increase in disintegrant level did not correlate with as much of a decrease in disintegration time as would be expected.
  • Example 4d [00219] Since disintegrant level was not the dominating parameter for disintegration at this high sofosbuvir level prototype, a soluble structured composition containing spray dried lactose (filler) was prepared at approximately equal proportions of lactose to that of microcrystalline lactose. This resulted in dilution of other excipients (disintegrant and lubricant). Surprisingly, despite there being the lowest level of disintegrant, the disintegration time was substantially decreased down to 1 minute 30 seconds. Given the different tablet hardness characteristics of lactose compared with microcrystalline cellulose, it was not surprising that the tablet hardness decreased to 8.2 kp.
  • a base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
  • Daclatasvir dihydrochloride active substance was weighed and then blended in
  • the tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
  • tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula.
  • a summary of daclatasvir dihydrochloride single drug formulation examples are tabulated and described below.
  • the base excipient formula was further functionalized using crystalline lactose monohydrate seeking to space apart the active substance particles partially with not just microcrystalline cellulose as filler, but also lactose as a hard and less compressible filler material. This did not substantially improve the disintegration characteristics.
  • a prototype was prepared using the base excipient formula and lactose, similarly to that used in Example 4d, and further functionalized by increasing the croscarmellose sodium disintegrant level substantially. This formulation had a surprising and very effective impact on reducing the tablet disintegration time down to 1 minute 30 seconds. It also had an acceptable tablet hardness
  • the most effective formulation parameter for optimizing the tablet performance of daclatasvir dihydrochloride was the disintegrant level croscarmellose sodium. This was further improved with addition of water soluble lactose monohydrate filler to the base formula with high disintegrant level, although the favorable impact of lactose was not as substantial as that of croscarmellose sodium disintegrant.
  • Daclatasvir daily dose for HCV treatment is 60mg. therefore, it was not as important to increase the active substance proportion for daclatasvir as it was for sofosbuvir in Example 4.
  • Simeprevir sodium is an amorphous active ingredient that has low aqueous solubility and poor wettability characteristics.
  • the marketed product of simeprevir sodium is an amorphous active ingredient that has low aqueous solubility and poor wettability characteristics.
  • OLYSIOTM is presented as a powder filled into a capsule.
  • OLYSIO (simeprevir, 150 mg) capsules contain the following inactive ingredients: colloidal anhydrous silica, croscarmellose sodium, lactose monohydrate, magnesium stearate and sodium lauryl sulphate.
  • the white capsule contains gelatin and titanium dioxide and is printed with ink containing iron oxide black and shellac.
  • the capsule presentation of simeprevir sodium limits the possibility to fix dose combine with other drugs for the treatment of hepatitis C infection.
  • a capsule has weight limitations because of the volume constraints of the capsule shell that are filled with loosely compacted powders. This volume constraint prohibits the addition of other drugs.
  • a tablet presentation allows for higher mass loading by compressing under high pressures to increase the density of the combined products. Developing a tablet formulation of simeprevir sodium is highly desirable to prepare for the next steps in fix dose combining with other drugs.
  • a base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
  • Model C hydraulic press using 1 ⁇ 4-inch round tooling 4.
  • the tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
  • tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula.
  • a summary of simeprevir single drug formulation examples are tabulated and described below.
  • Sodium lauryl sulfate surfactant was added at 1.96% w/w to aid in the wettability of simeprevir sodium in the tablet matrix. Surprisingly, including sodium lauryl sulfate, a component of the commercial capsule formulation, did not correlate with a decrease in disintegration time as would be expected.
  • Example 2a Step 2 details the loading of simeprevir sodium onto a silicon dioxide carrier at a ratio of 0.56 g of simeprevir sodium to 1 g of simeprevir sodium on silicon dioxide.
  • a 16.82% concentration of simeprevir sodium on silicon dioxide has the same simeprevir sodium loading as Example 5a, with the corresponding reductions in
  • microcrystalline cellulose, croscarmellose sodium and magnesium stearate Surprisingly the disintegration time reduced significantly to 20 seconds by using simeprevir sodium on silicon dioxide with no changes in any of the other ingredients.
  • Amorphous simeprevir sodium was blended at a high loading with the crystalline active ingredient ribavirin. Ribavirin also has high aqueous solubility. Surprisingly at a very low disintegrant level of 1.03% the tablet disintegrated within 40 seconds. Additionally, Example 6f, which is a compact of 100% ribavirin, took 20 minutes to fully disintegrate indicating some level of synergy for disintegration of a mixture of simeprevir sodium and a crystalline active ingredient with high aqueous solubility.
  • Simeprevir sodium loaded onto silicon dioxide enabled a tablet formulation for the poorly wetting simeprevir.
  • Simeprevir loaded onto silicon dioxide greatly improved the disintegration and dispersibility of the tablet matrix as compared to amorphous simeprevir sodium in the powdered state.
  • simeprevir sodium formulation compositions were determined for the reported fixed-dose combination examples.
  • a base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
  • the tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
  • tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of dasabuvir single drug formulation examples are tabulated and described below.
  • Example 7a Improving on the prototype and observations in Example 7a, a surfactant was added to the base formula mixture with dasabuvir. Sodium lauryl sulfate (SLS) was used for this.
  • SLS Sodium lauryl sulfate
  • FBRM measurements for Example 7b indicated a finer dispersion of particles coincident with use of SLS surfactant as an excipient to improve the wettability of the dasabuvir active substance. This resulted in maintaining acceptable hardness, and disintegration characteristics of the tablet.
  • surfactants for example ionic Tween 80, and non-ionic lecithin types of surfactants
  • ionic Tween 80 and non-ionic lecithin types of surfactants

Abstract

The present invention relates to formulations including combination drugs, e.g., fixed-dose combinations, methods for formulating including combination drugs, e.g., fixed-dose combinations comprising two or more drugs in a single delivery vehicle, and methods for treating diseases, including viral disease, with formulation including combination drugs, e.g., fixed-dose combinations. The invention also provides for pharmaceutically acceptable combination drugs, e.g., fixed-dose combinations for administration, including oral administration, for the treatment of various diseases, such as, viral diseases, viral conditions, or viral disorders. The invention also provides for pharmaceutically acceptable drugs including combination drugs, e.g., fixed-dose combinations for the treatment of viral diseases caused by RNA viruses, including Hepatitis C (HCV). The invention also provides for compositions with drugs loaded onto carrier particles, and novel methods for formulating drugs onto carrier particles, which can also be further used in single drugs or combination drugs, e.g., fixed-dose combinations.

Description

FIXED DOSE COMBINATIONS FOR THE TREATMENT OF VIRAL DISEASES
CROSS REFERENCE TO RELATED APPLICATION
[0001] This application claims the benefit of U.S. Provisional Application No. 62/071,716, filed on September 30, 2014, which is incorporated herein by reference in its entirety.
BACKGROUND
[0002] Millions of people around the world are chronically infected with viral disease, such as Hepatitis C virus (HCV). The primary effect of HCV is liver inflammation, or hepatitis, that is often asymptomatic, but chronic infection can result in cirrhosis of the liver, and liver cancer. The virus is spread by blood-to-blood contact with an infected person's blood.
[0003] The symptoms of HCV infection can often be medically managed. Early medical intervention is helpful, but people who are infected often experience mild symptoms, and thus do not seek treatment. Indeed, some people infected with the virus can be cleared of it with therapies that involve treatment with the appropriate anti-viral medicines.
[0004] HCV is an enveloped virion that contains a positive stranded RNA genome encoding all known virus-specific proteins in one single, uninterrupted, open reading frame. The virus has several genotypes, 1-4, with genotype 1 being the most prevalent. The open reading frame comprises approximately 9500 nucleotides encoding a single large polyprotein of about 3000 amino acids. The polyprotein comprises a core protein, envelope proteins El and E2, a membrane bound protein p7, and the non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B. Upon infection, a cellular protease cleaves the viral protein at the NS2- NS3 junction allowing a viral protease (NS3 protease) to mediate subsequent cleavages. The NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. It is thought that NS2 and NS4A are involved in proteolytic activity. NS5A is a phosphoprotein involved in viral replication. NS5B is a RNA-dependent RNA polymerase. The inhibition of the nonstructural proteins inhibits HCV replication. Thus, these non-structural proteins have been targets for drug development to treat HCV infection, alone, or in combination with existing therapies. Inhibitors of these viral proteins are often referred to as "direct acting antiviral, or HCV, inhibitors."
[0005] The FDA recognizes that combination drugs, such as fixed-dose combinations have become increasingly prevalent in certain therapeutic areas (including cancer, cardiovascular, and infectious disease) and that these products play an important role in optimizing adherence to dosing regimens and improving patient outcomes. [0006] Combination drugs, such as fixed-dose combinations may be particularly beneficial for the treatment of certain viral diseases, including HCV, where a combination may comprise two or more drugs. Nevertheless, such combinations are just now being developed. In part, this is due to having to define the proper doses of the drugs to be combined, and solving critical technical issues having to do with material physical and chemical properties, such as differential solubility, granulation, powder compressibility, tablet disintegration, and achieving tablet of acceptable size to be swallowed, yet containing a full dose payload of active substances and necessary excipients. Resolving these complex technical issues permits combining antiviral drugs, such as anti-HCV drugs in a combination drug, such as a fixed- dose combination product. The present invention describes surprising and unexpected conditions and materials that permit the fixed dose formulation of certain compounds that target a variety of proteases and proteins that are responsible for its replication.
INCORPORATION BY REFERENCE
[0007] All publications, patents, and patent applications herein are incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event of a conflict between a term herein and a term in an incorporated reference, the term herein controls.
SUMMARY OF THE INVENTION
[0008] Disclosed herein are pharmaceutical compositions comprising a nonstructural protein 5A (NS5A) inhibitor or salt thereof; a nonstructural protein 3/4A (NS3/4A) inhibitor or salt thereof; and a nonstructural protein 5B (NS5B) inhibitor or salt thereof.
[0009] In some cases, the NS5A inhibitor or salt thereof can comprise ACH-3102 or salt thereof._In other cases, the NS3/4A inhibitor or salt thereof can comprise simeprevir or salt thereof. In some cases, the composition can comprise further interferon. The pharmaceutical composition can further comprise ribavirin or salt thereof. In some cases, NS5B inhibitor or salt thereof can also comprise a non-nucleoside inhibitor or salt thereof. The NS5B inhibitor or salt thereof can also comprise a nucleoside inhibitor or salt thereof.
[0010] In certain instances, the pharmaceutical composition can further comprise one or more additional drugs or salts thereof. In some cases, the one or more additional drugs or salts thereof can be daclatasvir or salt thereof, an NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, or any combination thereof.
[0011] The pharmaceutical composition can be a tablet.
[0012] In some cases, the pharmaceutical composition can comprise a sustained release composition. In certain instances, the sustained release composition can release at least or at least about 50, 60, 70, 80, 90, 95, or 99% by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, before or before about 24 hours based on the weight of the drug of the composition. In certain cases, the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
[0013] In some cases, the pharmaceutical composition can comprise an extended release composition. In some instances, the extended release composition can release not more than or more than about 5, 10, 20, 25, or 30% after 90 minutes, from or from about 35, 40, or 45% to 50, 55, 60, or 65% after 4 hours, from or from about 50, 55, 60, or 65% to 70, 75, 80 or 85% after 8 hours; from or from about 70, 75, 80, or 85 to 90, 95, or 99% after 14 hours; and not less than or not less than about 90, 95, 97, or 99% after 24 hours by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition. In some cases, the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
[0014] In some cases, the pharmaceutical composition can comprise a delayed release composition. In some cases, the delayed release composition can release from or from about 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 15, 20, 25, or 30% after 90 minutes by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition. In some cases, the release can be determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle.
[0015] In some instances, the NS5A inhibitor or salt thereof, said NS3/4A inhibitor or salt thereof, and said NS5B inhibitor or salt thereof, can be in separate sections or layers. In some cases, the pharmaceutical composition can comprise at least one immediate release, at least one extended release, or at least one delayed release section or layer. In some cases, the pharmaceutical composition can comprise at least two or at least three or two or three layers or sections, wherein each section or layer comprises independently an immediate release, an extended release, or a delayed release section or layer.
[0016] In some cases, the pharmaceutical composition can further comprise a coating. In some cases, the coating can comprise an enteric coating. In some cases, the pharmaceutical composition can comprise an immediate release composition.
[0017] In some cases, one or more active ingredients or salt thereof of the pharmaceutical composition can inhibit HCV viral replication of one or more HCV genotypes. The one or more HCV genotypes can comprise a type I or IV genotype. The one or more HCV genotypes can comprise a type I genotype. The one or more HCV genotypes can comprise a type IV genotype. The one or more HCV genotypes can comprise a type II genotype. The one or more HCV genotypes can comprise a type III genotype.
[0018] Further disclosed herein are pharmaceutical compositions comprising simeprevir or salt thereof loaded onto silica carrier particles. In some instances, the silica carrier particles can comprise fumed silica.
[0019] In some instances, the pharmaceutical composition can comprise one or more additional drugs or salts thereof. In some cases, the one or more additional drugs or salts thereof can comprise an NS5A inhibitor or salt thereof. The NS5A inhibitor or salt thereof can also comprise ACH-3102 or salt thereof. _In some cases, the one or more additional drugs or salts thereof can comprise an NS5B inhibitor or salt thereof. In some instances, the NS5B inhibitor or salt thereof can comprise a non-nucleoside inhibitor or salt thereof. In some instances, the NS5B inhibitor or salt thereof can comprise a nucleoside inhibitor or salt thereof. [0020] In some cases, the pharmaceutical composition can comprise interferon. The pharmaceutical composition can also further comprise ribavirin or salt thereof. The pharmaceutical compositions can also comprise the dissolution profile of the sustained release, extended release, or delayed release composition.
[0021] The pharmaceutical composition can also be in the form of a tablet. In some cases, the tablet can exhibit a hardness of less than or less than about 40, 30, 20, 10, 5, or 1 kp.
[0022] In some embodiments, the pharmaceutical composition can further comprise fumed silica.
[0023] In some cases, the NS3/4A inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS3/4A inhibitor or salt thereof can comprise from or from about 50 mg to 1000 mg. In some cases, the NS5A inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS5A inhibitor or salt thereof can comprise from or from about 10 mg to 250 mg. In some cases, the NS5B inhibitor or salt thereof can range from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition. In some cases, the NS5B inhibitor or salt thereof can comprise from or from about 10 mg to 1000 mg.
[0024] In some embodiments, after oral administration the pharmaceutical composition can comprise a Cmax from or from about 1.0, 1.5, 2.0, 2.5, or 3.0 pg/ml to 5.0, 10.0, 15.0, 20.0, or 25.0 ng/ml with plasma of patients with an average distribution volume of 50 L. In other embodiments, after oral administration, the pharmaceutical composition can comprise a Tmax from or from about 30, 60, 90, 120, 150, 180, 210, or 240 minutes. In another embodiment, after oral administration, the pharmaceutical composition can comprise a Tmax from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours.
[0025] Further disclosed herein are methods of treating or preventing a viral infection comprising administering to a patient in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of an NS 5 A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, and an NS5B inhibitor or salt thereof. [0026] Also disclosed herein are methods of treating or preventing viral infections comprising administering to a patient in need thereof the pharmaceutical composition as described throughout.
[0027] In some cases, the viral infection can be hepatitis. The viral infection can also be hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
[0028] Disclosed herein are also methods of making a pharmaceutical composition comprising loading simeprevir or a salt thereof onto silica carrier particles to form silica carrier particles loaded with simeprevir or salt thereof. The method can further comprise adding to said composition one or more additional drugs or salts thereof. The one or more additional drugs can comprise an NS5A inhibitor or salt thereof. The NS5A inhibitor or salt thereof can comprise ACH-3102 or salt thereof. _The one or more additional drugs can comprise an NS5B inhibitor or salt thereof. The NS5B inhibitor or salt thereof can comprise a non-nucleoside inhibitor or salt thereof. The NS5B inhibitor or salt thereof can comprise a nucleoside inhibitor or salt thereof. The method can further comprise adding to the composition interferon. The method can further comprise adding to the composition ribavirin or salt thereof.
[0029] In some cases, the composition can be compressed into a tablet. In some cases, the composition can be formed into a bilayer or multilayer tablet.
[0030] Further disclosed herein are pharmaceutical compositions comprising two or more compositions that inhibit two or more components of Figure 1. The pharmaceutical composition can also comprise three or more compositions that inhibit three or more components of Figure 1. The pharmaceutical composition can also comprise four or more compositions that inhibit four or more components of Figure 1.
[0031] Also disclosed herein uses of the pharmaceutical composition as described throughout, for the manufacture of a medicament for the treatment of viral disease in a subject in need thereof. The viral infection can be hepatitis. The viral infection can be hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
BRIEF DESCRIPTION OF THE DRAWINGS [0032] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
[0033] Fig. 1 shows the different components of the Hepatitis C pathway.
DETAILED DESCRIPTION OF THE INVENTION
[0034] The following description and examples illustrate embodiments of the invention in detail. It is to be understood that this invention is not limited to the particular embodiments described herein and as such can vary. Those of skill in the art will recognize that there are numerous variations and modifications of this invention, which are encompassed within its scope.
DEFINITIONS
[0035] The following definitions will aid in understanding the present invention.
[0036] The term "fixed-dose combination" and its grammatical equivalents as used herein can refer to a formulation that can comprise two or more active pharmaceutical ingredients (APIs) combined in a single dosage form. The dosages can be any type of dosage form, including but not limited to dosages forms for oral administration (for example, a pill, tablet, capsule, or other delivery vehicle).
[0037] The term "targeted drug" and "direct targeted drug" and their grammatical equivalents as used herein can refer to and can encompass any compound that can inhibit the activity of viral proteins involved in viral replication. For example, the term "targeted drug" can refer to inhibitors of the activity of HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B. The term "targeted drug" can encompass all proteins involved in viral replication for all the genotypes of HCV.
[0038] The term "immunotherapy" and its grammatical equivalents as used herein can refer to a drug or an approach for the treatment of a disease such as viral diseases, including HCV, cancer, or other immunological disorder where the drug or treatment acts on the immune system, to harness its ability to combat the disease. For example, immunotherapy can include an antibody that affects immune function, a cell cycle checkpoint inhibitor, and other small molecule immunomodulator drugs such as thalidomide, lenalidomide, and pomalidomide. [0039] The term "about" as used herein and its grammatical equivalents, in relation to a reference numerical value can include a range of values plus or minus 10% from that value. For example the amount "about 10 " can include amounts from 9 to 11. The term "about " in relation to a reference numerical value can also include a range of values plus or minus 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% from that value.
[0040] The term "treating" as used herein and its grammatical equivalents can include achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit can be eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit can be achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement can be observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions can be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
[0041] 'Therapeutically effective amount" as used herein and its grammatical equivalents can refer to the amount of an active ingredient, with or without additional active ingredients, which can be effective to achieve its intended purpose. While individual patient needs may vary, determination of optimal ranges for effective amounts of the compounds and compositions is within the skill of an ordinary practitioner of the art. Generally, the dosage required to provide an effective amount of the composition, and which can be adjusted by one of ordinary skill in the art, can vary depending on the age, health, physical condition, sex, weight, extent of the dysfunction of the recipient, frequency of treatment and the nature, and scope of the dysfunction.
[0042] The terms "patient" or "subject" as used herein and its grammatical equivalents can include mammals, such as humans, including those in need of treatment thereof. Depending on the context, the terms "patient " and "subject" can sometimes be used interchangeably.
[0043] The term "combination thereof" as used herein and its grammatical equivalents can refer to one or more members of the recited group. For example, if the group comprises A, B, or any combination thereof, each of A individually, B individually, and the group A and B are contemplated.
COMPOSITIONS [0044] Disease, for example, viral diseases, including but not limited to HCV, can be treated with a variety of different compositions. Many of the compositions that are effective to treat disease, including viral disease, have varying degree of efficacy. Many of these compositions are insoluble in water, causing unpredictability when formulating the drugs. Putting aside these formulation complications, this section focuses on the compositions that can be used to treat disease, for example, viral diseases, including but not limited to HCV. The
compositions, and combinations of compositions presented in this section can be used for the different types of formulations presented thought this application.
DRUGS
[0045] Traditionally, viral disease, such as HCV, was treated essentially with injections of interferon, or peg-interferon-alpha in combination with orally administered ribavirin. Peg- interferon-alpha has the advantage over interferon in that it is much slower to be eliminated from the body (longer half-life), and thus more efficacious. Nevertheless, pegylation of interferon can impart side effects to the molecule, and despite its enhanced efficacy and the treatment is often not optimal in terms of viral elimination. Further, when combined with ribavirin, the combination of peg-interferon and ribavirin has additional and undesirable side effects, such as flu-like symptoms spanning months in duration.
[0046] More promising approaches for the treatment of viral disease, such as HCV infection, has come from targeting of specific proteins encoded by the viral genome. To date, compounds have been, or are being developed, that target: NS3/NS4A, protease inhibitors, NS5A inhibitors, and NS5B polymerase inhibitors. Compounds that target the NS5B polymerase can be analogs or derivatives of either nucleosides, nucleotides, or other compounds that are neither of these; so called, non-nucleoside/tide inhibitors, or non-nuc inhibitors. Initially these compounds have been approved, or are being approved, in combination with known therapies. More recently, they are being approved for use alone, or in combination, with other compounds that target the aforementioned viral proteins.
[0047] Examples of direct acting antiviral HCV drugs that inhibit one or more of HCV's proteins include Incivek® (telaprevir) and Victrelis® (boceprevir), which are used in combination with interferon and ribavirin. These are being developed by Vertex and Merck Pharmaceuticals, respectively. Both drugs target the viral proteases NS3/NS4A of genotype 1 (the most common type) of the virus. Another drug, Olysio® (simeprevir) (by Johnson and Johnson), is a hepatitis C virus (HCV) NS3/4A protease inhibitor indicated for the treatment of chronic hepatitis C (CHC) genotype 1 infection as a component of a combination antiviral treatment regimen. Olysio® also targets the NS3/NS4 proteases. [0048] Newer drugs that don't have to be administered with interferon and ribavirin have been, or are being developed. For example, Bristol-Myers Squibb (BMS) ( See WO
2004/014852), has described iminothiazolidinones, including fused-bicyclic derivatives of 2- (4-aminophenyl)-5H-thiazolo[2,3-6]quinazolin-3-one, as NS5A-protein-inhibitors proposed to prevent HCV replication, alone, or in combination (See, WO 2004/014313) with other inhibitors known to interfere with HCV function. BMS recently received European approval for its NS5A inhibitor, Daklinza® (daclatasvir dihydrochloride) for use in tandem with Gilead's Sovaldi® (sofosbuvir). BMS is now running further clinical trials with this
Daklinza-Sovaldi all-oral regimen comprising 2 single drugs administered once daily each.
[0049] Sovaldi® (sofosbuvir) is a nucleotide analogue inhibitor of NS5B. The use of Sovaldi® is approved for people with genotypes 1 and 4, in combination with interferon and ribavirin. People with genotypes 2 and 3 can use Sovaldi® with ribavirin alone (an all oral regimen); they no longer have to take injections of interferon.
[0050] Abb Vie, Inc., (Abbott Pharmaceuticals) is developing dasabuvir (ABT-333), aNS5B polymerase, non-nucleotide inhibitor. See, US Patent No. 8,466,159. Dasabuvir targets genotype 1 of the virus. However, non-nucleotide inhibitors tend to have the disadvantage of requiring that the patient take a pill more than once a day.
[0051] Achillion Pharmaceuticals' ACH-3102 is a nucleoside NS5A inhibitor. See, U.S. Patent 8,809,313. Achillion also has another drug in phase 1 development, ACH-3422. It is a nucleotide pro-drug of a uridine analog designed to inhibit Hepatitis HCV NS5B polymerase.
[0052] Generally, the aforementioned viral targeted drugs can be administered individually, that is, as single drugs. However, combination drugs can be used. For example, Gilead developed a single, once-daily pill which combines Sovaldi® and ledipasvir (an NS5A inhibitor). Harvoni®, the ledipasvir/sofosbuvir combination, has two direct-acting antiviral drugs that interfere with HCV replication that can be used to treat patients with genotypes la or lb without PEG-interferon. Recently, Harvoni® was approved for treating chronic (lasting a long time) hepatitis C genotype 1 infection in adults, but not children.
[0053] Merck Pharmaceuticals is also developing and testing a combination therapy with MK-5172/MK-8742.
[0054] Table 1 below summarizes antivirals to treat viral infection.
TABLE 1: Direct Acting Antivirals (DAA) - Oral HCV compounds and classes
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
[0055] Combination drugs, such as fixed-dose combination products, can have numerous advantages over co-administration of their respective single drug products. These advantages can include more convenient dosing for patients that may lead to improved compliance, and thereby improved patient outcomes. Synergistic activity can also occur when multiple drugs are administered in combination therapies, however, the combination of drugs and dosing amounts are unpredictable. In cases where drug-drug interactions may occur, or where there are accidental risks of either over or under dosing, the combination drugs, e.g., fixed-dose combination products, can be beneficial since they are formulated in a manner to
accommodate the necessary dose adjustments due to metabolic or other drug interactions. This can permit simpler prescribing for physicians and reduce the risk of patients being over or under dosed with any of the drugs normally prescribed individually. Combination drugs, e.g., fixed-dose combination products may have cost advantages due to reduced co-pays for patients. Where therapeutic effects of one drug depends on concurrent dosing of another drug, a combination drugs, e.g., a fixed-dose combination can enable additive,
complementary, or synergistic efficacy to be more reliably achieved. For example, patients may be able to take a dose of one drug, but skip or delay taking another in the prescribed regimen. In this scenario, the beneficial effects of combination dosage, can be negative. There can be, in some cases, potential disadvantages associated with combination drugs, e.g., fixed-dose combinations, such as less flexibility in fixed dose ratios, and difficulty to separate timing of one drug from another in the combination where desired. However, in most cases, combination drugs, e.g., fixed-dosed combination can be beneficial.
[0056] The drugs listed in Table 1 (above) can be formulated in combination drugs, e.g. fixed-drug combinations. In some instances, the formulations can lead to unexpected synergy and greater efficacy for the treatment of viral disease, including but not limited to HCV. [0057] One aspect of the present invention is a method for formulating a combination drug, e.g., a fixed-dose combination for treating viral diseases comprising two or more anti-viral drugs in a single delivery vehicle, where the method comprises using excipients such as fillers, disintegrants, lubricants, surfactants, and carriers in amounts which facilitate rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
[0058] Another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination products for treating viral diseases comprising two or more direct acting anti-viral drugs, where the drugs inhibit virally encoded proteins involved in viral replication, and where the method comprises using excipients and active substances in amounts which facilitate commercially useful tablet hardness characteristics and achieve rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
[0059] For example, the compositions as described throughout can be formulated to achieve a hardness of less than 50 kilopond (kp). Other effective hardness that can be useful can include less than 45, 40, 35, 30, 25, 20, 15, 10, 5, 4, 3, 2, 1, or less than 1 kp. Achieving this level of hardness can result in rapid disintegration and dissolution characteristics that are beneficial. In some cases, the formulation can include one or more of the drugs or any combination, e.g., fixed-dose combination as described herein.
[0060] A further aspect of the present invention are methods for formulating a combination drug, e.g., a fixed-dose combination for treating viral disease comprising two or more drugs, where the drugs inhibit HCV viral replication of one or more viral genotypes by inhibiting the activity of certain proteins encoded by the HCV genome and that are involved in its replication, where those proteins encoded by the HCV genome comprise non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the method comprises using fillers, disintegrants, or excipients, or other active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
[0061] For example, in some cases, the formulation can include one or more drugs that inhibit NS2, NS3, NS4A, NS4B, NS5A, NS5B, and/or any combinations thereof. In some instances, an NS2 inhibitor can be combined with an NS3 inhibitor. Further, any one or more of an NS4A, NS4B, NS5A, or NS5B inhibitor can be added to the NS2/NS3 combination. For example, the combination can be an NS2/NS3/NS4B combination, or an
NS2/NS3/NS4B/NS5A combination. [0062] In some instances, an NS3 inhibitor can be combined with an NS4A and inhibitor. Further, any one or more of an NS2, NS4B, NS5A, or NS5B inhibitor can be added to the NS3/NS4A combination.
[0063] In some instances, an NS4A inhibitor can be combined with an NS4B inhibitor. Further, any one or more of an NS2, NS3, NS5A, or NS5B inhibitor can be added to the NS4A/NS4B combination.
[0064] In some instances, an NS4B inhibitor can be combined with an NS5A inhibitor. Further, any one or more of an NS2, NS3, NS4A, or NS5B inhibitor can be added to the NS4B/NS5A combination.
[0065] In some instances, an NS5A inhibitor can be combined with an NS5B inhibitor. Further, any one or more of an NS2, NS3, NS4A, or NS4B inhibitor can be added to the NS5A/NS5B combination.
[0066] Yet another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination of daclatasvir (Daklinza®) comprising one or more other drugs that inhibit those direct acting viral proteins that are involved in viral replication, e.g., HCV replication, where the method comprises using fillers, disintegrants, or other excipients and active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof.
[0067] Yet another aspect of the invention are methods for formulating a combination drug, e.g., a fixed-dose combination of simeprevir (Olysio®), e.g., in a tablet, comprising one or more other drugs that target and inhibit direct acting viral proteins that are involved in viral replication, e.g., HCV replication, comprising contacting simeprevir with an inorganic porous silica carrier particle.
[0068] Another aspect of the invention are methods of making a combination drug, e.g., a fixed-dose combination of daclatasvir with other compounds that can inhibit one or more HCV non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, where the combinations can comprise one or more of: daclatasvir and simeprevir; daclatasvir and sofosbuvir; daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and sofosbuvir; daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the method comprises using excipients and the active substances in amounts which facilitate commercially useful hardness for packaging, storage, and transportation, and rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof. Such methods of making a combination drug, e.g., a fixed-dose combination may also comprise ribavirin. [0069] An aspect of the invention are methods for treating viral diseases, including HCV, with a combination drug, e.g., a fixed-dose combination of daclatasvir where the
combinations can comprise inhibitors that can inhibit the activity of one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the inhibitors can be one or more of: daclatasvir and simeprevir; daclatasvir and sofosbuvir; daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and sofosbuvir;
daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the method comprises using excipient and the active substances in amounts which facilitate commercially useful tablet size, hardness, and rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof. Such methods may also comprise using ribavirin. Such methods for treating viral diseases can also be used in combination with other therapies, including interferons, other antiviral classes, immunotherapy, chemotherapy, or liver disease treatments for fibrosis or cirrhosis.
[0070] Another aspect of the invention are compositions of a combination drug, e.g., a fixed- dose combination for treating viral diseases comprising two or more direct acting anti-viral drugs that can be formulated in a device for oral delivery, where the device comprises a pill, tablet, capsule, or film comprising excipients and active substances in amounts which facilitate the dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof. If pills, capsules, or tablets are formulated, the pills or tables can be small, e.g., less than 1cm in length, circumference, and/or girth.
[0071] A further aspect of the invention are compositions of a combination drug, e.g., a fixed-dose combination comprising two or more drugs in a device for oral delivery, where one of the drugs is daclatasvir, and the drugs inhibit one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and the device comprises a pill, tablet, or capsule comprising excipients and active substances in amounts which facilitate the disintegration and dissolution of the combination drug, e.g., the fixed-dose combination when administered to a patient in need thereof. If pills, capsules, or tablets are formulated, the pills or tables can be small.
[0072] An additional aspect of the present invention are compositions of a combination drug, e.g., a fixed-dose combination in a device for oral delivery to a patient in need thereof, comprising two or more drugs, where the drugs inhibit HCV viral replication of one or more genotypes by inhibiting the activity of proteins encoded by the HCV genome that are involved in HCV replication, where said proteins comprise non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, and the device comprises a pill, tablet, or capsule comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination. If pills, capsules, or tablets are formulated, the pills or tables can be small
[0073] A further aspect of the invention are compositions of a combination drug, e.g., a fixed-dose combination of daclatasvir with one or more other drugs in a device for oral delivery to a patient in need thereof that inhibit HCV's non-structural proteins, NS3, and NS4A, where one of said other drugs comprises simeprevir, and the device comprises a tablet, comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
[0074] Yet another aspect of the present invention are compositions of a combination drug, e.g., a fixed-dose combination of daclatasvir in a device for oral delivery to a patient in need thereof with at least one other drug that inhibit one or more of HCV's non-structural proteins, NS2, NS3, NS4A, NS4B, NS5A and NS5B, where the drugs can be one or more of:
daclatasvir and sofosbuvir; daclatasvir and simeprevir; daclatasvir and simeprevir and ribavirin; daclatasvir and simeprevir and sofosbuvir; daclatasvir and ABT-333; simeprevir and sofosbuvir; simeprevir and sofosbuvir; simeprevir and ABT-333, and where the device comprises a tablet comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination.
[0075] Further aspects of the invention are compositions of a combination drug, e.g., a fixed- dose combination of daclatasvir with other drugs in a device for oral delivery to a patient in need thereof, where the combinations can inhibit one or more of HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B, and where the device comprises a tablet comprising excipients and active substances in amounts which facilitate the rapid disintegration and dissolution of the combination drug, e.g., the fixed-dose combination. The combinations may also comprise ribavirin.
[0076] Excipients. Depending on the type of formulation, one or more excipients can be used. The one or more excipients can include those found in the Handbook of
Pharmaceutical Excipients, Sixth Edition (2009), Eds. R.C. Rowe, P.J. Shesky, and M.E. Quinn. For example, it is contemplated that the following excipients can be added separately or in any combination, to any of the drugs or combination of drugs: Acacia, Acesulfame Potassium, Acetic Acid - Glacial, Acetone, Acetyltributyl Citrate, Acetyltriethyl Citrate, Adipic Acid, Agar, Albumin, Alcohol, Alginic Acid, Aliphatic Polyesters, Alitame, Almond Oil, Alpha Tocopherol, Aluminum Hydroxide Adjuvant, Aluminum Monostearate,
Aluminum Oxide, Aluminum Phosphate Adjuvant, Ammonia Solution, Ammonium Alginate, Ammonium Chloride, Ascorbic Acid, Ascorbyl Palmitate, Aspartame, Attapulgite, Bentonite, Benzalkonium Chloride, Benzethonium Chloride, Benzoic Acid, Benzyl Alcohol, Benzyl Benzoate, Boric Acid, Bronopol, Butylated Hydroxyanisole, Butylated Hydroxytoluene, Butylene Glycol, Butylparaben, Calcium Acetate, Calcium Alginate, Calcium Carbonate, Calcium Chloride, Calcium Hydroxide, Calcium Lactate, Calcium Phosphate - Dibasic Anhydrous, Calcium Phosphate - Dibasic Dihydrate, Calcium Phosphate - Tribasic, Calcium Silicate, Calcium Stearate, Calcium Sulfate, Canola Oil, Carbomer, Carbon Dioxide, Carboxymethylcellulose Calcium, Carboxymethylcellulose Sodium, Carrageenan, Castor Oil, Castor Oil - Hydrogenated, Cellulose - Microcrystalline, Cellulose - Microcrystalline and Carboxymethylcellulose Sodium, Cellulose - Powdered, Cellulose - Silicified
Microcrystalline, Cellulose Acetate, Cellulose Acetate Phthalate, Ceratonia, Ceresin, Cetostearyl Alcohol, Cetrimide, Cetyl Alcohol, Cetylpyridinium Chloride, Chitosan, Chlorhexidine, Chlorobutanol, Chlorocresol, Chlorodifluoroethane (HCFC),
Chlorofluorocarbons (CFC), Chloroxylenol, Cholesterol, Citric Acid Monohydrate, Coconut Oil, Colloidal Silicon Dioxide, Coloring Drugs, Copovidone, Corn Oil, Corn Starchand Pregelatinized Starch, Cottonseed Oil, Cresol, Croscarmellose Sodium, Crospovidone, Cyclodextrins, Cyclomethicone, Denatonium Benzoate, Dextrates, Dextrin, Dextrose, Dibutyl Phthalate, Dibutyl Sebacate, Diethanolamine, Diethyl Phthalate, Difluoroethane (HFC), Dimethicone, Dimethyl Ether, Dimethyl Phthalate, Dimethyl Sulfoxide,
Dimethylacetamide, Disodium Edetate, Docusate Sodium, Edetic Acid, Erythorbic Acid, Erythritol, Ethyl Acetate, Ethyl Lactate, Ethyl Maltol, Ethyl Oleate, Ethyl Vanillin,
Ethylcellulose, Ethylene Glycol Stearates, Ethylene Vinyl Acetate, Ethylparaben, Fructose, Fumaric Acid, Gelatin, Glucose - Liquid, Glycerin, Glyceryl Behenate, Glyceryl Monooleate, Glyceryl Monostearate, Glyceryl Palmitostearate, Glycine, Glycofurol, Guar Gum, Hectorite, Heptafluoropropane (HFC), Hexetidine, Hydrocarbons (HC), Hydrochloric Acid,
Hydrophobic Colloidal Silica, Hydroxyethyl Cellulose, Hydroxyethylmethyl Cellulose, Hydroxypropyl Betadex, Hydroxypropyl Cellulose, Hydroxypropyl
Cellulose - Low-substituted, Hydroxypropyl Starch, Hypromellose, Hypromellose Acetate Succinate, Hypromellose Phthalate, Imidurea, Inulin, Iron Oxides, Isomalt, Isopropyl Alcohol, Isopropyl Myristate, Isopropyl Palmitate, Kaolin, Lactic Acid, Lactitol,
Lactose - Anhydrous, Lactose - Inhalation, Lactose - Monohydrate, Lactose - Monohydrate and Corn Starch, Lactose - Monohydrate and Microcrystalline Cellulose, Lactose - Monohydrate and Povidone, Lactose - Monohydrate and Powdered Cellulose, Lactose - Spray-Dried, Lanolin, Lanolin - Hydrous, Lanolin Alcohols, Laurie Acid, Lecithin, Leucine, Linoleic Acid, Macrogol 15 Hydroxystearate, Magnesium Aluminum Silicate, Magnesium Carbonate, Magnesium Oxide, Magnesium Silicate, Magnesium Stearate, Magnesium Trisilicate, Maleic Acid, Malic Acid, Maltitol, Maltitol Solution, Maltodextrin, Maltol, Maltose, Mannitol, Medium-chain Triglycerides, Meglumine, Menthol, Methionine, Methylcellulose, Methylparaben, Mineral Oil, Mineral Oil - Light, Mineral Oil and Lanolin Alcohols, Monoethanolamine, Monosodium Glutamate, Monothioglycerol, Myristic Acid, Myristyl Alcohol, Neohesperidin Dihydrochalcone, Neotame, Nitrogen, Nitrous Oxide, Octyldodecanol, Oleic Acid, Oleyl Alcohol, Olive Oil, Palmitic Acid, Paraffin, Peanut Oil, Pectin, Pentetic Acid, Petrolatum, Petrolatum and Lanolin Alcohols, Phenol, Phenoxyethanol, Phenylethyl Alcohol, Phenylmercuric Acetate, Phenylmercuric Borate, Phenylmercuric Nitrate, Phospholipids, Phosphoric Acid, Polacrilin Potassium, Poloxamer, Polycarbophil, Polydextrose, Poly (DL-Lactic Acid), Polyethylene Glycol, Polyethylene Oxide,
Polymethacrylates, Poly(methyl vinylether/maleic anhydride), Polyoxyethylene Alkyl Ethers, Polyoxyethylene Castor Oil Derivatives, Polyoxyethylene Sorbitan Fatty Acid Esters, Polyoxyethylene Stearates, Polyoxylglycerides, Polyvinyl Acetate Phthalate, Polyvinyl Alcohol, Potassium Alginate, Potassium Alum, Potassium Benzoate, Potassium Bicarbonate, Potassium Chloride, Potassium Citrate, Potassium Hydroxide, Potassium Metabisulfite, Potassium Sorbate, Povidone, Propionic Acid, Propyl Gallate, Propylene Carbonate, Propylene Glycol, Propylene Glycol Alginate, Propylparaben, Propylparaben Sodium, Pyrrolidone, Raffinose, Saccharin, Saccharin Sodium, Safflower Oil, Saponite, Sesame Oil, Shellac, Simethicone, Sodium Acetate, Sodium Alginate, Sodium Ascorbate, Sodium
Benzoate, Sodium Bicarbonate, Sodium Borate, Sodium Carbonate, Sodium Chloride, Sodium Citrate Dihydrate, Sodium Cyclamate, Sodium Formaldehyde Sulfoxylate, Sodium Hyaluronate, Sodium Hydroxide, Sodium Lactate, Sodium Lauryl Sulfate, Sodium
Metabisulfite, Sodium Phosphate - Dibasic, Sodium Phosphate - Monobasic, Sodium
Propionate, Sodium Starch Glycolate, Sodium Stearyl Fumarate, Sodium Sulfite, Sodium Thiosulfate, Sorbic Acid, Sorbitan Esters (Sorbitan Fatty Acid Esters), Sorbitol, Soybean Oil, Starch, Starch - Pregelatinized, Starch - Sterilizable Maize, Stearic Acid, Stearyl Alcohol, Sucralose, Sucrose, Sucrose Octaacetate, Sugar - Compressible, Sugar - Confectioner's, Sugar Spheres, Sulfobutylether b-Cyclodextrin, Sulfur Dioxide, Sulfuric Acid, Sunflower Oil, Suppository Bases - Hard Fat, Tagatose, Talc, Tartaric Acid, Tetrafluoroethane (HFC), Thaumatin, Thimerosal, Thymol, Titanium Dioxide, Tragacanth, Trehalose, Triacetin, Tributyl Citrate, Tricaprylin, Triethanolamine, Triethyl Citrate, Triolein, Vanillin, Vegetable Oil - Hydrogenated, Vitamin E Polyethylene Glycol Succinate, Water, Wax - Anionic Emulsifying, Wax - Carnauba, Wax - Cetyl Esters, Wax - Microcrystalline, Wax - Nonionic Emulsifying, Wax - White, Wax - Yellow, Xanthan Gum, Xylitol, Zein, Zinc Acetate, and/or Zinc Stearate.
[0077] Oral Administration. The combination drugs, e.g., the fixed-dose combinations of the invention can be delivered orally. Thus, the combination drugs, e.g., the fixed-dose combinations that are suitable for oral administration can be formulated as discrete dosage forms, such as, but are not limited to, pills, tablets (e.g., chewable tablets), caplets, capsules, and films. Such dosage forms can contain predetermined amounts of active ingredients, and may be generally, but not totally, prepared by any methods of pharmacy. See generally, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990). An aspect of the instant invention is a description of novel and inventive, and previously, unknown ways to formulate certain targeted drugs that inhibit HCV's non-structural proteins; NS2, NS3, NS4A, NS4B, NS5A and NS5B.
[0078] Typical oral dosage forms of the invention can prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients that can be suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring drugs, preservatives, and coloring drugs.
Examples of excipients that can be suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro- crystalline cellulose, diluents, granulating drugs, lubricants, binders, and disintegrating drugs.
[0079] Because of ease of administration, pills, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or non-aqueous techniques. Such dosage forms can be prepared by methods of pharmacy by skilled practitioners of the art.
[0080] In general, pharmaceutical compositions and dosage forms can be prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then optionally shaping the product into the desired presentation if necessary.
[0081] For example, a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[0082] Examples of excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
[0083] Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler in pharmaceutical compositions of the invention can be present in from or from about 50 to 99 (e.g., from 50 to 99 or from about 50 to about 99) weight percent of the pharmaceutical composition or dosage form. The binder or filler can also be present from or from about 20 to 30, 30 to 40, or 40 to 50, 50 to 60, 60 to 70, 70 to 80, 80 to 90, or 90 to 98 weight percent of the pharmaceutical composition. The binder or filler can also be present from or from about greater than 20, 30, 40, 50, 60, 70, 80, 90, or 95, weight percent of the pharmaceutical composition.
[0084] Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101.TM., AVICEL-PH- 103.TM., AVICEL RC-581.TM., AVICEL-PH-105.TM. (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa.), and mixtures thereof. A specific binder can be a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.TM. Suitable anhydrous or low moisture excipients or additives can include AVICEL-PH- 103.TM, and Starch 1500 LM.TM.
[0085] Silicified microcrystalline cellulose can also be used. For example, CSD5866 can be used.
[0086] Other microcrystalline cellulose can include any of the following in Table 2.
Table 2: Types of Microcrystalline Cellulose
Figure imgf000022_0001
[0087] Disintegrants can be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. However, tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from or from about 0.5 to 15 weight percent of disintegrant. For example, disintegrants that can be used in the formulations described herein can be from or from about 1 to 5 weight percent of disintegrant. In some cases, the disintegrants can be from or from about 0 to 0.1; 0.05 to 0.15; 0.125 to 0.2; 0.15 to 0.30; 0.25 to 0.5; 0.4 to 0.7; 0.6 to 0.75; 0.725 to 0.9; 0.8 to 1.0; 0.9 to 1.1; 1.0 to 1.3; 1.2 to 1.5; 1.4 to 2.0; 1.9 to 2.5; 2.4 to 3.0; 2.9 to 3.5; 3.4 to 3.8; 3.7 to 3.9; 3.85; to 5.0; 5.0 to 6.0; 6.0 to 7.0; 7.0 to 8.0; 8.0 to 9.0; 9.0 to 10.0; 10.0 to 11.0; 11.0 to 12.0; 12.0 to 13.0; 13.0 to 14.0; 14.0 to 15.0; 15.0 to 16.0; 16.0 to 17.0; 17.0 to 18.0; 18.0 to 19.0; 19.0 to 20.0; 20.0 to 21.0; 21.0 to 22.0; 22.0 to 23.0; 23.0 to 24.0; 24.0 to 25.0; less than 0.5; less than 0.7; less than 0.75; less than 0.9; less than 1.0; less than 1.1; less than 1.5; less than 1.75; less than 2.0; less than 2.25; less than 5.0; less than 6.0; less than 7.0; less than 10.0; or less than 15.0 weight percent of disintegrant.
[0088] Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof. For example, croscarmellose sodium can be used when formulating fixed dose combinations of daclatasvir.
Croscarmellose sodium can be used from or from about 0.1 to 25 weight percent, for example as any of the weight percentages described above.
[0089] Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof. Other lubricants that can be used include, but are not limited to, for example, a solid silica gel (AE OSIL 200®, manufactured by W.R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SIL.TM. (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about one weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated. However, in some cases up to 15 weight percent can be used. For example, lubricants in the amount of less than 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5, 0.25, or 0.1 weight percent can be used. Lubricants in the amount of from or from about 0.01 to 0.1; 0.1 to 0.2; 0.2 to 0.3; 0.3 to 0.4; 0.4 to 0.5; 0.5 to 0.6; 0.6 to 0.7; 0.7 to 0.8; 0.8 to 0.9; 0.9 to 1; 1 to 2; 2 to 3; 3 to 4; 4 to 5; 5 to 6; 6 to 7; 7 to 8; 8 to 9; 9 to 10; 10 to 11; 11 to 12; 12 to 13; 13 to 14; or 14 to 15 weight percent can be used.
[0090] One aspect of the present invention is concerned with a process for making rapidly dissolving and dispersing dosage forms, e.g., orally consumable formulations, such as films, for the delivery of pharmaceutically active drugs and with the dosage forms so obtained.
[0091] For example, the inventors were surprised to have found that certain antiviral drugs, such as daclatasvir, can be formulated as a tablet using water soluble fillers in the formulation process. Any such water soluble fillers can be used including those described above. In certain embodiments where a formulation is sought comprising daclatasvir alone or in combination with ribavirin, ribavirin can be used as the filler. In some instances, ribavirin possesses desired filler properties. Without intending to be bound to any particular theory, it is thought that use of an appropriate filler in this way avoids the stalling of disintegration and dissolution events of daclatasvir in the intestinal tract of a patient which occurs when daclatasvir is formulated using standard methods. If daclatasvir is formulated without the appropriate filler, it gels up, and does not disintegrate and dissolve, significantly decreasing or eliminating absorption rates.
[0092] The present invention also presents novel ways of formulating simeprevir (along or in combination with dalclatsavir or any other drug disclosed throughout). For example, simeprevir can be combined with any suitable solid carrier, provided the resulting combination has physical properties that allow it to be more easily formulated than the parent compound. Non-limiting suitable solid carriers can comprise kaolin, bentonite, hectorite, colloidal magnesium-aluminum silicate, silicon dioxide, magnesium trisilicate, aluminum hydroxide, magnesium hydroxide, magnesium oxide and talc. In one embodiment of the invention, the solid carrier can comprise calcium silicate (such as Zeopharm), or magnesium aluminometasilicate (aka magnesium aluminum silicate; such as Neusilin). Calcium silicate can be a material containing not less than 4% calcium oxide and not less than 35% silicon dioxide. Thus, a person of skill in the art can substitute calcium silicate with a material that contains calcium oxide and silicon dioxide in the correct amounts or add them to the composition individually. Magnesium aluminometasilicate can be a material that contains magnesium, aluminum, silicon, oxygen, and water. For example, magnesium
aluminometasilicate can contain or can contain about silicon dioxide 61.1%, magnesium oxide 13.7%, aluminum oxide 9.3%, titanium dioxide 0.1%, ferric oxide 0.9%, calcium oxide 2.7%, sodium oxide 2.9%, potassium oxide 0,3%, carbon dioxide 1.8%, and/or water of combination 7.2%.
[0093] Suitable silica derivatives for use in the compositions of the invention and methods for preparing such silica derivatives include those that are described in international patent application publication number WO 03/037379 and the references cited therein. These silica derivatives can comprise a granular hydrophilic fumed silica that has a mean particle diameter of or of about 10 to 120 micron (e.g., 5 to 25; 20 to 50; 40 to 80; 75 to 100; 85 to 110; 100 to 120; or 110 to 125 micron) and a BET surface area of or of about 40 to 400 m2/g (e.g., 30 to 70; 60 to 100; 90 to 150; 130 to 180; 170 to 200; 190 to 240; 230 to 290; 280 to 320; 300 to 350; 340 to 400; 380 to 450; or 410 to 500 m2/g) (determined according to DIN 66 131 with nitrogen).
[0094] The silica derivatives can also have a pore volume of or of about 0.5 to 2.5 mL/g (e.g., 0.4 to 0.8; 0.7 to 0.9; 0.8 to 1.2; 1.1. to 1.6; 1.4 to 1.9; 1.7 to 2.2; 2.0 to 2.5; 2.3 to 3.0 mL/g), where less than or less than about 5% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10%) of the overall pore volume has a pore diameter of less than or less than about 5 nm (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nm), the remainder being mesopores and macropores. Additionally, the silica derivatives typically have a pH in the range of or about 3.6 to 8.5 (e.g., 3.5 to 4.0; 4.0 to 5.0; 5.0 to 6.0; 6.0 to 7.0; 7.0 to 8.0; or 8.0 to 9.0) and a tamped density of or of about 220 to 700 g/L ( e.g., 200 to 300; 300 to 400; 400 to 500; 500 to 600; 600 to 700; or 700 to 770 g/L).
[0095] A specific silica material that can be useful in the compositions and methods of the invention is AEROPERL® 300 (fumed silica), which is available from Evonik Degussa AG, Dusseldorf, Germany. Other collodial silicon dioxide can be used, e.g., Aerosil, Cab-O-Sil, and or Wacker HDK. Other materials having physical and chemical properties similar to the silica materials described herein can also be used.
[0096] The silica particles can have a mean grain diameter of or of about 20-40 micron (e.g., 10 to 20; 15 to 25; 20 to 30; 25 to 35; 30 to 40; 35 to 45; 40 to 50 micron), in one
embodiment of the invention the silica particles have a BET surface area of at least or of at least about 150 m2/g (e.g., 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, or 350 m2/g). In one embodiment of the invention the silica particles have a BET surface area of at least 200 m2/g. In one embodiment of the invention the silica particles have a BET surface area of at least 250 m2/g. In one embodiment of the invention the silica particles have a BET surface area of at least 275 m2/g.
[0097] When composition comprise simeprevir, the composition can be coated in the pores and on the surface of the fumed silica particles. It has been determined that up to or up to about 60% (w/w) of simeprevir can be loaded on these silica particles. Other amounts of simeprevir can be loaded on these silica particles such as up to or up to about 10, 20, 30, 40, 50, 70, 80, or 90% (w/w). This high loading capacity can be beneficial for pharmaceutical applications. In one embodiment of the invention the weight percentage of simeprevir to the silica particles is 20% ± 15%. In one embodiment of the invention the weight percentage of simeprevir to the silica particles is 60% ± 15%. In one embodiment of the invention the weight percentage of simeprevir to the silica particles is 56% ± 15%. In other embodiments of the invention, the weight percentage of simeprevir can be 10, 20, 30, 40, 50, 70, 80, or 90% ± 5, 10, 15, 20, 25, or 30% (or any combinations such as 10% ± 25% or 70% ± 10%.
[0098] Simeprevir can be loaded on the solid carrier using any suitable method. As used herein, the term "loaded" on a solid carrier and its grammatical equivalents includes, but is not limited to drugs, e.g., simeprevir, coated with the pores and on the surface of a solid carrier. For example, simeprevir can be loaded on the solid carrier by: a) spraying a solution of the compound (e.g. a solution of the compound in dichloromethane solvent) onto the solid carrier. Any solvent in which simeprevir is soluble can be used. For example, the solvent can comprise a volatile organic solvent. At a later time, b) The volatile solvent can then be evaporated resulting in a dry flowable powder containing simeprevir loaded on the carrier particles.
[0099] One or more pharmaceutically acceptable excipients can be combined with the mixture to provide a second mixture. These pharmaceutically acceptable excipients can include fillers, binders, and disintegrants, including those that were disclosed above in the excipient section. In order to improve the processability of the mixture in the subsequent aqueous granulation process, it can be beneficial to select fillers and disintegrants that are compatible with this aqueous process. For example microcrystalline cellulose (filler) and croscarmellose sodium (disintegrant) were found to be particularly compatible with the subsequent aqueous granulation process. In one embodiment of the invention the weight percentage of microcrystalline cellulose to the total weight of the second mixture is or is about 50% ± 25% (e.g., 10, 20, 30, 40, 60, 70, or 80% ± 5, 10, 15, 20, 25, 30, 35, or 40%). In one embodiment of the invention the weight percentage of croscarmellose sodium is or is about 2% ± 2% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% ± 0.5, 1, 1.5, 2.5, 3, 3.5, 4, 4.5, or 5%). Following addition of the pharmaceutically acceptable excipients, the second mixture can be mixed, for example, using a mechanical mixer. The mixture can be further processed for tableting or capsule filling. This further processing can entail unit operations such as blending, or roller compaction, wet granulation, and/or milling. It can also entail co- formulation with other active drugs.
[00100] A suitable pharmaceutically acceptable lubricant/glidant (e.g. , magnesium stearate, stearic acid, calcium stearate, zinc stearate, or pregelatinized starch or any disclosed in the excipients section) can be combined with the third mixture to provide a fourth mixture. In one embodiment, the weight percentage of magnesium stearate to the total weight of the fourth mixture can be 1% ± 0.5% (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% ± 0.5, 1, 1.5, 2.5, 3, 3.5, 4, 4.5, or 5%).
[00101] Other forms suitable for oral administration include liquid form preparations including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions, or solid form preparations which are intended to be converted shortly before use to liquid form
preparations. Emulsions can be prepared in solutions, for example, in aqueous propylene glycol solutions or can contain emulsifying drugs, for example, such as lecithin, sorbitan monooleate, or acacia. Aqueous solutions can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening drugs. Aqueous suspensions can be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending drugs. Suitable fillers or carriers with which the compositions can be administered include agar, alcohol, fats, lactose, starch, cellulose derivatives, polysaccharides, polyvinylpyrrolidone, silica, sterile saline and the like, or mixtures thereof used in suitable amounts. Solid form preparations include solutions, suspensions, and emulsions, and can contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing drugs, and the like.
[00102] A syrup or suspension can be made by adding the active compound to a concentrated, aqueous solution of a sugar, e.g., sucrose, to which can also be added any accessory ingredients. Such accessory ingredients can include flavoring, an drug to retard crystallization of the sugar or an drug to increase the solubility of any other ingredient, e.g., as a polyhydric alcohol, for example, glycerol or sorbitol. [00103] Pharmaceutically acceptable derivative. It will also be appreciated that the drugs that are administered in a fixed dose combination in a single pill can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative can include, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, free- base, acid, mixtures thereof, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
[00104] As used herein, the term "pharmaceutically acceptable salt" and its grammatical equivalents can refer to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a pharmaceutically acceptable salt reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" can mean any non-toxic salt or salt of an ester of a compound of this invention. The term "inhibitorily active metabolite or residue thereof and its grammatical equivalent can refer to a metabolite or residue of the parent drug is also an inhibitor.
[00105] If a drug or composition is a salt, it can refer to a pharmaceutically acceptable salt, including but not limited to the salts found in the handbook of
Pharmaceutical Salts: Properties, Selection, and Use, " R. Heinrich Stahl and Camile G. Wermuth, eds., Wiley-VCH, 2nd Edition (2011) or S. M. Berge et al, J. Pharmaceutical Sciences, 1977, 66, 1-19. For example, the drugs can be formulated into, but not limited to, hydrochloride salts, hydrobromide salts, hydroiodide salts, fumaric acid salts, maleic acid salts, amino acid salts, mineral acid salts, addition salts, nitrate salts, phosphate salts, succinate salts, maleate salts, fumarate salts, citrate salts, tartrate salts, gluconate salts, lactate salts, lactobionate salts, lauryl sulfate salts, glutamate salts, acetamidobenzoate salts, potassium salts, sodium salts, calcium salts, tromethamine salts, 2-aminoethanol salts, lysine salts, and/or arginine salts. Pharmaceutically acceptable salts of the compounds of this invention suitable for inclusion in a fixed dose combination in a single pill can include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts can include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N4" (Ci-4alkyl)4 salts. This invention also envisions the quatemization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products can be obtained by such quatemization. Representative alkali or alkaline earth metal salts can include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts can include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
[00106] Pharmaceutically acceptable carrier, adjuvant, or vehicle. As described above, the present invention composition, e.g., fixed-dose combinations, can additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, can include any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active drugs, isotonic drugs, thickening or emulsifying drugs, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers that can be used in formulating
pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as pharmaceutically acceptable carriers can include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, mesoporous silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering drugs such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring drugs, releasing drugs, coating drugs, sweetening, flavoring, and preservatives and antioxidants.
[00107] Weight, Volume, and Percent Weight/Volume. The weight of the composition can vary and total weight can have an effect on the effectiveness of the composition, e.g., by affecting dissolution rates. In some cases the weight of the composition (in whatever form, e.g., tablet, capsule, powder, pill, etc.) can be less than or less than about 10 grams (g). In some cases, the composition can be less than or less than about 9, 8, 7, 6, 5, 4, 3, 2, or 1 grams. The composition can be less than or less than about 3 grams. In some cases, the composition can be less than or less than about 1.6 grams. In some cases, the composition can be less than or less than about 1.2 grams. In other cases, the composition can be less than or less than about 0.9 grams. The composition can also be from or from about 0.1 to 0.5; 0.4 to 0.9; 0.8 to 1.3; 1.2 to 1.8; 1.7 to 2.3; 2.2 to 2.8; 2.7 to 3.3; or 3.2 to 4.0 grams.
[00108] The volume of the composition can vary and total volume can have an effect on the effectiveness of the composition, e.g., by affecting dissolution rates. In some cases the volume of the composition (in whatever form, e.g., tablet, capsule, powder, pill, etc.) can be less than or less than about 100ml. In some cases, the composition can be less than or less than about 90, 80, 70, 60, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 mis. The composition can be less than or less than about 3 mis. In some cases, the composition can be less than or less than about 1.6 mis. In some cases, the composition can be less than or less than about 1.2 mis. In other cases, the composition can be less than or less than about 0.9 mis. The composition can also be from or from about 0.1 to 0.5; 0.4 to 0.9; 0.8 to 1.3; 1.2 to 1.8; 1.7 to 2.3; 2.2 to 2.8; 2.7 to 3.3; or 3.2 to 4.0 mis.
[00109] In some cases, the weight/volume percent (mass/volume) can be important.
Weight volume percent can be calculated by:
Weight Volume percent = (weight of solute (in g)/volume of solution (in mL)) X 100 For example, in some of the compositions disclosed herein, the weight/volume can be less than or less than about 50%. For example, the compositions disclosed herein can contain a weight/volume percent of less than or less than about 45, 40, 35, 30, 25, 20, 15, 10, 5, or 1%. The compositions can also have a weight/volume percent of from or from about 0.1 to 0.2; 0.2 to 0.3; 0.3 to 0.4; 0.4 to 0.5; 0.5 to 0.6; 0.6 to 0.7; 0.7 to 0.8; 0.8 to 0.9; 0.9 to 1.0; 1.0 to 1.3; 1.2 to 1.4; 1.3 to 1.5; 1.4 to 2.1; 2 to 8; 5 to 10; 6 to 14; 13 to 16; 15 to 20; 19 to 30; 22 to 38; 35 to 45; or 40 to 50%.
[00110] Routes of Administration and Effective Doses. Yet another aspect of the present disclosure relates to routes of administration and effective doses for pharmaceutical compositions comprising an drug or combination of drugs of the instant disclosure. Such pharmaceutical compositions can be used to treat disease such as viral disease, including hepatitis, e.g., HCV.
[00111] Compounds of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, transdermal patch, pulmonary, vaginal, suppository, or parenteral (including intramuscular, intraarterial, intrathecal, intradermal, intraperitoneal, subcutaneous and intravenous) administration or in a form suitable for administration by aerosolization, inhalation or insufflation. General information on drug delivery systems can be found in Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott Williams & Wilkins, Baltimore Md. (1999).
[00112] Compounds can also be encapsulated within liposomes using well-known technology. Biodegradable microspheres can also be employed as carriers for the
pharmaceutical compositions of this disclosure. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268, 5,075,109, 5,928,647, 5,811,128, 5,820,883, 5,853,763, 5,814,344 and 5,942,252.
[00113] The compound can be administered in liposomes or microspheres (or microparticles). Methods for preparing liposomes and microspheres for administration to a subject are well known to those of skill in the art. U.S. Pat. No. 4,789,734, the contents of which are hereby incorporated by reference, describes methods for encapsulating biological materials in liposomes. Essentially, the material is dissolved in an aqueous solution, the appropriate phospholipids and lipids added, and along with surfactants if required, and the material dialyzed or sonicated, as necessary. A review of known methods is provided by G. Gregoriadis, Chapter 14, "Liposomes," Drug Carriers in Biology and Medicine, pp. 2.sup.87- 341 (Academic Press, 1979).
[00114] Microspheres formed of polymers or polypeptides are well known to those skilled in the art, and can be tailored for passage through the gastrointestinal tract directly into the blood stream. Alternatively, the compound can be incorporated and the microspheres, or composite of microspheres, implanted for slow release over a period of time ranging from days to months. See, for example, U.S. Pat. Nos. 4,906,474, 4,925,673 and 3,625,214, and Jein, TIPS 19:155-157 (1998).
[00115] The concentration of drug can be adjusted, the pH of the solution buffered and the isotonicity adjusted to be compatible with intravenous injection, as is well known in the art.
[00116] The compounds of the disclosure can be formulated as a sterile solution or suspension, in suitable vehicles, well known in the art. The pharmaceutical compositions can be sterilized by conventional, well-known sterilization techniques, or can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. Suitable formulations and additional carriers are described in Remington "The Science and Practice of Pharmacy" (20th Ed., Lippincott Williams & Wilkins, Baltimore MD).
[00117] The drugs or their pharmaceutically acceptable salts can be provided alone or in combination with one or more other drugs or with one or more other forms. For example, a formulation can comprise one or more drugs in particular proportions, depending on the relative potencies of each drug and the intended indication. For example, in compositions for targeting two different host targets, and where potencies are similar, about a 1:1 ratio of drugs can be used. The two forms can be formulated together, in the same dosage unit, e.g., in one cream, suppository, tablet, capsule, aerosol spray, or packet of powder to be dissolved in a beverage; or each form can be formulated in a separate unit, e.g., two creams, two suppositories, two tablets, two capsules, a tablet and a liquid for dissolving the tablet, two aerosol sprays, or a packet of powder and a liquid for dissolving the powder, etc.
[00118] In some embodiments, a drug can be administered in combination with one or more other compounds, forms, and/or drugs, e.g., as described above. Pharmaceutical compositions comprising combinations a first viral drugs, e.g., HCV drugs, with one or more other second viral drug can be formulated to comprise certain molar ratios. For example, molar ratios of about 99: 1 to about 1 : 99 of a first (viral) drug to the second (viral) drug can be used. In some subset of the embodiments, the range of molar ratios of a first (viral) drug to the second (viral) drug can be from about 80:20 to about 20:80; about 75:25 to about 25:75, about 70:30 to about 30:70, about 66:33 to about 33:66, about 60:40 to about 40:60; about 50:50; and about 90: 10 to about 10:90. The molar ratio of a first (viral) drug to the second (viral) drug can be used can be about 1 :9, and in some embodiments can be about 1:1. The two drugs, forms and/or compounds can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, or packet of powder to be dissolved in a beverage; or each drug, form, and/or compound can be formulated in separate units, e.g., two creams, suppositories, tablets, two capsules, a tablet and a liquid for dissolving the tablet, an aerosol spray a packet of powder and a liquid for dissolving the powder, etc.
[00119] If necessary or desirable, the drugs and/or combinations of drugs can be administered with still other drugs. The choice of drugs that can be co-administered with the drugs and/or combinations of drugs of the instant disclosure can depend, at least in part, on the condition being treated. Drugs of particular use in the formulations of the present disclosure can include, for example, any drug having a therapeutic effect for a viral infection, including, e.g., drugs used to treat inflammatory conditions. For example, in treatments for HCV, in some embodiments formulations of the instant disclosure can additionally contain one or more conventional anti-inflammatory drugs, such as an NSAID, e.g., ibuprofen, naproxen, acetaminophen, ketoprofen, or aspirin. In some alternative embodiments for the treatment of hepatitis, formulations of the instant disclosure can additionally contain one or more conventional hepatitis antiviral drugs as disclosed throughout. As another example, formulations can additionally contain one or more supplements, such as vitamin C, E or other anti-oxidants.
[00120] When formulating compounds of the disclosure for oral administration, it can be desirable to utilize gastroretentive formulations to enhance absorption from the gastrointestinal (GI) tract. A formulation which is retained in the stomach for several hours can release compounds of the disclosure slowly and provide a sustained release that can be preferred in some embodiments of the disclosure. Disclosure of such gastro-retentive formulations are found in Klausner, E.A.; Lavy, E.; Barta, M.; Cserepes, E.; Friedman, M.; Hoffman, A. 2003 "Novel gastroretentive dosage forms: evaluation of gastroretentivity and its effect on levodopa in humans." Pharm. Res. 20, 1466-73, Hoffman, A.; Stepensky, D.; Lavy, E.; Eyal, S. Klausner, E.; Friedman, M. 2004 "Pharmacokinetic and pharmacodynamic aspects of gastroretentive dosage forms" Int. J. Pharm. 11, 141-53, Streubel, A.; Siepmann, J.; Bodmeier, R.; 2006 "Gastroretentive drug delivery systems" Expert Opin. Drug Deliver. 3, 217-3, and Chavanpatil, M.D.; Jain, P.; Chaudhari, S.; Shear, R.; Vavia, P.R. "Novel sustained release, swellable and bioadhesive gastroretentive drug delivery system for olfoxacin" Int. J. Pharm. 2006. Expandable, floating and bioadhesive techniques can be utilized to maximize absorption of the compounds of the disclosure. If the compositions contain multiple sections or layers, each layer or section can independently can be designed to release the drug at different times.
[00121] The compounds of the disclosure can be formulated for parenteral
administration (e.g., by injection, for example, bolus injection or continuous infusion) and can be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example, solutions in aqueous polyethylene glycol.
[00122] For injectable formulations, the vehicle can be chosen from those known in art to be suitable, including aqueous solutions or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles. The formulation can also comprise polymer compositions which are biocompatible, biodegradable, such as poly(lactic-co- glycolic)acid. These materials can be made into micro or nanospheres, loaded with drug and further coated or derivatized to provide superior sustained release performance. Vehicles suitable for periocular or intraocular injection include, for example, suspensions of therapeutic drug in injection grade water, liposomes and vehicles suitable for lipophilic substances. Other vehicles for periocular or intraocular injection are well known in the art.
[00123] In some embodiments, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition can also include a solubilizing drug and a local anesthetic such as lidocaine to ease pain at the site of the injection.
Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active drug. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
[00124] When administration is by injection, the active compound can be formulated in aqueous solutions, specifically in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer. The solution can contain formulatory drugs such as suspending, stabilizing and/or dispersing drugs. Alternatively, the active compound can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In some embodiments, the pharmaceutical composition does not comprise an adjuvant or any other substance added to enhance the immune response stimulated by the peptide. In some embodiments, the pharmaceutical composition comprises a substance that inhibits an immune response to the peptide. Methods of formulation are known in the art, for example, as disclosed in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Co., Easton P.
[00125] In addition to the formulations described previously, the drugs can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation or transcutaneous delivery (for example, subcutaneously or intramuscularly), intramuscular injection or use of a transdermal patch. Thus, for example, the drugs can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[00126] In some embodiments, pharmaceutical compositions comprising one or more drugs of the present disclosure exert local and regional effects when administered topically or injected at or near particular sites of infection. Direct topical application, e.g., of a viscous liquid, solution, suspension, dimethylsulfoxide (DMSO)-based solutions, liposomal formulations, gel, jelly, cream, lotion, ointment, suppository, foam, or aerosol spray, can be used for local administration, to produce for example, local and/or regional effects.
Pharmaceutically appropriate vehicles for such formulation include, for example, lower aliphatic alcohols, polyglycols (e.g., glycerol or polyethylene glycol), esters of fatty acids, oils, fats, silicones, and the like. Such preparations can also include preservatives (e.g., p- hydroxybenzoic acid esters) and/or antioxidants (e.g., ascorbic acid and tocopherol). See also Dermatological Formulations: Percutaneous absorption, Barry (Ed.), Marcel Dekker Incl, 1983.
[00127] The solubility of the components of the present compositions can be enhanced by a surfactant or other appropriate co-solvent in the composition. Such cosolvents include polysorbate 20, 60, and 80, Pluronic F68, F-84 and P-103, cyclodextrin, or other drugs known to those skilled in the art. Such co-solvents can be employed at a level of from about 0.01% to 2% by weight.
[00128] In some embodiments, the pharmaceutical dosage forms can be formulated to provide a controlled release of one or more drugs. The term "controlled release" and its grammatical equivalents can refer to the release of the compound from a dosage form in which it is incorporated according to a desired profile over a period of time, e.g., an extended period of time. Controlled release profiles include, for example, extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled. In contrast to immediate release compositions, controlled release compositions can allow delivery of an agent to a subject over an extended period of time. Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response while minimizing side effects as compared to conventional rapid release dosage forms. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding short acting, immediate release preparations.
[00129] The compositions herein can be formulated into an immediate release composition. However, the compositions as disclosed herein can also be formulated into extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled formulations. Further, if a composition is formulated into one or more sections, then each section can be independently formulated into an extended release, delayed release, sustained release, prolonged release, pulsatile release, programmed release, time release, and/or rate controlled formulations. This can be desirable, if a first drug is required as precursor to increase the effectiveness of the second drug. Or in another case, if the same drug is formulated to be released at different areas of the gastrointestinal tract.
[00130] A sustained release composition, e.g., in beads, can release drugs from or from about 20 to 80%, 20 to 70%, 25 to 60%, and 30 to 50%, of the drug within 2 hours. For example, a sustained release composition can release at least or at least about 50, 60, 70, 80, 90, 95, or 99% by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, before or before about 24 hours based on the weight of the drug of the composition. Further, the release profile of the sustained release compositions can often include achieving 90% after or after about 4 hours (i.e., beyond the 4 hour point of the dissolution test), 6 hours, or 8 hours. Some preferred sustained release compositions are so prolonged that they do not release 90% of the drug before or before about 10 hours, 12 hours, 20 hours, or 24 hours. These release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1N HC1 followed by pH 6.8 phosphate buffer, 900 ml, and 50 rptn.
[00131] An extended released composition can release drugs not more than or more than about 30% after 90 minutes, from or from about 45% to 60% after 4 hours, from or from about 65% to 80% after 8 hours; from or from about 80 to 90% after 14 hours; and not less than or not less than about 90% after 24 hours. For example, an extended release composition can releases not more than or more than about 5, 10, 20, 25, or 30% after 90 minutes, from or from about 35, 40, or 45% to 50, 55, 60, or 65% after 4 hours, from or from about 50, 55, 60, or 65% to 70, 75, 80 or 85% after 8 hours; from or from about 70, 75, 80, or 85 to 90, 95, or 99% after 14 hours; and not less than or not less than about 90, 95, 97, or 99% after 24 hours by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition. These release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1 N HC1 followed by pH 6.8 phosphate buffer, 900 ml, and 50 rpm.
[00132] A delayed released composition, e.g., in beads, can release drugs from or from about 0 to 25% of the drug within 90 minutes (e.g., when inside the gastrointestinal tract). For example, a delayed release composition can release from or from about 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 15, 20, 25, or 30% after 90 minutes by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug of the composition. Further, the release profile of the delayed release compositions can often include and immediate release or sustained release after the initial hour period. For example, a composition can be coated with an enteric coating which is designed to release drug as it enters the small intestine, but not within the stomach. Any enteric coating can be used. For example, materials used for enteric coatings can include fatty acids, waxes, shellac, plastics, and plant fibers. This can include methyl acrylate-methacrylic acid copolymers, cellulose acetate succinate, dydroxy propyl methyl cellulose phthalate, hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate), polyvinyl acetate phthalate (PVAP), methyl methacrylate- methacrylic acid copolymers, shellac, cellulose acetate trimellitate, sodium alginate, zein or anyone combination thereof.
[00133] Once in the small intestine, the coating is typically breached and the drug can be immediately released. This can be good for drugs that are pH sensitive. Further, the delay in drug release can be even later, e.g., within the large intestine. This delayed release can be good for those drugs that have an effect within the large intestine, in which absorption prior to the large intestine is undesirable. These release profiles can be determined by any method, for example, in Type II USP apparatus, 0.1N HC1 followed by pH 6.8 phosphate buffer, 900 ml, and 50 rpm.
[00134] Pharmacokinetics. In standard pharmacokinetic studies, a sufficient number of data collection points should be included, and pharmacokinetic parameters ,such as area under the blood concentration-time curve (AUC), clearance, maximum blood concentration (Cmax), minimum blood concentration (Cmin), time to reach maximum blood concentration (Tmax), volume of distribution at steady state (Vdss), mean residence time (MRT), and half-life (t1/2), should be determined by using model independent analytical methods. In addition to the above parameters, the rate constant and information about the volume of distribution (V1, Vd%, and Vdss) can be obtained using pharmacokinetic models like a compartment model. Models that can describe drug concentrations in a blood-versus-time profile are useful to estimate changes in blood concentrations caused by differences in dosage and dose regimen, and may be used to tailor individual dosing plans. The analysis should also be extended to include a PK/PD analysis.
[00135] Cmax. The pharmaceutical compositions described throughout can possess certain unique characteristics including Cmax. The pharmaceutical compositions as described throughout, can exhibit a Cmax from or from about 1.0, 1.5, 2.0, 2.5, or 3.0 pg/ml to 5.0, 10.0, 15.0, 20.0, or 25.0 ng/ml {e.g., 1.0 pg/ml to 25.0 ng/ml) with plasma of patients with an average distribution volume of 50 L, after oral administration. For example, the Cmax value can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or lOOpg/ml. Cmax can also be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 ng/ml.
[00136] Cmax can be calculated by any standard method, e.g., the USP. Cmax is the peak plasma concentration of a drug after administration. The mathematical formula to calculate Cmax (for 1 -compartment models) can be Cmax = (F * Xo * exp (-k * tmax)) / V. [00137] Tmax. The pharmaceutical compositions described throughout can possess certain unique characteristics including Tmax. The pharmaceutical compositions as described throughout, can exhibit a Tmax from or from about 30, 60, 90, 120, 150, 180, 210, or 240 minutes with plasma of patients with an average distribution volume of 50 L after oral administration. Tmax can also be from or from about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, in some cases, e.g., in certain controlled release formulations.
Tmax can be calculated by any standard method. Tmax is the time it takes to reach Cmax. The mathematical formula to calculate Tmax (for 1 -compartment models) can be Tmax = (ln(ka) - ln(k)) / (ka - k).
METHODS OF TREATMENT
[00138] The compositions described herein can be used to treat disease, for example, viral disease. The viral disease can be a virus that infects the liver such as the hepatitis virus. Some examples of the hepatitis virus can include the virus that causes hepatitis A (HAV); hepatitis B (HBV); hepatitis C (HCV); hepatitis D (HDV); hepatitis E (HEV); hepatitis F (HFV); and/or hepatitis G (HGV).
[00139] Some embodiments of the present disclosure relates to methods of using pharmaceutical compositions to treat disease, such as viral disease, including hepatitis, e.g., HCV. Another embodiment of the present disclosure provides methods, pharmaceutical compositions, and kits for the treatment of animal subjects. The term "animal subject" and its grammatical equivalents as used herein includes humans as well as other mammals.
[00140] For embodiments where a prophylactic benefit is desired, a pharmaceutical composition of the disclosure can be administered to a subject at risk of contracting a disease, such as viral disease, including hepatitis, e.g., HCV, or to a subject reporting one or more of the physiological symptoms of a disease, such as viral disease, including hepatitis, e.g., HCV, even though in some cases, the screening of the condition cannot have been made.
Administration can prevent a disease, such as viral disease, including hepatitis, e.g., HCV from developing, or it can reduce, lessen, shorten and/or otherwise ameliorate the progression of a disease, such as viral disease, including hepatitis, e.g, HCV, or symptoms that develop. The pharmaceutical composition can modulate or target a disease, such as viral disease, including hepatitis, e.g., HCV, associated biomarker. Wherein, the term modulate includes inhibition of a developmental disorder associated biomarkers or alternatively activation of a developmental disorder associated biomarkers. [00141] In some cases, the drugs or the combination of drugs have the ability to modify enzyme activity (including proteases). The ability to reduce enzyme activity can be a measure of the potency or the activity of an drug, or combination of drugs, towards or against the enzyme or other biologically important molecular process. Potency can be measured by cell free, whole cell and/or in vivo assays in terms of IC50, Ki and/or ED50 values. An IC50 value represents the concentration of an drug required to inhibit enzyme activity by half (50%) under a given set of conditions. A Ki value represents the equilibrium affinity constant for the binding of an inhibiting drug to the enzyme or other relevant biomolecule. An ED50 value represents the dose of an drug required to affect a half-maximal response in a biological assay. Further details of these measures will be appreciated by those of ordinary skill in the art, and can be found in standard texts on biochemistry, enzymology, and the like.
[00142] The present disclosure also includes kits that can be used to disease, such as viral disease, including hepatitis, e.g., HCV. These kits comprise an drug or combination of drugs that inhibits the causation agent of a disease, such as viral disease, including hepatitis, e.g., HCV and in some embodiments instructions teaching the use of the kit according to the various methods and approaches described herein. Such kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the drug. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like.
METHODS OF MAKING
[00143] A variety of different methods can be used to make the different formulations of the compositions as described herein.
[00144] Dry granulation can be used to granulate one or more drugs (a first drug) (as disclosed throughout) with intragranular excipients. For example, any of the excipients listed throughout can be used. Exemplary excipients that can be used are fillers (e.g.,
microcrystalline cellulose, and/or lactose), disintegrants (e.g., croscarmellose sodium), and lubricants (e.g., magnesium stearate).
[00145] For combination compositions, the second drug can be blended for 3 minutes in a tumbling container, e.g., at approximately 25 rpm. The blend can then be rolled and compacted on a formulation prototyping machine with counter rotating stainless steel rollers. In some cases, the stainless steel rollers can have or have about 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness, leading the formation of a compact. These compacts can then be milled to produce granules through a screen, e.g., a stainless steel rotary screen mill of 16 mesh size (1.2 mm). Other mesh sizes can be used, for example, 8, 9, 10, 11, 12, 13, 14, 15, 17, 18, 19, or 20. The granules are milled until they are free flowing.
[00146] The free base of the drugs can also be dissolved and crystallized. These solids can then be dried and the resulting powdered blended with excipients for formulation, such as tableting.
[00147] Direct blending of two or more drugs that were previously processed with excipients can be used.
[00148] If the compositions are tableted, they can be tableted into a bilayer drug. For example, the drugs can be configured into a bilayer using a tablet tooling (e.g., a round ¼ inch tableting tool) at a compression force of or of about 2,000 lbs (e.g., 1000, 1500, 2500, or 3000 lbs). The compression force can also be or be about 5,000 lbs (e.g., 4000, 4500, 5500, or 6000 lbs).
[00149] Tablets can have a certain thickness of for example, of or of about 0.5 to 5 mm. For example, the tablet can have a thickness of or of about 0.5, 1.0, 2.0, 3.0, 4.0, or 5.0 mm. The table can also have a thickness of or of about 2.58 mm. The table can also have a thickness of or of about 3.13 mm.
[00150] The tablet can also exhibit different hardness. For example, the hardness can be hardness of or of about less than 20 kp (e.g., less than 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 kp). The hardness can also be or be about 11.8 kp. The hardness can also be or be about 8.9 kp. The hardness can also be or be about 2.9 kp.
[00151] Tablets can be tested for disintegration time in deionized water at 20 °C, to determine acceptable disintegration characteristics breaking up into dispersed granules and powder within a certain amount of time. The disintegration from can vary. However, some disintegration times can be or can be about 3 minutes, 2 minutes 30 seconds, 1 minute 40 seconds, or 40 seconds.
[00152] In some cases, two or more of the drugs can be directly bending with excipients at the same time. If there is a third drug, then this can be either directly blending with the first two drugs, or later added after it has been processed with its own excipients (e.g., in bilayer tableting).
[00153] In some cases, one or more drugs can be formulated into an amorphous solid form. These amorphous solids can be produced via spray drying after dissolving in a solvent. The amorphous form can improve dissolution and bioavailability. To improve the formulation, amorphous drugs can be loaded onto hard readily flowable porous silica carrier particles, e.g., AeroPerl 300, by Evonik. If the silica carrier particles are used, then the drug can be dissolved with a solvent and then missed thoroughly until homogenous with the silica carrier particle. These AeroPerl loaded particles and then be blended with other drugs (that were processed with excipients) and further process into a desired formulation, such as a tablet.
[00154] Surfactants can be used as well to improve dissolution characteristics. For example, such as sodium lauryl sulfate (SLS), ionic Tween 80, non-ionic lecithin types of surfactants, or any sorbitan esters (e.g., sorbitan diisostearate, sorbitan dioleate, sorbitan monoisostearate, sorbitan monolaurate, sorbitan monooleate, sorbitan monopalmiiate, sorbitan monostearate, sorbitan sesquiisostearate, sorbitan sesquioleate, sorbitan sesquistearate, sorbitan
triisostearate, sorbitan trioleate, or sorbitan tristearate). The amount of surfactants used is typically less than or less than about 20% w/w. For example, less than or less than about 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1% surfactant can be used.
EXAMPLES
Example la: NS5B/NS5A Tableting
[00155] A fixed dose combination HCV treatment product was formulated and manufactured with two active drugs in a single tablet. This comprised a nucleotide NS5B class RNA polymerase inhibitor, and a NS5A class viral replication complex inhibitor. This product contained sofosbuvir (400 mg) as the NS5B inhibitor, and daclatasvir dihydrochloride (60 mg) as the NS5A inhibitor.
[00156] Preparation of a Representative Bilayer Tablet
[00157] The manufacturing procedure was broken down into multiple segments: dry granulation of sofosbuvir with intragranular excipients, direct blending of daclatasvir dihydrochloride with excipients, bilayer tableting. Further unit processes such as film coating, and bottling and blister packaging are also added. The stepwise procedure is detailed below.
[00158] Step 1: Dry Granulation, Milling, and Blending of Sofosbuvir
[00159] Crystalline sofosbuvir and dry granulation excipients were weighed in the following mass proportions:
• Sofosbuvir (nucleotide NS5B active substance) 40.0 % (w/w)
• Microcrystalline cellulose (filler) 29.05 % (w/w)
Lactose (filler) 30.0 % (w/w) • Croscarmellose sodium (disintegrant) 0.73 % (w/w)
• Magnesium stearate (lubricant) 0.22 % (w/w)
[00160] The sofosbuvir and dry granulation excipients were blended for 3 minutes in a tumbling container at approximately 25 rpm. The blend was then roller compacted on a formulation prototyping machine with counter rotating stainless steel rollers of 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness. The compacts formed were ribbon-like of 25 mm width, and had processing and mechanical properties desirable for larger-scale roller compaction processing. The compacts were milled to produce granules through a stainless steel rotary screen mill of 16 mesh size (1.2 mm). The granules were readily free flowing, which would be useful for reliable processing on high-speed commercial scale tablet manufacturing equipment.
[00161] Step 2: Direct Blending of Daclatasvir dihydrochloride
[00162] Daclatasvir free base was dissolved and crystallized as the dihydrochloride salt following the procedure recorded for seed crystal preparation in the patent with US publication number: US 20090041716 Al. The harvested solids were dried and the resulting powder was blended with excipients for tableting in the following mass proportions:
• Daclatasvir dihydrochloride (NS5A active substance) 20.0 % (w/w)
• Microcrystalline cellulose (filler) 67.79 % (w/w)
• Croscarmellose sodium (disintegrant) 11.70 % (w/w)
• Magnesium stearate (lubricant) 0.51 % (w/w)
[00163] Step 2: Direct Blending of Daclatasvir dihydrochloride
[00164] Step 3: Bilayer Tableting
[00165] Sofosbuvir granules from #1 and daclatasvir powder from #2 above were compressed in a bilayer tableting configuration using ¼-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 83 mg. Tablets had a measured thickness of 2.58 mm, and a tablet hardness of 8.9 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 3 minutes.
[00166] Step 4: Full Strength Bilayer Tableting
[00167] The tableting procedure of Step 3 was scaled-up in proportion to yield total individual tablet mass of 1,330 mg, 21 mm x 10 mm (length and width) for a caplet shaped tablet tooling type, and a compression pressure of 5,000 lb. The tablet thickness was 7.0 mm, and was therefore comparable in dimensions to Atripla®, which is a commercial once daily oral antiviral fixed-dose combination product. The active substance strengths of the bilayers corresponded to 400mg sofosbuvir and 60 mg daclatasvir parent, which are respectively the recommended daily doses for these active substances for purposes of HCV infection treatment.
Example lb: Sofosbuvir/Daclatasvir Tablet Use
[00168] A fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained viralogic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example lc: Sofosbuvir/Daclatasvir Tablet Use with Ribavirin
[00169] A fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. The patient concurrently is administered 500 mg ribavirin twice daily. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example Id: Sofosbuvir/Daclatasvir/Ribavirin Fixed Dose Combination Tablet
[00170] Step 1: Direct Blending of sofosbuvir and ribavirin
[00171] Sofosbuvir and ribavirin active substances were blended with excipients for tableting in the following mass proportions:
• Sofosbuvir (nucleotide NS5B active substance) 20.0 % (w/w)
• Ribavirin (nucleoside active substance) 49.60 % (w/w)
Microcrystalline cellulose (filler) 29.44 % (w/w)
• Croscarmellose sodium (disintegrant) 0.74 % (w/w)
• Magnesium stearate (lubricant) 0.22 % (w/w)
[00172] Step 2: Direct Blending of Daclatasvir dihydrochloride
[00173] Daclatasvir free base was dissolved and crystallized as the dihydrochloride salt following the procedure recorded for seed crystal preparation in the patent with US publication number: US 20090041716 Al. The harvested solids were dried and the resulting powder was blended with excipients for tableting in the following mass proportions:
• Daclatasvir dihydrochloride (NS5A active substance) 20.0 % (w/w)
• Microcrystalline cellulose (filler) 67.79 % (w/w)
• Croscarmellose sodium (disintegrant) 11.70 % (w/w) • Magnesium stearate (lubricant) 0.51 % (w/w)
[00174] Step 3: Bilayer Tableting
[00175] Sofosbuvir/ribavirin excipient blend from Step 1 and daclatasvir powder from Step 2 above were compressed in a bilayer tableting configuration using ¼-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 73 mg. Tablets had a measured thickness of 2.69 mm, and a tablet hardness of 2.9 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 40 seconds.
[00176] Step 4: Full Strength Bilayer Tableting
[00177] The tableting procedure of Step 3 is scaled-up in proportion to yield total individual tablet mass of 1,330 mg, 21 mm x 10 mm (length and width) for a caplet shaped tablet tooling type, and a compression pressure of 5,000 lb. The active substance strengths of the bilayers correspond to 200mg sofosbuvir, 500 mg ribavirin, and 30 mg daclatasvir parent, which the appropriate doses for this product to be taken twice daily for HCV infection treatment.
Example le: Sofosbuvir/Daclatasvir/Ribavirin Fixed Dose Combination Tablet Use
[00178] The fixed-dose combination single tablet from Example Id containing
sofosbuvir/daclatasvir/ribavirin (200mg / 30mg / 250 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example 2a: NS5B/NS3/4A Tableting
[00179] A fixed dose combination HCV treatment product was formulated and manufactured with two active drugs in a single tablet. This comprised a nucleotide NS5B class RNA polymerase inhibitor, and a NS3/4A class serine protease inhibitor. This product contained sofosbuvir (400 mg) as the NS5B inhibitor, and simeprevir sodium (150 mg) as the NS3/4A inhibitor.
[00180] Preparation of a Representative Bilayer Tablet of the Invention
[00181] In one embodiment of the invention the manufacturing procedure was broken down into multiple segments: dry granulation of sofosbuvir with intragranular excipients, loading of amorphous simeprevir sodium onto AeroPerl 300 porous silica carrier particles, direct blending of simeprevir- AeroPerl particles with excipients, bilayer tableting. Further unit processes such as film coating, and bottling and blister packaging can be conceived. The stepwise procedure is detailed below.
[00182] Step 1: Dry Granulation, Milling, and Blending of Sofosbuvir
[00183] Crystalline sofosbuvir and dry granulation excipients were weighed in the following mass proportions:
• Sofosbuvir (nucleotide NS5B active substance) 40.0 % (w/w)
Microcrystalline cellulose (filler) 29.05 % (w/w)
Lactose (filler) 30.0 % (w/w)
• Croscarmellose sodium (disintegrant) 0.73 % (w/w)
• Magnesium stearate (lubricant) 0.22 % (w/w)
[00184] The sofosbuvir and dry granulation excipients were blended for 3 minutes in a tumbling container at approximately 25 rpm. The blend was then roller compacted on a formulation prototyping machine with counter rotating stainless steel rollers of 50 mm diameter, at approximately 5 rpm, and approximately 1.0 mm gap thickness. The compacts formed were ribbon-like of 25 mm width, and had processing and mechanical properties desirable for larger-scale roller compaction processing. The compacts were milled to produce granules through a stainless steel rotary screen mill of 16 mesh size (1.2 mm). The granules were readily free flowing, which would be useful for reliable processing on high-speed commercial scale tablet manufacturing equipment.
[00185] Step 2: Loading Simeprevir on Silica AeroPerl Carrier Particles
[00186] The commercial product of simeprevir sodium exists as capsules (rather than tablets) and uses the amorphous solid form of simeprevir sodium produced via spray drying from dichloromethane solvent. The amorphous form improves dissolution and bioavailability. Our attempts to prepare tablets with amorphous simeprevir were hindered by a material property where the amorphous solid form of simeprevir tended to compress in a plastic manner under tablet compression forces, yielding tablets that were highly resilient against disintegration, dispersion, and dissolution in aqueous media. To overcome this material limitation which would make fixed-dose combinations tablets less simple to formulate and produce, amorphous simeprevir sodium was loaded onto hard readily flowable porous silica carrier particles. AeroPerl 300, by Evonik, is an example carrier of this type and available for pharmaceutical use. Simeprevir sodium was dissolved dichloromethane (the same solvent as used in commercial amorphous simeprevir manufacture, except there by a spray drying process). The concentration of simeprevir sodium in solution was 49.5 mg/mL. The simeprevir solution was added to unloaded silica AeroPerl 300 particles, thoroughly mixed until homogeneous, and the solvent evaporated to dryness. The resulting dry powder contained amorphous simeprevir sodium loaded onto AeroPerl silica carrier particles. The material properties included a dense highly free flowing powder containing hard, small particles that were amenable to tableting with desirable disintegration and dispersion characteristics. The particles contained 0.56 g simeprevir sodium per gram of powder. Other loading levels (both higher and lower) are conceivable.
[00187] Step 3: Direct Blending of Simeprevir AeroPerl Particles with Excipients
Simeprevir sodium AeroPerl particles were directly blended with further excipients for tableting in the following mass proportions:
• Simeprevir sodium AeroPerl (NS3/4A active substance on silica carrier) 36.96 % (w/w)
• Microcrystalline cellulose (filler) 61.05 % (w/w)
• Croscarmellose sodium (disintegrant) 1.53 % (w/w)
• Magnesium stearate (lubricant) 0.46 % (w/w)
[00188] Step 4: Bilayer Tableting
[00189] Sofosbuvir granules from Step 1 and simeprevir sodium powder from Step 3 above were compressed in a bilayer tableting configuration using ¼-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 109 mg. Tablets had a measured thickness of 3.13 mm, and a tablet hardness of 11.8 kp. Tablets were tested for disintegration time in deionized water at 20 °C and 37 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 2 minutes 30 seconds, and within 1 minute 40 seconds respectively.
[00190] Step 5: Full Strength Bilayer Tableting
[00191] The tableting procedure of Step 4 was scaled-up in the following proportions to yield total individual tablet mass of 1,740 mg, 21 mm x 10mm (length and width) for a caplet shaped tablet tooling type, and a compression pressure of 5,000 lb.
• Simeprevir sodium AeroPerl (NS3/4A active substance on silica carrier) 37.57 % (w/w)
• Microcrystalline cellulose (filler) 60.46 % (w/w)
• Croscarmellose sodium (disintegrant) 1.52 % (w/w)
• Magnesium stearate (lubricant) 0.46 % (w/w)
[00192] The tablet thickness was 8.6 mm, and was therefore comparable in dimensions to Atripla®, which is a commercial once daily oral antiviral fixed-dose combination product. The active substance strengths of the bilayers corresponded to 400mg sofosbuvir and 150 mg simeprevir parent, which are respectively the recommended daily doses for these active substances for purposes of HCV infection treatment.
Example 2b: Sofosbuvir/Simeprevir Tablet Use
[00193] A fixed-dose combination of sofosbuvir/Simeprevir (400mg / 150mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained viralogic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example 2c: Sofosbuvir/Simeprevir Tablet Use with Ribavirin
[00194] A fixed-dose combination of sofosbuvir/daclatasvir (400mg / 60mg) as a single tablet is administered orally once daily to a patient with Hepatitis C infection for 12 weeks. The patient is also administered 500 mg ribavirin twice daily. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example 2d: Sofosbuvir/Simeprevir/Ribavirin Fixed Dose combination Tablet
[00195] A fixed dose combination tablet containing sofosbuvir 200mg, simeprevir 75mg, and ribavirin 500 mg for twice daily administration in treatment of HCV infection is prepared as follows:
[00196] Step 1: Blending of Ribavirin with Sofosbuvir
[00197] To the sofosbuvir granules yielded in Example 2a Step 1, ribavirin active substance is added in a 1 : 1 mass ratio and further blended.
[00198] Step 2: Bilayer Tableting
[00199] Fixed-dose combination tablets are compressed in a bilayer configuration where the of sofosbuvir/ribavirin material from Example 2d Step 1 form 1 layer, and simeprevir blend from Example 2a Step 3 form the second layer. The mass ratios are l,000mg : 370mg respectively. This yields a single tablet product of 1,370 mg total mass and containing 3 active substances in the following doses: 200 mg sofosbuvir / simeprevir 75 mg / ribavirin 500 mg. These are the appropriate doses for twice daily administration for patients with Hepatitis C infection. Example 2e: Sofosbuvir/Sim eprevir/Ribavirin Fixed Dose Combination Tablet Use
[00200] The fixed-dose combination single tablet from Example 2d, containing
sofosbuvir/simeprevir/ribavirin (200mg / 75mg / 250 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example 3a: NS5B /NS5A /NS3/4A Tableting
[00201] A fixed dose combination HCV treatment product was developed with three active drugs in a single tablet. This comprised a non-nucleoside/tide NS5B class RNA polymerase inhibitor, a NS5A class viral replication complex inhibitor, and a NS3/4A class serine protease inhibitor. The active substances include dasabuvir (ABT-333) as the NS5B inhibitor, daclatasvir dihydrochloride (60 mg) as the NS5A inhibitor, and simeprevir sodium as the NS3/4A inhibitor.
[00202] Preparation of a Representative Bilayer Tablet of the Invention
[00203] Step 1: Blending of dasabuvir, simeprevir loaded AeroPerl, and excipients
[00204] Dasabuvir was crystallized as the sodium salt according to the procedure of patent US 20120014913. The salt was isolated and dried. The resulting powder of dasabuvir active substance was used for further processing.
[00205] Dasabuvir sodium, and simeprevir sodium loaded AeroPerl particles from Example 2a Step 2, were directly blended with further excipients for tableting in the following mass proportions:
• Dasabuvir sodium (NS5B non-nucleaside/tide active substance) 41.77 % (w/w)
• Simeprevir sodium AeroPerl (NS3/4A active substance on silica carrier) 22.78 % (w/w)
Microcrystalline cellulose (filler) 29.42 % (w/w)
• Croscarmellose sodium (disintegrant) 3.27 % (w/w)
• Sodium lauryl sulfate (surfactant) 2.53 % (w/w)
• Magnesium stearate (lubricant) 0.22 % (w/w)
[00206] Step 2: Direct Blending of Daclatasvir dihydrochloride
[00207] Daclatasvir dihydrochloride was blended with excipients in preparation for tableting according to Example la, Step 2.
[00208] Step 3: Bilayer Tableting [00209] Dasabuvir sodium / simeprevir sodium on AeroPerl blend from Step 1, and daclatasvir powder from Step 2 above, were compressed in a bilayer tableting configuration using ¼-inch round tablet tooling at a compression force of 2,000 lb, for a total tablet mass of 99 mg. Tablets had a measured thickness of 3.23 mm, and a tablet hardness of 8.0 kp. Tablets were tested for disintegration time in deionized water at 20 °C, and exhibited acceptable disintegration characteristics breaking up into dispersed granules and powder within 2 minutes.
[00210] Step 4: Bilayer Tableting Scale-up
[00211] The materials from Steps 1 and 2 are roller compacted and granulated separately, then fed to a tableting press to manufacture fixed-dose combination tablets in a bilayer configuration where the dasabuvir/simeprevir comprise one layer, and daclatasvir comprises the second layer. The mass ratios are 758mg : 152mg respectively. This yields a single tablet product of 910 mg total mass and containing 3 active substances in the following doses: dasabuvir 250 mg / simeprevir 75 mg / daclatasvir 30 mg. These are the appropriate doses for twice daily administration for patients with Hepatitis C infection. The tablets are
manufactured at 1.5kg batch size. The tablets are coated with 36mg (dry weight) of a polymeric film comprising hypromellose, titanium dioxide, and polyethylene glycol 400, in a pan coater.
Example 3b: Dasabuvir/Simeprevir/Daclatasvir Fixed Dose Combination Tablet Use
[00212] The fixed-dose combination single tablet from Example 3a, containing
dasabuvir/simeprevir/daclatasvir (250mg / 75mg / 30 mg doses) is administered orally twice daily to a patient with Hepatitis C infection for 12 weeks. After 12 weeks continuous daily dosing with the fixed dose combination tablet, the patient is medically assessed for HCV infection and found to have a sustained virologic response (that is, a functional cure). The fixed-dose combination product is preferred by the patient and physician due to its greater convenience of administration, higher level of patient compliance to daily dosing, and reduced risk of resistance development from poor compliance.
Example 4: Sofosbuvir Tablet Formulation Development
[00213] Experiments were conducted to determine the optimal formulation composition for sofosbuvir as a single drug to identify important formulation constituents and working levels to yield tablets of commercially useful hardness, size, and disintegration characteristics. The improved single drug formulations were useful for purposes of the fixed dose-combination examples that included sofosbuvir.
[00214] Procedure: 1. A base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
2. Sofosbuvir active substance was weighed and then blended in specific proportions of the base formula from #1 in this procedure.
3. Tablets of specific accurate masses were prepared individually using a Carver Model C hydraulic press using ¼-inch round tooling.
4. The tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
[00215] Ideally tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of sofosbuvir single drug formulation examples are tabulated and described below.
Example 4a:
[00216] At 10% sofosbuvir in the base formula, acceptable hardness, thickness, and disintegration characteristics were observed. For utility in fixed dose combination tablets of modest total tablet size, an increase to the sofosbuvir proportion was required.
Example 4b:
[00217] At 25% sofosbuvir in the base formula, acceptable hardness, thickness, were observed, but the disintegration characteristics were slowed substantially and indicated that other formulations would be more optimal to facilitate further increases in sofosbuvir proportion.
Example 4c:
[00218] The disintegrant level was increased significantly to 6.70%. Surprisingly, this large increase in disintegrant level did not correlate with as much of a decrease in disintegration time as would be expected.
Example 4d: [00219] Since disintegrant level was not the dominating parameter for disintegration at this high sofosbuvir level prototype, a soluble structured composition containing spray dried lactose (filler) was prepared at approximately equal proportions of lactose to that of microcrystalline lactose. This resulted in dilution of other excipients (disintegrant and lubricant). Surprisingly, despite there being the lowest level of disintegrant, the disintegration time was substantially decreased down to 1 minute 30 seconds. Given the different tablet hardness characteristics of lactose compared with microcrystalline cellulose, it was not surprising that the tablet hardness decreased to 8.2 kp. This however is still acceptable tablet hardness for a lOOmg total tablet mass for large scale production purposes. To confirm, a simple binary mixture of 1:1 mass ratio sofosbuvir with lactose was also prepared. This did not disintegrate as fast as the lactose/base formula mixture and confirms that lactose alone is not sufficient to optimally formulate sofosbuvir, but the other excipients used had important synergistic roles in the formulation.
[00220] Other soluble filler materials or soluble active substances that have hard crystalline or partially crystalline physical states would similarly be useful to that of lactose in this composition.
[00221] With this knowledge, sofosbuvir formulation compositions were determined for the reported fixed-dose combination examples.
TABLE 3
Figure imgf000052_0001
Example 5a: Daclatasvir Tablet Formulation Development
[00222] Experiments were conducted to determine the optimal formulation composition for daclatasvir dihydrochloride as a single drug to identify important formulation constituents and working levels to yield tablets of commercially useful hardness, size, and disintegration characteristics. The improved single drug formulations were useful for purposes of the fixed dose-combination examples that included daclatasvir.
[00223] Procedure:
1. A base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
2. Daclatasvir dihydrochloride active substance was weighed and then blended in
specific proportions of the base formula from #1 in this procedure.
3. Tablets of specific accurate masses were prepared individually using a Carver
Model C hydraulic press using ¼-inch round tooling.
4. The tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
[00224] Ideally tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of daclatasvir dihydrochloride single drug formulation examples are tabulated and described below.
Example 5a:
[00225] At 10% daclatasvir dihydrochloride in the base formula, the disintegration stalled after 5 minutes indicating an unusual property. Uncompressed pure daclatasvir was rapidly wet into water and would dissolve, however in the compressed state its rapid wetting and dissolution properties were not reflected in disintegration and dissolution of this tablet formulation.
Example 5b:
[00226] Also at 10% daclatasvir dihydrochloride level, the base excipient formula was further functionalized using crystalline lactose monohydrate seeking to space apart the active substance particles partially with not just microcrystalline cellulose as filler, but also lactose as a hard and less compressible filler material. This did not substantially improve the disintegration characteristics.
Example 5c:
[00227] A strategy similar to that used in Example 5b was attempted, however using an insoluble hard filler material (silicon dioxide). This did not substantially improve the disintegration characteristics and indicated that physical spacing out of daclatasvir particles using fillers was not an effective formulation intervention.
Example 5d:
[00228] Given the pH dependence of solubility of daclatasvir, a formulation prototype was prepared using the base excipient formula plus fumaric acid as an additional excipient to create an acidic microenvironment with slowly dissolving acidic excipient to prolong the low pH microenvironment as long as possible. This approach was unsuccessful showing no significant disintegration and dissolution after 30 minutes.
Example 5e:
[00229] A prototype was prepared using the base excipient formula and lactose, similarly to that used in Example 4d, and further functionalized by increasing the croscarmellose sodium disintegrant level substantially. This formulation had a surprising and very effective impact on reducing the tablet disintegration time down to 1 minute 30 seconds. It also had an acceptable tablet hardness
Example 5f:
[00230] To assess the relative importance of lactose filler versus croscarmellose sodium disintegrant and synergies between them, a prototype was prepared similarly to Example 5e, but the lactose was omitted. This formulation still maintained a high level of disintegrant and exhibited acceptably rapid tablet disintegration and hardness.
[00231] The most effective formulation parameter for optimizing the tablet performance of daclatasvir dihydrochloride was the disintegrant level croscarmellose sodium. This was further improved with addition of water soluble lactose monohydrate filler to the base formula with high disintegrant level, although the favorable impact of lactose was not as substantial as that of croscarmellose sodium disintegrant.
[00232] Other disintegrants (for example crospovidone and sodium starch glycolate) as well as other water soluble filler materials (for example mannitol, or dextrose) would similarly improve the formulation performance in place of croscarmellose sodium and lactose as used here.
[00233] Daclatasvir daily dose for HCV treatment is 60mg. therefore, it was not as important to increase the active substance proportion for daclatasvir as it was for sofosbuvir in Example 4.
[00234] With this knowledge, daclatasvir formulation compositions were determined for the reported fixed-dose combination examples.
TABLE 4
Figure imgf000055_0001
TABLE 5
Figure imgf000055_0002
Figure imgf000056_0001
Example 6a: Simeprevir Tablet Formulation Development
[00235] Simeprevir sodium is an amorphous active ingredient that has low aqueous solubility and poor wettability characteristics. The marketed product of simeprevir sodium
(OLYSIO™) is presented as a powder filled into a capsule. OLYSIO (simeprevir, 150 mg) capsules contain the following inactive ingredients: colloidal anhydrous silica, croscarmellose sodium, lactose monohydrate, magnesium stearate and sodium lauryl sulphate. The white capsule contains gelatin and titanium dioxide and is printed with ink containing iron oxide black and shellac.
[00236] The capsule presentation of simeprevir sodium limits the possibility to fix dose combine with other drugs for the treatment of hepatitis C infection. A capsule has weight limitations because of the volume constraints of the capsule shell that are filled with loosely compacted powders. This volume constraint prohibits the addition of other drugs. On the other hand, a tablet presentation allows for higher mass loading by compressing under high pressures to increase the density of the combined products. Developing a tablet formulation of simeprevir sodium is highly desirable to prepare for the next steps in fix dose combining with other drugs.
[00237] Experiments were conducted to determine the optimal formulation composition for simeprevir as a single drug to identify important formulation constituents and working levels to yield tablets of commercially useful hardness, size, and disintegration characteristics. The improved single drug formulations were useful for purposes of the fixed dose-combination examples that included simeprevir.
[00238] Procedure:
1. A base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
2. Simeprevir sodium active substance was weighed and then blended in specific
proportions of the base formula from #1 in this procedure.
3. Tablets of specific accurate masses were prepared individually using a Carver
Model C hydraulic press using ¼-inch round tooling. 4. The tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
[00239] Ideally tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of simeprevir single drug formulation examples are tabulated and described below.
Example 6a:
[00240] At 10% simeprevir sodium in the base formula, acceptable hardness, and thickness. The disintegration characteristics of the tablet were slow for this size of tablet at a low simeprevir sodium concentration.
Example 6b:
[00241] Because of the fine particle size of the amorphous simeprevir sodium and the plastic deformation characteristics under tablet preparation, a brittle crystalline excipient, lactose monohydrate, was included as a filler to impart a soluble, disruptive component to the tablet matrix. The relative composition of simeprevir sodium, croscarmellose disintegrant and magnesium stearate lubricant was comparable to Example 5a. The resulting tablet had acceptable hardness, and thickness. The disintegration characteristics of the tablet were surprisingly faster than a tablet containing microcrystalline cellulose as the filler.
Example 6c:
[00242] Sodium lauryl sulfate surfactant was added at 1.96% w/w to aid in the wettability of simeprevir sodium in the tablet matrix. Surprisingly, including sodium lauryl sulfate, a component of the commercial capsule formulation, did not correlate with a decrease in disintegration time as would be expected.
Example 6d:
[00243] Example 2a, Step 2 details the loading of simeprevir sodium onto a silicon dioxide carrier at a ratio of 0.56 g of simeprevir sodium to 1 g of simeprevir sodium on silicon dioxide. A 16.82% concentration of simeprevir sodium on silicon dioxide has the same simeprevir sodium loading as Example 5a, with the corresponding reductions in
microcrystalline cellulose, croscarmellose sodium and magnesium stearate. Surprisingly the disintegration time reduced significantly to 20 seconds by using simeprevir sodium on silicon dioxide with no changes in any of the other ingredients.
Example 6e:
[00244] Amorphous simeprevir sodium was blended at a high loading with the crystalline active ingredient ribavirin. Ribavirin also has high aqueous solubility. Surprisingly at a very low disintegrant level of 1.03% the tablet disintegrated within 40 seconds. Additionally, Example 6f, which is a compact of 100% ribavirin, took 20 minutes to fully disintegrate indicating some level of synergy for disintegration of a mixture of simeprevir sodium and a crystalline active ingredient with high aqueous solubility.
[00245] Simeprevir sodium loaded onto silicon dioxide enabled a tablet formulation for the poorly wetting simeprevir. Simeprevir loaded onto silicon dioxide greatly improved the disintegration and dispersibility of the tablet matrix as compared to amorphous simeprevir sodium in the powdered state.
[00246] Additionally, it is expected that other crystalline, soluble active substances that have properties similar to ribavirin would similarly be useful in this composition.
[00247] With this knowledge, simeprevir sodium formulation compositions were determined for the reported fixed-dose combination examples.
TABLE 6
Figure imgf000058_0001
TABLE 7
Figure imgf000059_0001
Example 7a: Dasabuvir Tablet Formulation Development
[00248] Experiments were conducted to determine the optimal formulation composition for dasabuvir sodium (ABT-333) as a single drug to identify important formulation constituents and working levels to yield tablets of commercially useful hardness, size, and disintegration characteristics. The improved single drug formulations were useful for purposes of the fixed dose-combination examples that included dasabuvir.
[00249] Procedure:
1. A base formula of excipients were prepared by weighing and blending. This included microcrystalline cellulose as a compressible filler material, croscarmellose sodium as a disintegrant, and magnesium stearate as a lubricant.
2. Dasabuvir sodium active substance was weighed and then blended in specific
proportions of the base formula from #1 in this procedure.
3. Tablets of specific accurate masses were prepared individually using a Carver Model C hydraulic press using ¼-inch round tooling.
4. The tablets were evaluated for thickness (by caliper measurement), hardness (using a Varian VK 200 model tablet hardness tester), and disintegration time (by placing tablets in deionized water at ambient temperature (20-25°C) and observing the time to disintegrate and disperse granules and particles into suspension.
[00250] Ideally tablets have an acceptably high hardness and disintegrate rapidly in water. Where disintegration characteristics indicated slow or stalled disintegration, formulation parameters were changed seeking to optimize the disintegration characteristics. This also included the option of introducing additional functional excipients beyond those of the base formula. A summary of dasabuvir single drug formulation examples are tabulated and described below.
Example 7a:
[00251] At 10% dasabuvir sodium, in the base formula, acceptable hardness, thickness, and disintegration characteristics were observed. Based on microscopic evaluation of the dispersions after disintegration and based on assessment of the particle size distribution using a Focussed Beam Reflectance Measurement probe (manufactured by Mettler Toledo), it was apparent that the primary active substance particles were not as dispersed as could potentially be achieved for a highly wetting material. For utility in fixed dose combination tablets and to ensure maximal release of the active substance into solution for absorption in vivo, a surfactant addition was subsequently attempted.
Example 7b:
[00252] Improving on the prototype and observations in Example 7a, a surfactant was added to the base formula mixture with dasabuvir. Sodium lauryl sulfate (SLS) was used for this. By comparison to Example 7a, FBRM measurements for Example 7b indicated a finer dispersion of particles coincident with use of SLS surfactant as an excipient to improve the wettability of the dasabuvir active substance. This resulted in maintaining acceptable hardness, and disintegration characteristics of the tablet.
[00253] Beyond the simplest formulation composition using just dasabuvir and the base formula, the most effective formulation parameter for improving the tablet performance of dasabuvir sodium was via the addition of surfactant, and in this case SLS was used.
[00254] Other surfactants (for example ionic Tween 80, and non-ionic lecithin types of surfactants) would similarly improve the formulation performance in place of SLS as used here.
[00255] With this knowledge, dasabuvir sodium formulation compositions were determined for the reported fixed-dose combination examples.
TABLE 7
Figure imgf000060_0001
Figure imgf000061_0001
[00256] While embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments can be provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein can be employed in practicing the invention.

Claims

WHAT IS CLAIMED IS:
1. A pharmaceutical composition comprising:
a) a nonstructural protein 5A (NS5A) inhibitor or salt thereof;
b) a nonstructural protein 3/4A (NS3/4A) inhibitor or salt thereof; and
c) a nonstructural protein 5B (NS5B) inhibitor or salt thereof.
2. The pharmaceutical composition of claim 1, wherein said NS5A inhibitor or salt thereof comprises ACH-3102 or salt thereof.
3. The pharmaceutical composition of any one of claims 1 to 2, wherein said NS3/4A
inhibitor or salt thereof comprises simeprevir or salt thereof.
4. The pharmaceutical composition of any one of claims 1 to 3, further comprising
interferon.
5. The pharmaceutical composition of any one of claims 1 to 4, further comprising ribavirin or salt thereof.
6. The pharmaceutical composition of any one of claims 1 to 5, wherein said NS5B
inhibitor or salt thereof comprises a non-nucleoside inhibitor or salt thereof.
7. The pharmaceutical composition of any one of claims 1 to 6, wherein said NS5B
inhibitor or salt thereof comprises a nucleoside inhibitor or salt thereof.
8. The pharmaceutical composition of any one of claims 1 to 7, further comprising one or more additional drugs or salts thereof.
9. The pharmaceutical composition of claim 8, wherein said one or more additional drugs or salts thereof are daclatasvir or salt thereof, an NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, or any combination thereof.
10. The pharmaceutical composition of any one of claims 1 to 9, wherein said
pharmaceutical composition is a tablet.
11. The pharmaceutical composition of any one of claims 1 to 10, wherein said composition comprises a sustained release composition.
12. The pharmaceutical composition of claim 11, wherein said sustained release composition releases at least or at least about: 50, 60, 70, 80, 90, 95, or 99% by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, before or before about 24 hours based on the weight of the drug (w/w) within the composition.
13. The pharmaceutical composition of claim 11 or 12, wherein said release is determined in a Type II USP apparatus in 900 ml of apH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle, at from or from about 25 to 37C°.
14. The pharmaceutical composition of any one of claims 1 to 10, wherein said composition comprises an extended release composition.
15. The pharmaceutical composition of claim 14, wherein said extended release composition releases not more than or more than about: 5, 10, 20, 25, or 30% after 90 minutes, from or from about: 35, 40, or 45% to 50, 55, 60, or 65% after 4 hours, from or from about: 50, 55, 60, or 65% to 70, 75, 80 or 85% after 8 hours; from or from about: 70, 75, 80, or 85 to 90, 95, or 99% after 14 hours; and not less than or not less than about: 90, 95, 97, or 99% after 24 hours by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug (w/w) within the composition.
16. The pharmaceutical composition of claim 14 or 15, wherein said release is determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle, at from or from about 25 to 37C°.
17. The pharmaceutical composition of any one of claims 1 to 10, wherein said composition comprises a delayed release composition.
18. The pharmaceutical composition of claim 17, wherein said delayed release composition releases from or from about: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 15, 20, 25, or 30% after 90 minutes by weight individually of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, based on the weight of the drug (w/w) within the composition.
19. The pharmaceutical composition of claim 17 or 18, wherein said release is determined in a Type II USP apparatus in 900 ml of a pH 6.8 phosphate buffered, 0.1 N HC1 aqueous solution being agitated at 50 rotations per minute (rpm) by a paddle, at from or from about 25 to 37C°.
20. The pharmaceutical composition of any one of claims 1 to 19, wherein said NS5A
inhibitor or salt thereof, said NS3/4A inhibitor or salt thereof, and said NS5B inhibitor or salt thereof, are in separate sections or layers in the composition.
21. The pharmaceutical composition of any one of claims 1 to 20, wherein said
pharmaceutical composition comprises at least one immediate release, at least one extended release, or at least one delayed release section or layer in the composition.
22. The pharmaceutical composition of claim 20 or 21, comprising at least two or at least three or two or three layers or sections, wherein each section or layer comprises independently an immediate release, an extended release, or a delayed release section or layer in the composition.
23. The pharmaceutical composition of any one of claims 1 to 22, further comprising a coating.
24. The pharmaceutical composition of claim 23, wherein said coating comprises an enteric coating.
25. The pharmaceutical composition of any one of claims 1 to 24, wherein said composition comprises an immediate release composition.
26. The pharmaceutical composition of any one of claims 1 to 25, wherein one or more active ingredients or salts thereof inhibit HCV viral replication of one or more HCV genotypes.
27. The pharmaceutical composition of claim 26, wherein said one or more HCV genotypes comprises type I or IV.
28. The pharmaceutical composition of claim 26 or 27, wherein said one or more HCV
genotypes comprises type II.
29. The pharmaceutical composition of any one of claims 26 to 28, wherein said one or more HCV genotypes comprises type III.
30. A pharmaceutical composition comprising simeprevir or salt thereof loaded onto silica carrier particles.
31. The pharmaceutical composition of claim 30, wherein said silica carrier particles
comprises fumed silica.
32. The pharmaceutical composition of claims 30 or 31, further comprising one or more additional drugs or salts thereof.
33. The pharmaceutical composition of claim 32, wherein said one or more additional drugs or salts thereof comprise an NS5A inhibitor or salt thereof.
34. The pharmaceutical composition of claim 33, wherein said NS5A inhibitor or salt thereof comprises ACH-3102 or salt thereof.
35. The pharmaceutical composition of any one of claims 32 to 34, wherein said one or more additional drugs or salts thereof comprise an NS5B inhibitor or salt thereof.
36. The pharmaceutical composition of claim 35, wherein said NS5B inhibitor or salt thereof comprises a non-nucleoside inhibitor or salt thereof.
37. The pharmaceutical composition of claim 35 or 36, wherein said NS5B inhibitor or salt thereof comprises a nucleoside inhibitor or salt thereof.
38. The pharmaceutical composition of any one of claims 30 to 37, further comprising
interferon.
39. The pharmaceutical composition of any one of claims 30 to 38, further comprising
ribavirin or salt thereof.
40. The pharmaceutical compositions of any one of claims 1 to 39, wherein said composition comprises a dissolution profile of any one of claims 11 to 19.
41. The pharmaceutical composition of any one of claims 1 to 40, wherein said composition is in the form of a tablet.
42. The pharmaceutical composition of claim 41, wherein said tablet exhibits a hardness of less than or less than about 40, 30, 20, 10, 5, or 1 kp.
43. The pharmaceutical composition of any one of claims 1 to 42, further comprising fumed silica.
44. The pharmaceutical composition of any one of claims 1 to 43, wherein said NS3/4A inhibitor or salt thereof ranges from or from about: 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition (w/w).
45. The pharmaceutical composition of any one of claims 1 to 44, wherein the amount of said NS3/4A inhibitor or salt thereof is comprised of an amount from or from about 50 mg to 1000 mg.
46. The pharmaceutical composition of any one of claims 1 to 45, wherein said NS5A
inhibitor or salt thereof ranges from or from about: 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition (w/w).
47. The pharmaceutical composition of any one of claims 1 to 46, wherein the amount of said NS5A inhibitor or salt thereof is comprised of an amount from or from about 10 mg to 250 mg.
48. The pharmaceutical composition of any one of claims 1 to 47, wherein said NS5B inhibitor or salt thereof ranges from or from about: 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% to 30, 35, 40, 45, 50, 55, 55, or 60% by weight of the composition (w/w).
49. The pharmaceutical composition of any one of claims 1 to 48, wherein the amount of said NS5B inhibitor or salt thereof is comprised of an amount from or from about 10 mg to 1000 mg.
50. The pharmaceutical composition of any one of claims 1 to 49, wherein after oral
administration, Cmax of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, individually is from or is from about: 1.0, 1.5, 2.0, 2.5, or 3.0 pg/ml to 5.0, 10.0, 15.0, 20.0, or 25.0 ng/ml with plasma of patients with an average distribution volume of 50 L.
51. The pharmaceutical composition of any one of claims 1 to 50, wherein after oral
administration, Tmax of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, individually is from or is from about: 30, 60, 90, 120, 150, 180, 210, or 240 minutes.
52. The pharmaceutical composition of any one of claims 1 to 50, wherein after oral
administration, Tmax of the NS2 inhibitor or salt thereof, an NS3 inhibitor or salt thereof, an NS4A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, an NS4B inhibitor or salt thereof, an NS5A inhibitor or salt thereof, an NS5B inhibitor or salt thereof, the one or more additional drugs or salts thereof, or any combination thereof, individually is from or is from about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours.
53. A method of treating or preventing a viral infection comprising administering to a patient in need thereof, a pharmaceutical composition comprising a therapeutically effective amount of an NS5A inhibitor or salt thereof, an NS3/4A inhibitor or salt thereof, and an NS5B inhibitor or salt thereof.
54. A method of treating or preventing viral infections comprising administering to a patient in need thereof the pharmaceutical composition of any one of claims 2 to 29.
55. A method of treating or preventing viral infections comprising the pharmaceutical
compositions of any one of claims 30 to 52.
56. The method of any one of claims 53 to 55, wherein said viral infection is hepatitis.
57. The method of any one of claims 53 to 56, wherein said viral infection is hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
58. A method of making a pharmaceutical composition comprising loading simeprevir or a salt thereof onto silica carrier particles to form silica carrier particles loaded with simeprevir or salt thereof.
59. The method of claim 58, further comprising adding to said composition one or more additional drugs or salts thereof.
60. The method of claim 59, wherein said one or more additional drugs comprise an NS5A inhibitor or salt thereof.
61. The method of claim 60, wherein said NS5A inhibitor or salt thereof comprises ACH- 3102 or salt thereof.
62. The method of any one of claims 58 to 61, wherein said one or more additional drugs or salts thereof comprise an NS5B inhibitor or salt thereof.
63. The method of claim 62, wherein said NS5B inhibitor or salt thereof comprises a non- nucleoside inhibitor or salt thereof.
64. The method of 62 or 63, wherein said NS5B inhibitor or salt thereof comprises a
nucleoside inhibitor or salt thereof.
65. The method of any one of claims 58 to 64, further comprising adding to said composition interferon.
66. The method of any one of claims 58 to 65, further comprising adding to said composition ribavirin or salt thereof.
67. The method of claim 58 or 66, wherein said composition is compressed into a tablet.
68. The method of any one of claims 58 to 67, wherein said composition is formed into a bilayer or multilayer tablet.
69. A pharmaceutical composition comprising two or more compositions that inhibit two or more components of Figure 1.
70. The pharmaceutical composition of claim 69, comprising three or more compositions that inhibit three or more components of Figure 1.
71. The pharmaceutical composition of claim 69 or 70, comprising three or more
compositions that inhibit four or more components of Figure 1.
72. A pharmaceutical composition comprising two or more compositions that inhibit two or more components of Figure 1.
73. The use of the pharmaceutical composition of any one of claims 1 to 52 and 69 to 72, for the manufacture of a medicament for the treatment of viral disease in a subject.
74. The method of claim 73, wherein said viral infection is hepatitis.
75. The method of claim 73 or 74, wherein said viral infection is hepatitis A (HAV),
hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), hepatitis E (HEV), hepatitis F (HFV), hepatitis G (HGV), or any combination thereof.
PCT/US2015/053268 2014-09-30 2015-09-30 Fixed dose combinations for the treatment of viral diseases WO2016054240A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462071716P 2014-09-30 2014-09-30
US62/071,716 2014-09-30

Publications (1)

Publication Number Publication Date
WO2016054240A1 true WO2016054240A1 (en) 2016-04-07

Family

ID=54292955

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/053268 WO2016054240A1 (en) 2014-09-30 2015-09-30 Fixed dose combinations for the treatment of viral diseases

Country Status (1)

Country Link
WO (1) WO2016054240A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017017604A1 (en) * 2015-07-27 2017-02-02 Lupin Limited Substantially pure amorphous simeprevir sodium salt and the process for preparation thereof
WO2019030387A1 (en) * 2017-08-11 2019-02-14 Sandoz Ag Solid composition comprising amorphous sofosbuvir and amorphous daclatasvir

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3625214A (en) 1970-05-18 1971-12-07 Alza Corp Drug-delivery device
US4789734A (en) 1985-08-06 1988-12-06 La Jolla Cancer Research Foundation Vitronectin specific cell receptor derived from mammalian mesenchymal tissue
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US4906474A (en) 1983-03-22 1990-03-06 Massachusetts Institute Of Technology Bioerodible polyanhydrides for controlled drug delivery
US4925673A (en) 1986-08-18 1990-05-15 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents encapsulated with proteinoids
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US5928647A (en) 1993-01-11 1999-07-27 Dana-Farber Cancer Institute Inducing cytotoxic T lymphocyte responses
WO2003037379A1 (en) 2001-10-30 2003-05-08 Degussa Ag Use of granular materials based on pyrogenically produced silicon dioxide in pharmaceutical compositions
WO2004014852A2 (en) 2002-08-12 2004-02-19 Bristol-Myers Squibb Company Iminothiazolidinones as inhibitors of hcv replication
US20090041716A1 (en) 2007-08-08 2009-02-12 Bristol-Myers Squibb Company Crystalline form of methyl ((1s)-1-(((2s)-2-(5-(4'-(2-((2s)-1((2s)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)-2-pyrrolidinyl)-1h-imidazol-5-yl)-4-biphenylyl)-1h-imidazol-2-yl)-1-pyrrolidinyl)carbonyl)-2-methylpropyl)carbamate dihydrochloride salt
US20110306634A1 (en) * 2009-02-27 2011-12-15 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Amorphous Salt of a Macrocyclic Inhibitor of HCV
US20120014913A1 (en) 2010-07-16 2012-01-19 Abbott Laboratories Process for preparing antiviral compounds
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US8809313B2 (en) 2011-05-27 2014-08-19 Achillion Pharmaceuticals, Inc. Substituted aliphanes, cyclophanes, heteraphanes, heterophanes, hetero-heteraphanes and metallocenes useful for treating HCV infections

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3625214A (en) 1970-05-18 1971-12-07 Alza Corp Drug-delivery device
US4906474A (en) 1983-03-22 1990-03-06 Massachusetts Institute Of Technology Bioerodible polyanhydrides for controlled drug delivery
US4789734A (en) 1985-08-06 1988-12-06 La Jolla Cancer Research Foundation Vitronectin specific cell receptor derived from mammalian mesenchymal tissue
US4925673A (en) 1986-08-18 1990-05-15 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents encapsulated with proteinoids
US5942252A (en) 1986-10-24 1999-08-24 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissues and controlling their release
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US5814344A (en) 1986-10-24 1998-09-29 Southern Research Institute Method for delivering bioactive agents into and through the mucosally associated lymphoid tissues and controlling their release
US5820883A (en) 1986-10-24 1998-10-13 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissues and controlling their release
US5853763A (en) 1986-10-24 1998-12-29 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissue and controlling their release
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US5928647A (en) 1993-01-11 1999-07-27 Dana-Farber Cancer Institute Inducing cytotoxic T lymphocyte responses
WO2003037379A1 (en) 2001-10-30 2003-05-08 Degussa Ag Use of granular materials based on pyrogenically produced silicon dioxide in pharmaceutical compositions
WO2004014852A2 (en) 2002-08-12 2004-02-19 Bristol-Myers Squibb Company Iminothiazolidinones as inhibitors of hcv replication
WO2004014313A2 (en) 2002-08-12 2004-02-19 Bristol-Myers Squibb Company Combination pharmaceutical agents as inhibitors of hcv replication
US20090041716A1 (en) 2007-08-08 2009-02-12 Bristol-Myers Squibb Company Crystalline form of methyl ((1s)-1-(((2s)-2-(5-(4'-(2-((2s)-1((2s)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)-2-pyrrolidinyl)-1h-imidazol-5-yl)-4-biphenylyl)-1h-imidazol-2-yl)-1-pyrrolidinyl)carbonyl)-2-methylpropyl)carbamate dihydrochloride salt
US20110306634A1 (en) * 2009-02-27 2011-12-15 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Amorphous Salt of a Macrocyclic Inhibitor of HCV
US20120014913A1 (en) 2010-07-16 2012-01-19 Abbott Laboratories Process for preparing antiviral compounds
US8809313B2 (en) 2011-05-27 2014-08-19 Achillion Pharmaceuticals, Inc. Substituted aliphanes, cyclophanes, heteraphanes, heterophanes, hetero-heteraphanes and metallocenes useful for treating HCV infections
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
"Dermatological Formulations: Percutaneous absorption", 1983, MARCEL DEKKER INCL
"Handbook of Pharmaceutical Excipients", 2009
"Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2011, WILEY-VCH
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
"The Science and Practice of Pharmacy", LIPPINCOTT WILLIAMS & WILKINS
ANITA KOHLI ET AL: "Combination Oral, Hepatitis C Antiviral Therapy for 6 or 12 Weeks: Results of the SYNERGY Trial", 21ST CONFERENCE ON RETROVIRUSES AND OPPORTUNISTIC INFECTIONS; BOSTON, MA; MARCH 3 - 6, 2014,, 3 March 2014 (2014-03-03), pages 1 - 11, XP002740375, Retrieved from the Internet <URL:http://www.natap.org/2014/CROI/croi_04.htm> [retrieved on 20140306] *
ANSEL ET AL.: "Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPENCOTT WILLIAMS & WILKINS
CHAVANPATIL, M.D.; CHAUDHARI, S.; SHEAR, R.; VAVIA, P.R.: "Novel sustained release, swellable and bioadhesive gastroretentive drug delivery system for olfoxacin", INT. J. PHARM, 2006
DE CLERCQ ERIK ED - POIROT MARC ET AL: "Current race in the development of DAAs (direct-acting antivirals) against HCV", BIOCHEMICAL PHARMACOLOGY, vol. 89, no. 4, 13 April 2014 (2014-04-13), pages 441 - 452, XP029028847, ISSN: 0006-2952, DOI: 10.1016/J.BCP.2014.04.005 *
E. W. MARTIN: "Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING CO.
G. GREGORIADIS: "Drug Carriers in Biology and Medicine", 1979, ACADEMIC PRESS, article "Liposomes", pages: 2
GREGORY T. EVERSON ET AL: "Efficacy of an Interferon- and Ribavirin-Free Regimen of Daclatasvir, Asunaprevir, and BMS-791325 in Treatment-Naive Patients With HCV Genotype 1 Infection", GASTROENTEROLOGY, vol. 146, no. 2, 1 February 2014 (2014-02-01), PHILADELPHIA, PA, pages 420 - 429, XP055232160, ISSN: 0016-5085, DOI: 10.1053/j.gastro.2013.10.057 *
HOFFMAN, A.; STEPENSKY, D.; LAVY, E.; EYAL, S.; KLAUSNER, E.; FRIEDMAN, M.: "Pharmacokinetic and pharmacodynamic aspects of gastroretentive dosage forms", INT. J. PHARM, vol. 11, 2004, pages 141 - 53
JEIN, TIPS, vol. 19, 1998, pages 155 - 157
KLAUSNER, E.A.; LAVY, E.; BARTA, M.; CSEREPES, E.; FRIEDMAN, M.; HOFFMAN, A: "Novel gastroretentive dosage forms: evaluation of gastroretentivity and its effect on levodopa in humans", PHARM. RES., vol. 20, 2003, pages 1466 - 73
S. M. BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
STREUBEL, A.; SIEPMANN, J.; BODMEIER, R.: "Gastroretentive drug delivery systems", EXPERT OPIN. DRUG DELIVER., vol. 3, 2006, pages 217 - 3

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017017604A1 (en) * 2015-07-27 2017-02-02 Lupin Limited Substantially pure amorphous simeprevir sodium salt and the process for preparation thereof
WO2019030387A1 (en) * 2017-08-11 2019-02-14 Sandoz Ag Solid composition comprising amorphous sofosbuvir and amorphous daclatasvir

Similar Documents

Publication Publication Date Title
AU2018203696B2 (en) Combination formulation of two antiviral compounds
EP1441713B1 (en) Modified release tamsulosin tablets
AU2005253957B2 (en) Pharmaceutical compositions
KR100869039B1 (en) Nateglinide-containing preparations
JP6173521B2 (en) Formulations containing nalbuphine and their use
JP5600747B2 (en) Oral dispersible formulation
AU2011273519B2 (en) Pharmaceutical compositions comprising 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl) -1H-benzimidazol-2-yl]-1H-quinolin-2-one lactate monohydrate
TW201526899A (en) Pharmaceutical compositions
WO2001095912A1 (en) COMPOSITIONS CONTROLLING RELEASE pH RANGE AND/OR SPEED
US10105365B2 (en) Solid antiviral dosage forms
WO2016054240A1 (en) Fixed dose combinations for the treatment of viral diseases
JP2001064177A (en) Pharmaceutical preparation including benzamide derivative as ingredient
JP2005112825A (en) Gastric floating solid preparation
CA3190856A1 (en) Solid dosage forms of palbociclib
EP3305282A2 (en) Composition of pranlukast-containing solid preparation with improved bioavailability and method for preparing same
EP2197423B1 (en) Improved pharmaceutical composition containing a selective estrogen receptor modulator and method for the preparation thereof
JP2009530284A (en) Methods and compositions for treating diastolic heart failure
Lodhi et al. Review Study on Fast Dissolving Tablets Formulation of Antiemetic Medication
ES2862209T3 (en) Solid non-pulsatile prolonged-release oral compositions containing betahistine
TW202317145A (en) Advantageous anti-hcv combination therapy
CN112843053A (en) NS5A inhibitor composition
OA17450A (en) Combination formulation of two antiviral compounds.
BRPI0607372B1 (en) MEDICINAL PRODUCT FOR ORAL ADMINISTRATION UNDERSTANDING A CYCLOOXYGENASE-2 INHIBITOR AND PREPARATION METHOD

Legal Events

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

Ref document number: 15778852

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15778852

Country of ref document: EP

Kind code of ref document: A1