WO2013115916A1 - Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d'infections virales - Google Patents

Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d'infections virales Download PDF

Info

Publication number
WO2013115916A1
WO2013115916A1 PCT/US2012/069853 US2012069853W WO2013115916A1 WO 2013115916 A1 WO2013115916 A1 WO 2013115916A1 US 2012069853 W US2012069853 W US 2012069853W WO 2013115916 A1 WO2013115916 A1 WO 2013115916A1
Authority
WO
WIPO (PCT)
Prior art keywords
cobicistat
pharmaceutically acceptable
tenofovir alafenamide
acceptable salt
alafenamide hemifumarate
Prior art date
Application number
PCT/US2012/069853
Other languages
English (en)
Inventor
Srinivasan Ramanathan
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Priority to AU2012327170A priority Critical patent/AU2012327170A1/en
Publication of WO2013115916A1 publication Critical patent/WO2013115916A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • 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
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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

Definitions

  • Tenofovir ⁇ 9-R-[(2-phosphonomethoxy)propyl]adenine ⁇ , an acyclic nucleotide analog of dAMP, is a potent in vitro and in vivo inhibitor of human immunodeficiency virus type 1 (HTV-1) replication.
  • Tenofovir is sequentially phosphorylated in the cell by AMP kinase and nucleoside diphosphate kinase to the active species, tenofovir diphosphate, which acts as a competitive inhibitor of HIV-1 reverse transcriptase that terminates the growing viral DNA chain.
  • tenofovir circumvents an initial phosphorylation step that can be rate limiting for the activation of nucleoside analog inhibitors of HIV reverse transcriptase. Due to the presence of a phosphonate group, tenofovir is negatively charged at neutral pH, thus limiting its oral bioavailability.
  • VREAD® Tenofovir disoproxil fumarate
  • U.S. Patent No. 7,390,791 describes certain prodrugs of phosphonate nucleotide analogs that are useful in therapy.
  • One such prodrug is 9-[(R)-2-[[(S)- l-[[(S)-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]- methoxy]propyl] adenine 16:
  • GS-7340 exhibits potent anti-HIV activity 500- to 1000-fold enhanced activity relative to tenofovir against HIV-1 in T cells, activated peripheral blood mononuclear lymphocytes (PBMCs), and macrophages. GS-7340 also has enhanced ability to deliver and increase the accumulation of the parent tenofovir into PBMCs and other lymphatic tissues in vivo. It is also a potent inhibitor of hepatitis B virus.
  • GS-7340 is metabolized to tenofovir, which is not dependent on an intracellular nucleoside kinase activity for the first step in the conversion to the active metabolite, tenofovir diphosphate (PMPApp).
  • the cellular enzymes responsible for tenofovir metabolism to the active diphosphorylated form are adenylate kinase and nucleotide diphosphate kinase, which are highly active and ubiquitous.
  • Adenylate kinase exists as multiple isozymes (AK1 to AK4), with the phosphorylation of tenofovir mediated most efficiently by AK2.
  • GS-7340 has limited potential to alter cytochrome P450 enzyme activity through inhibition (IC 50 > 7 ⁇ compared to all isoforms tested). Similarly GS-7340 does not inhibit UGT1A1 function at concentrations up to 50 ⁇ . In addition, GS-7340 is not an activator of either the aryl hydrocarbon receptor or human pregnane X receptor.
  • GS-7340 When administered with cobicistat, GS-7340 was calculated to have a systemic exposure equivalent 2.2 fold higher than a dose of GS-7340 alone. In another case, GS-7340 administered with cobicistat was calculated to have a systemic exposure equivalent 3-4 fold higher than a dose of GS-7340 alone. In another case, GS-7340 administered with cobicistat was calculated to have a systemic exposure equivalent 1.3 fold higher than a dose of GS-7340 alone.
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the cobicistat may be coadministered with GS-7340.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • GS-7340 or a pharmaceutically acceptable salt thereof, and the cobicistat or pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount GS-7340 or a pharmaceutically acceptable salt thereof, and a daily amount cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, for improving the pharmacokinetics of GS-7340.
  • the cobicistat may be coadministered with GS-7340.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg. GS-7340, or a
  • a unit dosage form comprising a daily amount GS-7340 or a pharmaceutically acceptable salt thereof, and a daily amount cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, for improving the C max of GS-7340.
  • the cobicistat may be coadministered with GS-7340.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • GS-7340 or a pharmaceutically acceptable salt thereof, and the cobicistat or pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount GS-7340 or a pharmaceutically acceptable salt thereof, and a daily amount cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, for improving blood levels of GS-7340.
  • the cobicistat may be coadministered with GS-7340.
  • pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg. GS-7340, or a
  • a unit dosage form comprising a daily amount GS-7340 or a pharmaceutically acceptable salt thereof, and a daily amount cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • HAV human immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for a composition comprising a unit-dosage form of GS-7340 or a pharmaceutically acceptable salt thereof; a unit- dosage form of cobicistat, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or diluent.
  • the composition may include GS-7340 or a pharmaceutically acceptable salt thereof in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the composition may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100- 300 mg or 150 mg.
  • the unit dosage form may be a single daily dosage.
  • the invention provides for a kit comprising: (1) GS-7340, or a pharmaceutically acceptable salt thereof; (2) cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the GS-7340 or a
  • the kit may include GS-7340 or a pharmaceutically acceptable salt thereof in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the kit may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the invention provides for a method of treating a viral infection in a human comprising coadministering GS-7340 with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of cobicistat coadministered with the GS-7340 provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat.
  • GS-7340 or a pharmaceutically acceptable salt thereof in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below may be coadministered with cobicistat.
  • Cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg may be coadministered with GS-7340.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for a method for inhibiting activity of a retroviral reverse transcriptase in a human comprising coadministering GS-7340 with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of cobicistat coadministered with the GS-7340 provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat.
  • cobicistat in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below may be coadministered with cobicistat.
  • Cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg may be coadministered with GS-7340.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection.
  • the invention further provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection in a human.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount (or, in some embodiments throughout, in a therapeutic amount). GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg. Cobicistat may be used in an amount that provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat is used in the manufacture of the medicament.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound GS-7340 or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase.
  • the invention further provides for the use of the compound GS-7340 or a
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the cobicistat may be used in an amount that provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat is used in the manufacture of the medicament.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of GS-7340, or a pharmaceutically acceptable salt thereof, following administration to a human.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the cobicistat may be used in an amount that provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat is used in the manufacture of the medicament.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or a pharmaceutically acceptable salt thereof, following administration to a human.
  • GS-7340 or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount.
  • adenine ⁇ or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the cobicistat may be used in an amount that provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat is used in the manufacture of the medicament.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of GS-7340 by about 30-70%, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of GS-7340 by about 2 to 4 fold, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of GS-7340 by about 3 fold, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for a method of treating a viral infection in a human comprising coadministering 1) GS-7340 or a
  • GS-7340 or a pharmaceutically acceptable salt thereof is administered in a subtherapeutic amount.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • HBV human immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for a use of a subtherapeutic dose of GS-7340 coadministered with cobicistat for treating a viral infection.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of a subtherapeutic dose of GS-7340 coadministered with cobicistat for inhibiting retroviral reverse transcriptase.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for an anti-virus agent(s) comprising (a) a compound GS-7340 or a pharmaceutically acceptable salt thereof and (b) cobicistat, or a pharmaceutically acceptable salt thereof, in combination.
  • the anti-virus agent(s) may include GS-7340 or a pharmaceutically acceptable salt thereof may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the anti-virus agent(s) may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the cobicistat may be used in an amount that provides a systemic exposure of GS-7340 comparable to the systemic exposure obtainable by administration of a greater dose of GS-7340 in the absence of cobicistat is used in the manufacture of the medicament.
  • the anti-virus agent may further include 200 mg of emtricitabine and 150 mg of elvitegravir.
  • the anti-virus agent may further include 150mg cobicistat, 8 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the antivirus agent may further include 150mg cobicistat, 25or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may further include 150mg cobicistat, 10 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may include 150mg cobicistat, 8 mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti -virus agent may include 150 mg cobicistat, 10 mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides for a unit-dosage of GS-7340 or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, wherein the unit-dosage is a daily dose.
  • GS-7340 may be present in a subtherapeutic amount.
  • the unit-dosage may further include 150mg cobicistat, 8 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the unit-dosage may further include 150mg cobicistat, 25or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the unit-dosage may further include 150mg cobicistat, 10 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the unit-dosage may include 150 mg cobicistat, 10 mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or a pharmaceutically acceptable salt thereof, following administration to a human.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides cobicistat for use in improving the pharmacokinetics of ⁇ 9-[(R)-2-[[(S)-[[(S)-l-
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides a kit comprising: (1) ⁇ 9-[(R)-2- [[(S)- [ [(S)- 1 -(isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] - methoxy]propyl]adenine ⁇ , or a pharmaceutically acceptable salt thereof; (2) cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the ⁇ 9-[(R)-2- [[(S)-[[(S)-l-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]- methoxy]propyl]adenine ⁇ or a pharmaceutically acceptable salt thereof with the cobicistat or a pharmaceutically acceptable salt thereof.
  • the invention provides a kit comprising: (1) a unit dosage form comprising 5-100 mg of ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ , or a pharmaceutically acceptable salt thereof; (2) a unit dosage form comprising 150 mg cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the ⁇ 9-[(R)-2- [[(S)- [ [(S)- 1 -(isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] - methoxy]propyl]adenine ⁇ or a pharmaceutically acceptable salt thereof with cobicistat or a pharmaceutically acceptable salt thereof.
  • the invention provides a use of ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or its pharmaceutically acceptable salt for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase in a human, comprising administering GS-7340 or a pharmaceutically acceptable salt thereof, and cobicistat, or a pharmaceutically acceptable salt thereof to the human.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or its pharmaceutically acceptable salt; and cobicistat, or a pharmaceutically acceptable salt thereof; for use in inhibiting activity of a retroviral reverse transcriptase in a human.
  • the invention provides a use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament for a human useful for reducing a dose between about 30-70% of ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides the use of (9-[(R)-2-[[(S)-[[(S)- l-(isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]- methoxy]propyl]adenine ⁇ or a pharmaceutically acceptable salt thereof; and cobicistat or a pharmaceutically acceptable salt thereof for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides an anti-viral agent(s) comprising (a) ⁇ 9-[(R)-2-[[(S)-[[(S)-l- (isopropoxycarbonyl)ethyl] amino]phenoxyphosphinyl] -methoxy]propyl] adenine ⁇ or a pharmaceutically acceptable salt thereof, which is used in combination with (b) cobicistat or a pharmaceutically acceptable salt thereof for use in the prophylactic or therapeutic treatment of a viral infection in a human.
  • the invention provides for the use of the compound tenofovir or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, for the prophylactic or therapeutic treatment of a viral infection in a human.
  • Tenofovir may be used in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg, and 150 mg.
  • the tenofovir or a pharmaceutically acceptable salt thereof, and the cobicistat or pharmaceutically acceptable salt thereof may be coadministered.
  • the use may provide a unit dosage form comprising a daily amount tenofovir or a pharmaceutically acceptable salt thereof, and a daily amount cobicistat or pharmaceutically acceptable salt thereof is administered.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for a composition
  • a composition comprising a unit-dosage form of tenofovir or a pharmaceutically acceptable salt thereof; a unit- dosage form of cobicistat, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or diluent.
  • Tenofovir may be present in the composition in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg, and 150 mg.
  • the invention provides for a kit that includes (1) tenofovir, or a pharmaceutically acceptable salt thereof; (2) cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the tenofovir or a
  • Tenofovir may be present in the kit in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg, and 150 mg.
  • the invention provides for a method of treating a viral infection in a human that includes coadministering tenofovir with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of cobicistat coadministered with the tenofovir provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat.
  • Tenofovir may be administered in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • Cobicistat may be administered in amounts of 50-500 mg, 100-400 mg, 100-300 mg, and 150 mg.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B
  • HBV virus
  • the invention provides for a method for inhibiting activity of a retroviral reverse transcriptase in a human comprising coadministering tenofovir with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of tenofovir coadministered with the cobicistat provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat.
  • Tenofovir may be coadministered in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • Cobicistat may be coadministered in amounts of 50-500 mg, 100-400 mg, 100-300 mg, and 150 mg.
  • the virus may be human immunodeficiency virus (HIV)
  • the invention provides for the use of the compound tenofovir or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound tenofovir or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection in a human.
  • the tenofovir or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount (or, in some embodiments throughout, in a therapeutic amount).
  • Tenofovir may be administered in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • the cobicistat may be administered in an amount that provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat is used in the manufacture of the medicament. Cobicistat in an amount of 150 mg may be used in the manufacture of the medicament.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of the compound tenofovir or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for use of the compound tenofovir or a pharmaceutically acceptable salt thereof coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase in a human.
  • the tenofovir or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount.
  • Tenofovir may be used in amounts of less than 300 mg, 200 mg or less and 100 mg or less.
  • the cobicistat may be coadministered in an amount that provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat is used in the manufacture of the medicament. Cobicistat in an amount of 150 mg may be coadministered.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of tenofovir, or a pharmaceutically acceptable salt thereof, following administration to a human.
  • the tenofovir or a pharmaceutically acceptable salt thereof may be used in a subtherapeutic amount.
  • Tenofovir or a pharmaceutically acceptable salt thereof may be coadministered to the human in an amount of 100 mg or less, 200mg or less or in amount less than 300 mg.
  • Cobicistat may be used in an amount that provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat is used in the manufacture of the medicament. Cobicistat in an amount 150 mg may be used to prepare the medicament.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir by about 30-70%, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir by about 2 to 4 fold, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir by about 3 fold, or a pharmaceutically acceptable salt thereof, upon administration of the cobicistat.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a method of treating a viral infection in a human comprising coadministering 1) tenofovir or a
  • the tenofovir or a pharmaceutically acceptable salt thereof may be administered in a subtherapeutic amount.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a use of a subtherapeutic dose of tenofovir coadministered with cobicistat for treating a viral infection.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a use of a subtherapeutic dose of tenofovir coadministered with cobicistat for inhibiting retroviral reverse transcriptase.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for an anti-virus agent(s) comprising (a) a compound tenofovir or a pharmaceutically acceptable salt thereof and (b) cobicistat, or a pharmaceutically acceptable salt thereof, in combination.
  • the tenofovir may be present in the anti-virus agent(s) in a subtherapeutic amount.
  • the tenofovir may be present in the anti-virus agent(s) in an amount of 100 mg or less, 200 mg or less or less than 300 mg.
  • the cobicistat coadministered with the tenofovir may be present in the anti-virus agent(s) in an amount that provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat.
  • the antivirus agent may further include cobicistat in an amount of 150 mg.
  • the anti-virus agent may further include 200 mg of emtricitabine and 150 mg of elvitegravir.
  • the anti-virus agent may include 150mg cobicistat, 100 or less mg tenofovir, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti -virus agent may include 150mg cobicistat, 200 or less mg tenofovir, 150 mg elvitegravir, and 200 mg
  • the anti-virus agent may include 150mg cobicistat, less than 300 mg tenofovir, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may include 150mg cobicistat, 50 mg tenofovir, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a unit-dosage of tenofovir or a pharmaceutically acceptable salt thereof and cobicistat, or a pharmaceutically acceptable salt thereof, wherein the unit-dosage is a daily dose.
  • Tenofovir may be present in a subtherapeutic amount.
  • the unit-dosage may include 100 mg or less, 200 mg or less or less than 300 mg of tenofovir.
  • the unit-dosage may include an amount of cobicistat that provides a systemic exposure of tenofovir comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir in the absence of cobicistat.
  • the unit-dosage may include 150 mg of cobicistat.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • tenofovir alafenamide (isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine is tenofovir alafenamide.
  • the hemifumarate form of tenofovir alafenamide is also referred to herein as tenofovir alafenamide hemifumarate.
  • tenofovir alafenamide hemifumarate especially in combination with cobicistat and/or with other an additional therapeutic agent or agents.
  • tenofovir alafenamide hemifumarate wherein the ratio of fumaric acid to tenofovir alafenamide is 0.5 ⁇ 0.1, or
  • tenofovir alafenamide hemifumarate in a solid form.
  • tenofovir alafenamide hemifumarate that has an X-ray powder diffraction (XRPD) pattern having 2theta values of 6.9 ⁇ 0.2° and 8.6 ⁇ 0.2°.
  • XRPD X-ray powder diffraction
  • tenofovir alafenamide hemifumarate wherein the XRPD pattern comprises 2theta values of 6.9 ⁇ 0.2°, 8.6 ⁇ 0.2°, 1 1.0 ⁇ 0.2°, 15.9 ⁇ 0.2°, and 20.2 ⁇ 0.2°.
  • tenofovir alafenamide hemifumarate that has a differential scanning calorimetry (DSC) onset endotherm of 131 ⁇ 2 °C, or 131 ⁇ 1 °C.
  • DSC differential scanning calorimetry
  • a pharmaceutical composition comprising tenofovir alafenamide hemifumarate and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition further comprising an additional therapeutic agent.
  • the additional therapeutic agent is selected from the group consisting of human immunodeficiency virus (HIV) protease inhibiting compounds, HIV nonnucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, CCR5 inhibitors, and additional protease inhibiting compounds.
  • HIV human immunodeficiency virus
  • HIV infection comprising administering to a subject in need thereof a therapeutically effective amount of tenofovir alafenamide hemifumarate.
  • a method for treating an HIV infection comprising administering to a subject in need thereof a therapeutically effective amount of a pharmaceutical composition comprising tenofovir alafenamide hemifumarate.
  • the method comprises administering to the subject one or more additional therapeutic agents selected from the group consisting of HIV protease inhibiting compounds, HIV
  • nonnucleoside inhibitors of reverse transcriptase HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, CCR5 inhibitors, and additional protease inhibiting compounds.
  • a method for treating a hepatitis B virus (HBV) infection comprising administering to a subject in need thereof a therapeutically effective amount of tenofovir alafenamide hemifumarate.
  • a method for treating an HBV infection comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical composition comprising tenofovir alafenamide hemifumarate.
  • a method for preparing a pharmaceutical composition comprising combining tenofovir alafenamide hemifumarate and a pharmaceutically acceptable excipient to provide the pharmaceutical composition.
  • tenofovir alafenamide hemifumarate for use in medical therapy.
  • tenofovir alafenamide hemifumarate for the prophylactic or therapeutic treatment of an HIV infection.
  • use of tenofovir alafenamide hemifumarate to treat an HIV infection is provided.
  • use of tenofovir alafenamide hemifumarate for the preparation or manufacture of a medicament for the treatment of an HIV infection.
  • tenofovir alafenamide hemifumarate for use in treating an HIV infection.
  • tenofovir alafenamide hemifumarate for the prophylactic or therapeutic treatment of an HBV infection.
  • use of tenofovir alafenamide hemifumarate to treat an HBV infection is provided.
  • tenofovir alafenamide hemifumarate for the preparation or manufacture of a medicament for the treatment of an HBV infection.
  • tenofovir alafenamide hemifumarate for use in treating an
  • the methods of treating and the like comprise administration of multiple daily doses. In other embodiments, the methods of treating and the like comprise administration of a single daily dose.
  • the invention provides for the use of tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the cobicistat may be coadministered with tenofovir alafenamide hemifumarate.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Tenofovir alafenamide hemifumarate and cobicistat or pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount of tenofovir alafenamide hemifumarate, and a daily amount of cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for improving the pharmacokinetics of tenofovir alafenamide
  • Cobicistat may be coadministered with tenofovir alafenamide hemifumarate.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Tenofovir alafenamide hemifumarate and cobicistat, or pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount of tenofovir alafenamide hemifumarate, and a daily amount of cobicistat or pharmaceutically acceptable salt thereof may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for improving the C max of tenofovir alafenamide hemifumarate.
  • the cobicistat may be coadministered with tenofovir alafenamide hemifumarate.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Tenofovir alafenamide hemifumarate and cobicistat, or pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount of tenofovir alafenamide hemifumarate, and a daily amount of cobicistat, or a pharmaceutically acceptable salt thereof, may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for the use of tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for improving blood levels of tenofovir alafenamide hemifumarate.
  • the cobicistat may be coadministered with tenofovir alafenamide hemifumarate.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat, or a pharmaceutically acceptable salt thereof may be used in an amount of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof may be coadministered.
  • a unit dosage form comprising a daily amount tenofovir alafenamide hemifumarate, and a daily amount cobicistat, or a pharmaceutically acceptable salt thereof, may be used.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a composition
  • a composition comprising a unit-dosage form of tenofovir alafenamide hemifumarate; a unit-dosage form of cobicistat, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or diluent.
  • the composition may include tenofovir alafenamide hemifumarate in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the composition may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the unit-dosage form may be a single daily dosage.
  • the invention provides for a kit comprising: (1) tenofovir alafenamide hemifumarate; (2) cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the tenofovir alafenamide hemifumarate with the cobicistat, or the pharmaceutically acceptable salt thereof.
  • the kit may include tenofovir alafenamide hemifumarate in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the kit may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • the invention provides for a method of treating a viral infection in a human comprising coadministering tenofovir alafenamide hemifumarate with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of cobicistat coadministered with the tenofovir alafenamide hemifumarate provides a systemic exposure of tenofovir alafenamide
  • tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat.
  • Tenofovir alafenamide hemifumarate in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below may be coadministered with cobicistat.
  • Cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg may be coadministered with tenofovir alafenamide hemifumarate.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a method for inhibiting activity of a retroviral reverse transcriptase in a human comprising coadministering tenofovir alafenamide hemifumarate with cobicistat, or a pharmaceutically acceptable salt thereof, wherein the dose of cobicistat coadministered with the tenofovir alafenamide hemifumarate provides a systemic exposure of tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat.
  • Tenofovir alafenamide hemifumarate or a pharmaceutically acceptable salt thereof in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below may be coadministered with cobicistat.
  • Cobicistat in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg may be coadministered with tenofovir alafenamide hemifumarate.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for the use of tenofovir alafenamide hemifumarate coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection.
  • the invention further provides for the use of tenofovir alafenamide hemifumarate coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a viral infection in a human.
  • Tenofovir alafenamide hemifumarate may be used in a subtherapeutic amount (or, in some embodiments throughout, in a therapeutic amount).
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Cobicistat may be used in an amount that provides a systemic exposure of tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat in the manufacture of the medicament.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of tenofovir alafenamide hemifumarate coadministered with cobicistat, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase.
  • the invention further provides for the use of tenofovir alafenamide hemifumarate coadministered with cobicistat, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase in a human.
  • Tenofovir alafenamide hemifumarate may be used in a subtherapeutic amount.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Cobicistat may be used in an amount that provides a systemic exposure of tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat in the manufacture of the medicament.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of tenofovir alafenamide hemifumarate following administration to a human.
  • Tenofovir alafenamide hemifumarate may be used in a subtherapeutic amount.
  • Tenofovir alafenamide hemifumarate may be used in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • Cobicistat may be used in amounts of 50-500 mg, 100-400 mg, 100-300 mg or 150 mg.
  • Cobicistat may be used in an amount that provides a systemic exposure of tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat in the manufacture of the medicament.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir alafenamide hemifumarate by about 30-70% upon administration of the cobicistat.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir alafenamide hemifumarate by about 2-4 fold upon administration of the cobicistat.
  • the invention provides for the use of cobicistat, or a pharmaceutically acceptable salt thereof; to prepare a medicament for a human useful for reducing a dose of tenofovir alafenamide hemifumarate by about 3 fold upon administration of the cobicistat.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for a method of treating a viral infection in a human comprising coadministering 1) tenofovir alafenamide hemifumarate; and 2) cobicistat, or a pharmaceutically acceptable salt thereof, to the human.
  • Tenofovir alafenamide hemifumarate is administered in a
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • HIV human immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides for a use of a subtherapeutic dose of tenofovir alafenamide hemifumarate coadministered with cobicistat for treating a viral infection.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides for the use of a subtherapeutic dose of tenofovir alafenamide hemifumarate coadministered with cobicistat for inhibiting retroviral reverse transcriptase.
  • the virus may be human
  • HIV immunodeficiency virus
  • the invention provides for an anti-virus agent(s) comprising (a) tenofovir alafenamide hemifumarate and (b) cobicistat, or a pharmaceutically acceptable salt thereof, in combination.
  • the anti-virus agent(s) may include tenofovir alafenamide hemifumarate in amounts of 3 mg, 8 ⁇ 3 mg, 10 ⁇ 5 mg, 25 ⁇ 5 mg, or 40 ⁇ 10 mg or other ranges as set forth below.
  • the antivirus agent(s) may include cobicistat in amounts of 50-500 mg, 100-400 mg, 100- 300 mg or 150 mg.
  • the cobicistat may be used in an amount that provides a systemic exposure of tenofovir alafenamide hemifumarate comparable to the systemic exposure obtainable by administration of a greater dose of tenofovir alafenamide hemifumarate in the absence of cobicistat in the manufacture of the medicament.
  • the anti-virus agent may further include 200 mg of emtricitabine and 150 mg of elvitegravir.
  • the anti-virus agent may further include 150mg cobicistat, 8 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may further include 150mg cobicistat, 25 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may further include 150mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may include 150mg cobicistat, 8 mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the anti-virus agent may include 150 mg cobicistat, 10 mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides for a unit-dosage of tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, wherein the unit-dosage is a daily dose.
  • Tenofovir alafenamide hemifumarate may be present in a subtherapeutic amount.
  • the unit-dosage may further include 150 mg cobicistat, 8 or less mg tenofovir alafenamide
  • the unit-dosage may further include 150 mg cobicistat, 25or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the unit-dosage may further include 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the unit-dosage may include 150 mg cobicistat, 10 mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides the use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament useful for improving the pharmacokinetics of tenofovir alafenamide hemifumarate following administration to a human.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides cobicistat for use in improving the pharmacokinetics of tenofovir alafenamide hemifumarate following administration to a human.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human
  • HIV immunodeficiency virus
  • HBV Hepatitis B virus
  • the invention provides a kit comprising: (1) tenofovir alafenamide hemifumarate; (2) cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the tenofovir alafenamide hemifumarate with the cobicistat or a pharmaceutically acceptable salt thereof.
  • the invention provides a kit comprising: (1) a unit dosage form comprising 5-100 mg of tenofovir alafenamide hemifumarate; (2) a unit dosage form comprising 150 mg cobicistat, or a pharmaceutically acceptable salt thereof; (3) one or more containers; and (4) prescribing information regarding administering the tenofovir alafenamide hemifumarate with cobicistat or a pharmaceutically acceptable salt thereof.
  • the invention provides a use of tenofovir alafenamide hemifumarate for the manufacture of a medicament for inhibiting activity of a retroviral reverse transcriptase in a human, comprising administering tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, to the human.
  • the virus may be human immunodeficiency virus (HIV).
  • the invention provides tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for use in inhibiting activity of a retroviral reverse transcriptase in a human.
  • the invention provides a use of cobicistat, or a pharmaceutically acceptable salt thereof, to prepare a medicament for a human useful for reducing a dose between about 30-70% of tenofovir alafenamide hemifumarate upon administration of the cobicistat.
  • the medicament may be used for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides the use of tenofovir alafenamide hemifumarate and cobicistat, or a pharmaceutically acceptable salt thereof, for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the use may be for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the virus may be human immunodeficiency virus (HIV) or Hepatitis B virus (HBV).
  • the invention provides an anti-viral agent(s) comprising (a) tenofovir alafenamide hemifumarate, which is used in combination with (b) cobicistat, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of a viral infection in a human.
  • the invention provides for the use of ritonavir in the compositions, kits, unit-dosages and uses set forth above in place of cobicistat.
  • the invention provides a method for inhibiting intestinal Pgp-mediated intestinal secretion of GS-7340, or a pharmaceutically acceptable salt thereof, in a human by coadministration of cobicistat, or a pharmaceutically acceptable salt thereof, with GS-7340, or a pharmaceutically acceptable salt thereof.
  • 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof is coadministered with 10 mg of GS-7340, or a pharmaceutically acceptable salt thereof.
  • the invention provides a method for inhibiting intestinal Pgp-mediated intestinal secretion of tenofovir alafenamide hemifumarate in a human by coadministration of cobicistat, or a pharmaceutically acceptable salt thereof, with tenofovir alafenamide hemifumarate.
  • cobicistat or a pharmaceutically acceptable salt thereof
  • 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof is coadministered with 10 mg of tenofovir alafenamide hemifumarate.
  • the invention provides the use of an anti-virus agent for the prophylactic or therapeutic treatment of a viral infection in a human, wherein the anti-virus agent comprises 150 mg cobicistat, 10 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides a method of treating a viral infection in a human comprising coadministering 150 mg cobicistat, 10 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine to the human.
  • the invention provides the use of 150 mg cobicistat, 10 or less mg GS-7340, 150 mg elvitegravir, and 200 mg emtricitabine for the manufacture of a medicament for treating a viral infection in a human.
  • the invention provides the use of an anti-virus agent for the prophylactic or therapeutic treatment of a viral infection in a human, wherein the anti-virus agent comprises 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine.
  • the invention provides a method of treating a viral infection in a human comprising coadministering 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg
  • the invention provides the use of 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 150 mg elvitegravir, and 200 mg emtricitabine for the manufacture of a medicament for treating a viral infection in a human.
  • the invention provides an anti-virus agent(s) comprising (a) tenofovir alafenamide hemifumarate, (b) cobicistat, or a pharmaceutically acceptable salt thereof, (c) emtricitabine, and (d) darunavir, in combination.
  • the invention provides an anti-virus agent(s) comprising (a) 8 or less mg of tenofovir alafenamide hemifumarate, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof, (c) 200 mg of emtricitabine, and (d) 800 mg of darunavir, in combination.
  • the invention provides an anti-virus agent(s) comprising (a) 25 or less mg of tenofovir alafenamide hemifumarate, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof, (c) 200 mg of emtricitabine, and (d) 800 mg of darunavir, in combination.
  • the invention provides an anti-virus agent(s) comprising (a) 10 mg of tenofovir alafenamide hemifumarate, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof, (c) 200 mg of emtricitabine, and (d) 800 mg of darunavir, in combination.
  • the invention provides an anti-virus agent(s) comprising (a) GS-7340, or a pharmaceutically acceptable salt thereof,
  • the invention provides an anti-virus agent(s) comprising (a) 8 or less mg of GS-7340, or a pharmaceutically acceptable salt thereof, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof,
  • the invention provides an anti-virus agent(s) comprising (a) 25 or less mg of GS-7340, or a pharmaceutically acceptable salt thereof, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof, (c) 200 mg of emtricitabine, and (d) 800 mg of darunavir, in combination.
  • the invention provides an anti-virus agent(s) comprising (a) 10 mg of GS-7340, or a pharmaceutically acceptable salt thereof, (b) 150 mg of cobicistat, or a pharmaceutically acceptable salt thereof, (c) 200 mg of emtricitabine, and (d) 800 mg of darunavir, in combination.
  • the invention provides the use of an anti-virus agent for the prophylactic or therapeutic treatment of a viral infection in a human, wherein the anti-virus agent comprises 150 mg cobicistat, 10 or less mg GS-7340, 800 mg of darunavir, and 200 mg emtricitabine.
  • the invention provides a method of treating a viral infection in a human comprising coadministering 150 mg cobicistat, 10 or less mg
  • GS-7340 800 mg of darunavir, and 200 mg emtricitabine to the human.
  • the invention provides the use of 150 mg cobicistat, 10 or less mg GS-7340, 800 mg of darunavir, and 200 mg emtricitabine for the manufacture of a medicament for treating a viral infection in a human.
  • the invention provides the use of an anti-virus agent for the prophylactic or therapeutic treatment of a viral infection in a human, wherein the anti-virus agent comprises 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 800 mg of darunavir, and 200 mg emtricitabine.
  • the invention provides a method of treating a viral infection in a human comprising coadministering 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 800 mg of darunavir, and 200 mg emtricitabine to the human.
  • the invention provides the use of 150 mg cobicistat, 10 or less mg tenofovir alafenamide hemifumarate, 800 mg of darunavir, and 200 mg emtricitabine for the manufacture of a medicament for treating a viral infection in a human.
  • the invention provides the use of a dose of a cytochrome p450 inhibitor, or a pharmaceutically acceptable salt thereof, to boost a dose GS-7340, or a pharmaceutically acceptable salt thereof, for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the cytochrome p450 inhibitor is cobicistat, or a pharmaceutically acceptable salt thereof.
  • the dose of GS-7340 would be a
  • the invention provides a composition comprising: a unit-dosage form of GS-7340, or a pharmaceutically acceptable salt thereof; a unit- dosage form of cobicistat, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or diluent, wherein the amount of GS-7340 in the unit-dosage form is a subtherapeutic amount.
  • the invention provides the use of a dose of a cytochrome p450 inhibitor, or a pharmaceutically acceptable salt thereof, to boost a dose tenofovir alafenamide hemifumarate for the prophylactic or therapeutic treatment of a viral infection in a human.
  • the cytochrome p450 inhibitor is cobicistat, or a pharmaceutically acceptable salt thereof.
  • the dose of tenofovir alafenamide hemifumarate would be a subtherapeutic amount absent the dose of cobicistat.
  • the invention provides a composition comprising: a unit-dosage form of tenofovir alafenamide hemifumarate; a unit-dosage form of cobicistat, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or diluent, wherein the amount of tenofovir alafenamide hemifumarate in the unit-dosage form is a subtherapeutic amount.
  • the invention provides the uses and methods related to treating a viral infection, as noted herein, wherein the viral infection is human immunodeficiency virus (HIV).
  • HIV human immunodeficiency virus
  • the invention provides the uses and methods related to treating a viral infection, as noted herein, wherein the viral infection is Hepatitis B virus (HBV).
  • HBV Hepatitis B virus
  • Figure 1 shows pharmacokinetic data from patients dosed with various doses of GS-7340 and tenofovir.
  • Figure 2 shows pharmacokinetic data from patients dosed with various doses of GS-7340 and tenofovir.
  • Figure 3 shows pharmacokinetic data from patients dosed with various formulations of GS-7340.
  • Figure 4 shows pharmacokinetic data from patients dosed with various formulations of GS-7340.
  • Figure 5 shows pharmacokinetic data from patients dosed with various formulations of GS-7340.
  • Figure 6 shows pharmacokinetic data from patients dosed with various formulations of GS-7340.
  • Figure 7 shows pharmacokinetic data from patients dosed with various formulations of GS-7340.
  • Figure 8 shows pharmacokinetic data from patients dosed with various formulations of GS-7340or tenofovir.
  • Figure 9 shows pharmacokinetic data from patients dosed with various formulations of GS-7340 or tenofovir.
  • Figure 10 shows results of substrate assays in cells transfected with the genes for human P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) genes.
  • Figure 1 1 shows results of bidirectional permeability assays in cells transfected with the genes for human P-glycoprotein and breast cancer resistance protein genes.
  • Figure 12 shows results of bidirectional permeability assays in cells transfected with the genes for human P-glycoprotein and breast cancer resistance protein genes.
  • Figure 13 shows the X-ray powder diffraction (XRPD) pattern of tenofovir alafenamide hemifumarate.
  • Figure 14 shows a graph of the DSC analysis of tenofovir alafenamide hemifumarate.
  • Figure 15 shows a graph of the thermogravimetric analysis (TGA) data for tenofovir alafenamide hemifumarate.
  • Figure 16 shows a graph of the dynamic vapor sorption (DVS) analysis of tenofovir alafenamide hemifumarate.
  • Cobicistat (chemical name l,3-thiazol-5-ylmethyl (2R,5R)-(5- ⁇ [(2S)-2- [(methyl ⁇ [2-(propan-2-yl)-l,3-thiazol-4-yl]methyl ⁇ carbamoyl)amino]]-4- (morpholin-4-yl)butanamido ⁇ -l,6-diphenylhexan-2-yl)carbamate) is a chemical entity that has been shown to be a mechanism-based inhibitor that irreversibly inhibits CYP3A enzymes.
  • Cobicistat was found to be an efficient inactivator of human hepatic microsomal CYP3A activity with kinetic parameters similar to those of ritonavir.
  • cobicistat is a moderate inhibitor of CYP2B6 (similar potency to ritonavir), a weak inhibitor of CYP2D6, and does not appreciably inhibit CYP1A2, CYP2C8, CYP2C9, CYP2C19, or uridine glucuronosyltransferase 1A1.
  • cobicistat displayed no/weak potential as an inducer of cytochrome P450, UGT1A1, or P-glycoprotein (at up to 30 ⁇ ). Permeability assays suggest that cobicistat is not a strong substrate or inhibitor of transporters including P-glycoprotein, MRP1, and MRP2. Inhibition of intestinal
  • cobicistat is only possible during absorption due to its high aqueous solubility, but it is not potent enough to inhibit transporters at systemic concentrations.
  • Cobicistat may also be present in compositions enriched with a stereoisomer of formula (la):
  • the cobicistat has an enriched concentration of 85 ⁇ 5% of the stereoisomer of formula (la). In another embodiment the cobicistat has an enriched concentration of 90 ⁇ 5% of the stereoisomer of formula (la). In another embodiment the cobicistat has an enriched concentration of 95 ⁇ 2% of the stereoisomer of formula (la). In another embodiment the cobicistat has an enriched concentration of 99 ⁇ 1% of the stereoisomer of formula (la). In another embodiment the cobicistat is present as the pure stereoisomer of formula (la).
  • GS-7340 or tenofovir alafenamide hemifumarate coadministered with cobicistat may be administered in lower amounts than previously thought to achieve a therapeutic effect. Such lower amounts may be subtherapeutic amounts if cobicistat is not coadministered.
  • cobicistat may be acting to inhibit intestinal Pgp-mediated intestinal secretion of GS-7340 or tenofovir alafenamide hemifumarate.
  • cobicistat and ritonavir significantly increased the accumulation of probe substrates (such as calcein AM and Hoechst 33342) in cells transfected with P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP), and cobicistat was found to be a substrate for these transporters.
  • probe substrates such as calcein AM and Hoechst 33342
  • Pgp P-glycoprotein
  • BCRP breast cancer resistance protein
  • Cobicistat appears to be a relatively weak inhibitor of Pgp and BCRP and may only have a transient effect on these transporters during intestinal absorption, facilitated by high solubility of and resulting high concentrations of cobicistat achievable in the gastrointestinal tract. Combined, these results suggest that cobicistat can effectively inhibit intestinal transporters and increase the absorption of coadministered substrates, including HIV protease inhibitors and GS-7340 or tenofovir alafenamide hemifumarate, contributing to its effectiveness as a pharmacoenhancer.
  • administer refers to administration of two or more agents within a 24 hour period of each other, for example, as part of a clinical treatment regimen.
  • dose refers to administration of two or more agents within a 24 hour period of each other, for example, as part of a clinical treatment regimen.
  • administer refers to administration of two or more agents within 2 hours of each other. In other embodiments, “coadminister” refers to administration of two or more agents within 30 minutes of each other. In other embodiments,
  • “coadminister” refers to administration of two or more agents within 15 minutes of each other. In other embodiments, “coadminister” refers to administration at the same time, either as part of a single formulation or as multiple formulations that are administered by the same or different routes.
  • unit dosage form refers to a physically discrete unit, such as a capsule, tablet, or solution that is suitable as a unitary dosage for a human patient, each unit containing a predetermined quantity of one or more active ingredient(s) calculated to produce a therapeutic effect, in association with at least one pharmaceutically acceptable diluent or carrier, or combination thereof.
  • Unit dosage formulations contain a daily dose or unit daily subdose or an appropriate fraction thereof, of the active ingredient.
  • subtherapeutic amount of a compound is any amount of the compound that upon dosing is insufficient to achieve the desired therapeutic benefit.
  • boosting amount or “boosting dose” is the amount of a compound needed to improve the pharmacokinetics of a second compound (or increase availability or exposure).
  • the boosting amount or boosting dose may improve the pharmacokinetics (or increase availability or exposure) of the second compound to a level to therapeutic levels in a subject.
  • the present invention also provides a method for the treatment or prophylaxis of diseases, disorders, and conditions.
  • An example of a disease, disorder, or condition includes, but is not limited to, a retrovirus infection, or a disease, disorder, or condition associated with a retrovirus infection.
  • Retroviruses are RNA viruses and are generally classified into the alpharetrovirus,
  • retroviruses include, but are not limited to, human immunodeficiency virus (HIV), human T-lymphotropic virus (HTLV), rous sarcoma virus (RSV), and the avian leukosis virus.
  • gag group-specific antigen
  • pol polymerase
  • env envelopee gene
  • retroviruses attach to and invade a host cell by releasing a complex of RNA and the pol products, among other things, into the host cell. The reverse transcriptase then produces double stranded DNA from the viral RNA.
  • the double stranded DNA is imported into the nucleus of the host cell and integrated into the host cell genome by the viral integrase.
  • a nascent virus from the integrated DNA is formed when the integrated viral DNA is converted into mRNA by the host cell polymerase and the proteins necessary for virus formation are produced by the action of the virus protease.
  • the virus particle undergoes budding and is released from the host cell to form a mature virus.
  • the active agents may be administered to a human in any conventional manner. While it is possible for the active agents to be administered as raw compounds, they are preferably administered as a pharmaceutical composition.
  • the salt, carrier, or diluent should be acceptable in the sense of being compatible with the other ingredients and not deleterious to the recipient thereof.
  • Examples of carriers or diluents for oral administration include cornstarch, lactose, magnesium stearate, talc, microcrystalline cellulose, stearic acid, povidone, crospovidone, dibasic calcium phosphate, sodium starch glycolate, hydroxypropyl cellulose (e.g., low substituted hydroxypropyl cellulose), hydroxypropylmethyl cellulose (e.g., hydroxypropylmethyl cellulose 2910), and sodium lauryl sulfate.
  • cornstarch lactose, magnesium stearate, talc, microcrystalline cellulose, stearic acid, povidone, crospovidone, dibasic calcium phosphate, sodium starch glycolate, hydroxypropyl cellulose (e.g., low substituted hydroxypropyl cellulose), hydroxypropylmethyl cellulose (e.g., hydroxypropylmethyl cellulose 2910), and sodium lauryl sulfate.
  • hydroxypropyl cellulose e
  • compositions may be prepared by any suitable method, such as those methods well known in the art of pharmacy, for example, methods such as those described in Gennaro et al., Remington's Pharmaceutical Sciences (18th ed., Mack Publishing Co., 1990), especially Part 8: Pharmaceutical
  • Such methods include the step of bringing into association GS-7340 or tenofovir alafenamide hemifumarate with the carrier or diluent and optionally one or more accessory ingredients.
  • accessory ingredients include those conventional in the art, such as, fillers, binders, excipients, disintegrants, lubricants, colorants, flavoring agents, sweeteners, preservatives (e.g., antimicrobial preservatives), suspending agents, thickening agents, emulsifying agents, and/or wetting agents.
  • GS-7340 or pharmaceutically acceptable salt thereof includes any amorphous, crystalline, co-crystalline, complex, or other physical form thereof.
  • a composition comprising a pharmaceutically acceptable coformer and GS-7340 is administered.
  • the pharmaceutically acceptable coformer can be any pharmaceutically acceptable compound that is capable of forming a "pharmaceutically acceptable salt" (as described above) with GS-7340.
  • the pharmaceutically acceptable coformer can be a pharmaceutically acceptable acid (e.g. adipic acid, L-aspartic acid, citric acid, fumaric acid, maleic acid, malic acid, malonic acid, succinic acid, tartaric acid, or oxalic acid).
  • the pharmaceutically acceptable coformer is a bis-acid. In another embodiment the pharmaceutically acceptable coformer is fumaric acid. In another embodiment, a composition comprising a coformer and GS-7340 in a ratio of about 0.5 ⁇ 0.05 can be administered.
  • GS-7340 is a hemifumarate form (tenofovir alafenamide hemifumarate).
  • the pharmaceutical compositions may provide controlled, slow release, or sustained release of the agents (e.g., GS-7340 or tenofovir alafenamide hemifumarate) over a period of time.
  • the controlled, slow release, or sustained release of the agents may maintain the agents in the bloodstream of the human for a longer period of time than with conventional formulations.
  • compositions include, but are not limited to, coated tablets, pellets, solutions, powders, and capsules, and dispersions of GS-7340 or tenofovir alafenamide hemifumarate in a medium that is insoluble in physiologic fluids or where the release of the therapeutic compound follows degradation of the pharmaceutical composition due to mechanical, chemical, or enzymatic activity.
  • the pharmaceutical compositions of the invention may be, for example, in the form of a pill, capsule, solution, powder, or tablet, each containing a predetermined amount of GS-7340 or tenofovir alafenamide hemifumarate.
  • the pharmaceutical composition is in the form of a tablet comprising GS-7340 or tenofovir alafenamide hemifumarate.
  • the pharmaceutical composition is in the form of a tablet comprising GS-7340 and the components of the tablet utilized and described in the Examples herein.
  • fine powders or granules may contain diluting, dispersing, and or surface active agents and may be present, for example, in water or in a syrup, in capsules or sachets in the dry state, or in a nonaqueous solution or suspension wherein suspending agents may be included, or in tablets wherein binders and lubricants may be included.
  • the formulation When administered in the form of a liquid solution or suspension, the formulation may contain GS-7340 or tenofovir alafenamide hemifumarate and purified water.
  • Optional components in the liquid solution or suspension include suitable sweeteners, flavoring agents, preservatives (e.g., antimicrobial preservatives), buffering agents, solvents, and mixtures thereof.
  • a component of the formulation may serve more than one function.
  • a suitable buffering agent also may act as a flavoring agent as well as a sweetener.
  • Suitable sweeteners include, for example, saccharin sodium, sucrose, and mannitol. A mixture of two or more sweeteners may be used. The sweetener or mixtures thereof are typically present in an amount of from about 0.001% to about 70% by weight of the total composition. Suitable flavoring agents may be present in the pharmaceutical composition to provide a cherry flavor, cotton candy flavor, or other suitable flavor to make the pharmaceutical composition easier for a human to ingest. The flavoring agent or mixtures thereof are typically present in an amount of about 0.0001% to about 5% by weight of the total composition.
  • Suitable preservatives include, for example, methylparaben,
  • propylparaben sodium benzoate
  • benzalkonium chloride A mixture of two or more preservatives may be used.
  • the preservative or mixtures thereof are typically present in an amount of about 0.0001% to about 2% by weight of the total composition.
  • Suitable buffering agents include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts. A mixture of two or more buffering agents may be used. The buffering agent or mixtures thereof are typically present in an amount of about 0.001% to about 4% by weight of the total composition.
  • Suitable solvents for a liquid solution or suspension include, for example, sorbitol, glycerin, propylene glycol, and water. A mixture of two or more solvents may be used. The solvent or solvent system is typically present in an amount of about 1% to about 90% by weight of the total composition.
  • the pharmaceutical composition may be coadministered with adjuvants.
  • adjuvants such as polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether may be administered with or incorporated into the pharmaceutical composition to artificially increase the permeability of the intestinal walls.
  • Enzymatic inhibitors may also be administered with or incorporated into the pharmaceutical composition.
  • GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 8 ⁇ 3 mg, 8 ⁇ 2 mg or 8 ⁇ 1 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a unit dosage form comprises a dose of
  • a dose of 8 ⁇ 3 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 25 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 10 ⁇ 5 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 25 ⁇ 5 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 25 ⁇ 2 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 40 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 40 ⁇ 5 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 40 ⁇ 2 mg of GS-7340 or a pharmaceutically acceptable salt thereof is administered.
  • a dose of 60 ⁇ 20 mg of GS-7340 or a pharmaceutically acceptable salt thereof is administered.
  • a dose of 60 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 100 ⁇ 20 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 100 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 125 ⁇ 20 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 125 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 150 ⁇ 20 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 150 ⁇ 10 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 200 ⁇ 40 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • a dose of 200 ⁇ 15 mg of GS-7340 or a pharmaceutically acceptable salt thereof, is administered.
  • the effective daily dose of GS-7340 may be administered as one, two, three, four, five, six, or more subdoses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • the concentration of GS-7340 in the bloodstream may be measured as the plasma concentration (e.g., ng/mL).
  • Pharmacokinetic parameters for determining the plasma concentration include, but are not limited to, the maximum observed plasma concentration (C max ), observed plasma concentration at the end of the dosing interval or "trough" concentration (C tau or C mm ), area under the plasma concentration time curve (AUC) from time zero up to the last quantifiable time point (AUCo-iast), AUC from time zero to infinity (AUCo-inf), AUC over the dosing interval (AUC tau ), time of maximum observed plasma concentration after administration (t max ), and half-life of GS-7340 in plasma (ti/ 2 ).
  • Administration of GS-7340 with food according to the methods of the invention may also increase absorption of GS-7340.
  • Absorption of GS-7340 may be measured by the concentration attained in the bloodstream over time after administration of GS-7340.
  • An increase in absorption by administration of GS-7340 with food may also be evidenced by an increase in C max and/or AUC of GS-7340 as compared to the values if GS-7340 was administered without food.
  • protease inhibitors are administered with food.
  • a hemifumarate form of tenofovir alafenamide i.e., tenofovir alafenamide hemifumarate.
  • This form may have a ratio (i.e., a stoichiometric ratio or mole ratio) of fumaric acid to tenofovir alafenamide of 0.5 ⁇ 0.1, 0.5 ⁇ 0.05, 0.5 ⁇ 0.01, or about 0.5, or the like.
  • tenofovir alafenamide hemifumarate consists of fumaric acid and tenofovir alafenamide in a ratio of 0.5 ⁇ 0.1.
  • tenofovir alafenamide hemifumarate consists essentially of fumaric acid and tenofovir alafenamide in a ratio of 0.5 ⁇ 0.1.
  • tenofovir alafenamide hemifumarate has an XRPD pattern comprising 2theta values of 6.9 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.0 ⁇ 0.2°, 1 1.0 ⁇ 0.2°, 12.2 ⁇ 0.2°, 15.9 ⁇ 0.2°, 16.3 ⁇ 0.2°, 20.2 ⁇ 0.2°, and 20.8 ⁇ 0.2°.
  • tenofovir alafenamide hemifumarate has an XRPD pattern comprising at least four 2theta values selected from 6.9 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.0 ⁇ 0.2°, 1 1.0 ⁇ 0.2°, 12.2 ⁇ 0.2°, 15.9 ⁇ 0.2°, 16.3 ⁇ 0.2°, 20.2 ⁇ 0.2°, and 20.8 ⁇ 0.2°.
  • tenofovir alafenamide hemifumarate has a DSC onset endotherm of 131 ⁇ 2 °C, or 131 ⁇ 1 °C.
  • a tenofovir alafenamide hemifumarate composition comprises less than about 5% by weight of tenofovir alafenamide monofumarate.
  • a tenofovir alafenamide hemifumarate composition comprises less than about 1% by weight of tenofovir alafenamide monofumarate.
  • a tenofovir alafenamide hemifumarate composition comprises less than about 0.5% by weight of tenofovir alafenamide monofumarate. [0200] In one embodiment, a tenofovir alafenamide hemifumarate composition comprises no detectable tenofovir alafenamide monofumarate.
  • a dose of 3 mg, 3 ⁇ 2 or 3 ⁇ 1 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 8 ⁇ 3 mg, 8 ⁇ 2 mg or 8 ⁇ 1 mg of tenofovir alafenamide hemifumarate is administered.
  • a unit dosage form comprises a dose of 8 ⁇ 2 mg of tenofovir alafenamide hemifumarate.
  • a dose of 8 ⁇ 3 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 25 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 10 ⁇ 5 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 25 ⁇ 5 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 25 ⁇ 2 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 40 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 40 ⁇ 5 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 40 ⁇ 2 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 60 ⁇ 20 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 60 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 100 ⁇ 20 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 100 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 125 ⁇ 20 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 125 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 150 ⁇ 20 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 150 ⁇ 10 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 200 ⁇ 40 mg of tenofovir alafenamide hemifumarate is administered.
  • a dose of 200 ⁇ 15 mg of tenofovir alafenamide hemifumarate is administered.
  • the effective daily dose of tenofovir alafenamide hemifumarate may be administered as two, three, four, five, six, or more subdoses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • the concentration of GS-7340 or tenofovir alafenamide hemifumarate in the bloodstream may be measured as the plasma concentration (e.g., ng/mL).
  • Pharmacokinetic parameters for determining the plasma concentration include, but are not limited to, the maximum observed plasma concentration (C max ), observed plasma concentration at the end of the dosing interval or "trough" concentration (Ctau or Cmin), area under the plasma concentration time curve (AUC) from time zero up to the last quantifiable time point (AUCo-i as t), AUC from time zero to infinity (AUCo-inf), AUC over the dosing interval (AUC tau ), time of maximum observed plasma concentration after administration (t max ), and half-life of GS-7340 or tenofovir alafenamide hemifumarate in plasma (t ⁇ ).
  • Administration of GS-7340 or tenofovir alafenamide hemifumarate with food according to the methods of the invention may also increase absorption of GS-7340 or tenofovir alafenamide hemifumarate.
  • Absorption of GS-7340 or tenofovir alafenamide hemifumarate may be measured by the concentration attained in the bloodstream over time after administration of GS-7340 or tenofovir alafenamide hemifumarate.
  • An increase in absorption by administration of GS-7340 or tenofovir alafenamide hemifumarate with food may also be evidenced by an increase in C max and/or AUC of GS-7340 or tenofovir alafenamide hemifumarate as compared to the values if GS-7340 or tenofovir alafenamide hemifumarate was administered without food.
  • protease inhibitors are administered with food.
  • tenofovir alafenamide hemifumarate can be prepared using selective crystallization.
  • An example of a scheme for this preparation method is as follows.
  • the method can be carried out by subjecting a solution comprising: a) a suitable solvent; b) fumaric acid; c) tenofovir alafenamide; and, optionally, d) one or more seeds comprising tenofovir alafenamide hemifumarate, to conditions that provide for the crystallization of fumaric acid and tenofovir alafenamide.
  • the starting solution can contain the single diastereomer of tenofovir alafenamide or a mixture of tenofovir alafenamide and one or more of its other diastereomers (e.g., GS-7339, as described in U.S. Patent No. 7,390,791).
  • the selective crystallization can be carried out in any suitable solvent.
  • the solvent comprises a protic solvent (e.g., water or isopropyl alcohol).
  • the solvent comprises an aprotic organic solvent (e.g., acetone, acetonitrile (ACN), toluene, ethyl acetate, isopropyl acetate, heptane, tetrahydrofuran (THF), 2-methyl THF, methyl ethyl ketone, or methyl isobutyl ketone, or a mixture thereof).
  • the solvent comprises ACN or a mixture of ACN and up to about 50% methylene chloride (by volume).
  • the selective crystallization also can be carried out at any suitable temperature, for example, a temperature in the range of from about 0 °C to about 70 °C. In one specific embodiment, the resolution is carried out at a temperature of about 0 °C.
  • tenofovir alafenamide (isopropoxycarbonyl)ethyl]amino]phenoxyphosphinyl]methoxy]propyl]adenine; described in, e.g., U.S. Patent No. 7,390,791), which is the major diastereomeric impurity in the active pharmaceutical ingredient.
  • the hemifumarate form of tenofovir alafenamide can be more readily and easily separated from impurities than the monofumarate form.
  • Other major advantages of tenofovir alafenamide hemifumarate over the monofumarate form include improved thermodynamic and chemical stability (including long-term storage stability), superior process reproducibility, superior drug product content uniformity, and a higher melting point.
  • Tenofovir alafenamide hemifumarate is useful in the treatment and/or prophylaxis of one or more viral infections in man or animals, including infections caused by DNA viruses.
  • RNA viruses herpesviruses (e.g., CMV, HSV 1, HSV 2, VZV), retroviruses, hepadnaviruses (e.g., HBV), papillomavirus, hantavirus, adenoviruses and HIV.
  • herpesviruses e.g., CMV, HSV 1, HSV 2, VZV
  • retroviruses e.g., hepadnaviruses
  • papillomavirus hantavirus
  • adenoviruses adenoviruses and HIV.
  • U.S. Patent No. 6,043,230 (incorporated by reference herein in its entirety) and other publications describe the antiviral specificity of nucleotide analogs, such as tenofovir disoproxil
  • Tenofovir alafenamide hemifumarate can be administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including ocular, buccal, and sublingual), vaginal, and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal, and epidural). Generally, tenofovir alafenamide hemifumarate is administered orally, but it can be administered by any of the other routes noted herein.
  • compositions include those suitable for topical or systemic administration, including oral, rectal, nasal, buccal, sublingual, vaginal, or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal, and epidural) administration.
  • parenteral including subcutaneous, intramuscular, intravenous, intradermal, intrathecal, and epidural administration.
  • the formulations are in unit dosage form and are prepared by any of the methods well known in the art of pharmacy.
  • hemifumarate may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such pharmaceutical compositions and preparations will typically contain at least 0.1% of tenofovir alafenamide hemifumarate.
  • the percentage of this active compound in the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% or more of the weight of a given unit dosage form.
  • the amount of active compound in such therapeutically useful pharmaceutical compositions is preferably such that an effective dosage level will be obtained upon administration of a single-unit dosage (e.g., tablet).
  • dosage formulations may provide therapeutically effective amounts of tenofovir alafenamide hemifumarate upon repeated administration of subclinically effective amounts of the same.
  • Preferred unit dosage formulations include those containing a daily dose (e.g., a single daily dose), as well as those containing a unit daily subclinical dose, or an appropriate fraction thereof (e.g., multiple daily doses), of tenofovir alafenamide
  • compositions suitable for oral administration may be presented as discrete units such as capsules, cachets, or tablets, each containing a predetermined amount of tenofovir alafenamide hemifumarate; as a powder or granules; as a solution or a suspension in an aqueous liquid or a nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • Tenofovir alafenamide hemifumarate may also be presented as a bolus, electuary, or paste.
  • Tenofovir alafenamide hemifumarate is preferably administered as part of a pharmaceutical composition or formulation.
  • Such pharmaceutical composition or formulation comprises tenofovir alafenamide hemifumarate together with one or more pharmaceutically acceptable carriers / excipients, and optionally other therapeutic ingredients.
  • the excipient(s) / carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the patient.
  • Excipients include, but are not limited to, substances that can serve as a vehicle or medium for tenofovir alafenamide hemifumarate (e.g., a diluent carrier). They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • the tablets, troches, pills, capsules, and the like may also contain, without limitation, the following: a binder(s), such as hydroxypropyl cellulose, povidone, or hydroxypropyl methylcellulose; a filler(s), such as microcrystalline cellulose, pregelatinized starch, starch, mannitol, or lactose monohydrate; a disintegrating agent(s), such as croscarmellose sodium, cross- linked povidone, or sodium starch glycolate; a lubricant(s), such as magnesium stearate, stearic acid, or other metallic stearates; a sweetening agent(s), such as sucrose, fructose, lactose, or aspartame; and/or a flavoring agent(s), such as peppermint, oil of wintergreen, or a cherry flavoring.
  • a binder(s) such as hydroxypropyl cellulose, povidone, or hydroxypropyl methylcellulose
  • the unit dosage form When the unit dosage form is a capsule, it may contain, in addition to materials of the above types, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, polymers, wax, shellac, or sugar and the like. Of course, any material used in preparing any unit dosage form typically will be pharmaceutically acceptable and substantially nontoxic in the amounts employed. In addition, tenofovir alafenamide hemifumarate may be incorporated into sustained-release preparations and devices.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, polymers, wax, shellac, or sugar and the like.
  • any material used in preparing any unit dosage form typically will
  • the pharmaceutical compositions are preferably applied as a topical ointment or cream containing tenofovir alafenamide hemifumarate in an amount of, for example, 0.01 to 10% w/w (including active ingredient in a range between 0.1% and 5% in increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 3% w/w and most preferably 0.5 to 2% w/w.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base.
  • compositions suitable for topical administration in the mouth include lozenges comprising tenofovir alafenamide hemifumarate in a flavored basis, for example, sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate.
  • compositions suitable for parenteral administration are sterile and include aqueous and nonaqueous injection solutions that may contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions that may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials with elastomeric stoppers, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier (e.g., water for injections) immediately prior to use.
  • Injection solutions and suspensions may be prepared from sterile powders, granules, and tablets of the kind previously described.
  • compositions / formulations may include other ingredients conventional in the art, having regard to the type of formulation in question.
  • veterinary compositions comprising tenofovir alafenamide hemifumarate together with a veterinary carrier therefor.
  • Veterinary carriers are materials useful for the purpose of administering the composition to cats, dogs, horses, rabbits, and other animals, and may be solid, liquid, or gaseous materials that are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally, or by any other desired route.
  • the tenofovir alafenamide hemifumarate can be used to provide controlled release pharmaceutical formulations containing a matrix or absorbent material and an active ingredient of the invention, in which the release of the active ingredient can be controlled and regulated to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the compound.
  • Controlled release formulations adapted for oral administration in which discrete units comprising a compounds of the invention, can be prepared according to conventional methods.
  • Useful dosages of tenofovir alafenamide hemifumarate can be determined by comparing in vitro activities, and the in vivo activities in animal models.
  • the amount of tenofovir alafenamide hemifumarate required for use in treatment will vary with several factors, including but not limited to the route of administration, the nature of the condition being treated, and the age and condition of the patient; ultimately, the amount administered will be at the discretion of the attendant physician or clinician.
  • the therapeutically effective amount / dose of tenofovir alafenamide hemifumarate depends, at least, on the nature of the condition being treated, any toxicity or drug interaction issues, whether the compound is being used prophylactically (e.g., sometimes requiring lower doses) or against an active disease or condition, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies.
  • the oral dose of tenofovir alafenamide hemifumarate may be in the range from about 0.0001 to about 100 mg/kg body weight per day, for example, from about 0.01 to about 10 mg/kg body weight per day, from about 0.01 to about 5 mg/kg body weight per day, from about 0.5 to about 50 mg/kg body weight per day, from about 1 to about 30 mg/kg body weight per day, from about 1.5 to about 10 mg/kg body weight per day, or from about 0.05 to about 0.5 mg/kg body weight per day.
  • the daily candidate dose for an adult human of about 70 kg body weight will range from about 0.1 mg to about 1000 mg, or from about 1 mg to about 1000 mg, or from about 5 mg to about 500 mg, or from about 1 mg to about 150 mg, or from about 5 mg to about 150 mg, or from about 5 mg to about 100 mg, or about 10 mg, and may take the form of single or multiple doses.
  • the oral dose of tenofovir alafenamide hemifumarate may be in the form of a combination of agents (e.g., tenofovir alafenamide hemifumarate / emtricitabine / elvitegravir / cobicistat).
  • compositions described herein may further include one or more therapeutic agents in addition to tenofovir alafenamide hemifumarate.
  • the additional therapeutic agent can be selected from the group consisting of HIV protease inhibiting compounds, HIV nonnucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucleotide inhibitors of reverse transcriptase, HIV integrase inhibitors, and CCR5 inhibitors.
  • Therapeutic methods include administering tenofovir alafenamide hemifumarate to a subject / patient in need of the same as a therapeutic or preventative treatment.
  • tenofovir alafenamide hemifumarate may be administered to a subject / patient having a medical disorder or to a subject who may acquire the disorder.
  • Such treatment is given in order to ameliorate, prevent, delay, cure, and/or reduce the severity of a symptom or set of symptoms of a disorder (including a recurring disorder).
  • the treatment may also be given to prolong the survival of a subject, e.g., beyond the survival time expected in the absence of such treatment.
  • the medical disorders that may be treated with tenofovir alafenamide hemifumarate include those discussed herein, including without limitation, HIV infection and HBV infection.
  • the resulting combination When cobicistat or a pharmaceutically acceptable salt thereof is combined with certain specific solid carrier particles (e.g. silica derivatives), the resulting combination possesses improved physical properties. Even though cobicistat is hygroscopic in nature, the resulting combination has comparatively low hygroscopicity. Additionally, the resulting combination is a free-flowing powder, with high loading values for cobicistat, acceptable physical and chemical stability, rapid drug release properties, and excellent compressibility. Thus, the resulting combination can readily be processed into solid dosage forms (e.g. tablets), which possess good drug release properties, low tablet friability, good chemical and physical stability, and a low amount of residual solvents.
  • the compositions of the invention represent a significant advance that facilitates the commercial development of cobicistat for use in treating viral infections such as HIV.
  • Cobicistat 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 include 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 such as NEUSILIN
  • loaded on a solid carrier includes, but is not limited to a compound of formula (I) being coated in the pores and on the surface of a solid carrier.
  • 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.
  • a specific silica material that is particularly 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 materials having physical and chemical properties similar to the silica materials described herein can also be used.
  • ritonavir may be used in the manner that cobicistat is used to boost the circulating levels of GS-7340, tenofovir, or tenofovir alafenamide hemifumarate, to improve the pharmacokinetics of GS-7340, tenofovir, or tenofovir alafenamide hemifumarate and achieve the other advantages of the use of cobicistat as disclosed herein.
  • an HIV nonnucleoside inhibitor of reverse transcriptase e.g., capravirine, emivirine, delaviridine, efavirenz, nevirapine, (+) calanolide A, etravirine, GW5634, DPC-083, DPC-961, DPC-963, MTV-150, and TMC-120, TMC-278 (rilpivirene), BILR 355 BS, VRX 840773, UK-453061, and RDEA806; [0257] 3) an HIV nucleoside inhibitor of reverse transcriptase, e.g., zidovudine, emtricitabine, didanosine, stavudine, zalcitabine, lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, racivir ( ⁇ -FTC), D-d4FC, phosphazide
  • an HIV nucleotide inhibitor of reverse transcriptase e.g., tenofovir disoproxil fumarate and adefovir dipivoxil;
  • an HIV integrase inhibitor e.g., curcumin, derivatives of curcumin, chicoric acid, derivatives of chicoric acid, 3,5-dicaffeoylquinic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of tyrphostin, quercetin, derivatives of quercetin, S-1360, zintevir (AR-177), L-870812, and L-870810, MK-0518 (raltegravir), elvitegravir, BMS-538158, GSK364735C, BMS-707035, MK-2048, and BA 011 ;
  • curcumin e.g., curcumin, derivatives of curcumin, chicoric acid, derivative
  • gp41 inhibitor e.g., enfuvirtide, sifuvirtide, FB006M, and TRI-1144;
  • a CXCR4 inhibitor e.g., AMD-070
  • an entry inhibitor e.g., SP01A
  • gpl20 inhibitor e.g., BMS-488043 or BlockAide/ CR
  • a G6PD and NADH-oxidase inhibitor e.g., immunitin
  • a CCR5 inhibitor e.g., aplaviroc, vicriviroc, maraviroc, PRO- 140,
  • [0266] 12) other drugs for treating HIV e.g., BAS-100, SPI-452, REP 9, SP-01A, TNX-355, DES6, ODN-93, ODN-112, VGV-1, PA-457 (bevirimat), Ampligen, HRG214, Cytolin, VGX-410, KD-247, AMZ 0026, CYT 99007A-221 HIV, DEBIO-025, BAY 50-4798, MDX010 (ipilimumab), PBS 119, ALG 889, and PA- 1050040 (PA-040);
  • BAS-100 e.g., BAS-100, SPI-452, REP 9, SP-01A, TNX-355, DES6, ODN-93, ODN-112, VGV-1, PA-457 (bevirimat), Ampligen, HRG214, Cytolin, VGX-410, KD-247, AMZ 0026, CYT 99007A-221 HIV, DEB
  • an interferon e.g., pegylated rIFN-alpha 2b, pegylated rIFN-alpha 2a, rIFN-alpha 2b, rIFN-alpha 2a, consensus IFN alpha (infergen), feron, reaferon, intermax alpha, r-IFN-beta, infergen + actimmune, IFN-omega with DUROS, albuferon, locteron, Albuferon, Rebif, Oral interferon alpha, IFNalpha-2b XL, AVI-005, PEG-Infergen, and Pegylated IFN-beta;
  • an interferon e.g., pegylated rIFN-alpha 2b, pegylated rIFN-alpha 2a, rIFN-alpha 2b, rIFN-alpha 2a, consensus IFN alpha (infergen), feron, reaferon
  • a ribavirin analog e.g., rebetol, copegus, viramidine (taribavirin);
  • a NS5b polymerase inhibitor e.g., NM-283, valopicitabine, R1626, PSI-6130 (R1656), HCV-796, BILB 1941, XTL-2125, MK-0608, NM-107, R7128 (R4048), VCH-759, PF-868554, and GSK625433;
  • a NS3 protease inhibitor e.g., SCH-503034 (SCH-7), VX-950 (telaprevir), BILN-2065, BMS-605339, and ITMN-191;
  • an alpha-glucosidase 1 inhibitor e.g., MX-3253 (celgosivir), UT-231B;
  • hepatoprotectants e.g., IDN-6556, ME 3738, LB-84451, and MitoQ;
  • a nonnucleoside inhibitor of HCV e.g., benzimidazole derivatives, benzo-l,2,4-thiadiazine derivatives, phenylalanine derivatives, A-831, GS-9190, and A-689; and
  • Exemplary combinations include (a) emtricitabine / darunavir / cobicistat / GS-7340;
  • combination (b) above can include 200 mg of emtricitabine, 800 mg of darunavir, 150 mg of cobicistat, and 10 mg of tenofovir alafenamide
  • the distillation apparatus was then removed and the solution was cooled to 20 °C.
  • the solution was seeded with 2.0% compound 16 and allowed to stir for one hour at 20 °C.
  • Phenol (9.9 g, 0.1 1 mol, 0.1 equiv) and DBU (16 g, 0.1 1 mol, 0.1 equiv) were added and the mixture was stirred for an additional 24 hours or until the weight percent of compound 16 remaining in solution was less than 12%.
  • the slurry was then cooled to 0 °C and stirred for an additional 18 hours at 0 °C.
  • the slurry was filtered and washed with a 1 : 1 solution of isopropyl acetate:acetonitrile (1.5 L) at 0 °C.
  • the mixture was transferred to a separatory funnel and washed sequentially with 10 % w/v aqueous solution of sodium phosphate monobasic (2 x 50 mL), 15 % w/v aqueous solution of potassium bicarbonate (2 x 20 mL), and water (50 mL).
  • the final organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to a viscous amber oil.
  • the intermediate formed is the PMPA anhydride and is at 6 ppm; The product is at 1 1 ppm.
  • the reaction is deemed complete when less than 5% anhydride is present).
  • the reaction mixture was distilled to -1.5 volumes of acetonitrile and diluted with ethyl acetate (200 mL) and water (300 mL).
  • the aqueous layer was separated and washed with ethyl acetate (200 mL) twice.
  • the aqueous layer was re-charged to the vessel and pH adjusted to pH 3 using 12.1 M HCl (21.0 mL).
  • the reaction was then seeded with 0.05% of compound 12 seed and allowed to stir at 25 °C.
  • the solids were dried in a vacuum oven at 40 °C overnight. 5.52 g off-white solids were obtained. The solids were analyzed by XRPD and found to contain tenofovir alafenamide monofumarate, GS-7339 monofumarate, and tenofovir alafenamide hemifumarate.
  • the mixture was cooled over a minimum of 4 hours to 0-6 °C, and then agitated at 0-6 °C for a minimum of 1 hour.
  • the resulting slurry was filtered and rinsed with chilled (0-6 °C) ACN (2 kg).
  • the product was dried under vacuum below 45 °C until loss on drying (LOD) and organic volatile impurities (OVI) limits were met (LOD ⁇ 1.0%, dichloromethane content ⁇ 0.19%, acetonitrile content ⁇ 0.19%) to afford the final compound of the hemifumarate form of tenofovir alafenamide as a white to off- white powder (typical yield is about 0.95 kg).
  • Tenofovir alafenamide hemifumarate is anhydrous, nonhygroscopic, and has a DSC onset endotherm of about 131 °C.
  • the XRPD pattern for tenofovir alafenamide hemifumarate is shown in Figure 13.
  • the characteristic peaks include: 6.9 ⁇ 0.2°, 8.6 ⁇ 0.2°, 10.0 ⁇ 0.2°, 1 1.0 ⁇ 0.2°, 12.2 ⁇ 0.2°, 15.9 ⁇ 0.2°, 16.3 ⁇ 0.2°, 20.2 ⁇ 0.2°, and 20.8 ⁇ 0.2°.
  • the crystal size was 0.32 x 0.30 x 0.20 mm 3 .
  • the sample was held at 123 K and the data was collected using a radiation source with a wavelength of 0.71073 A in the theta range of 1.59 to 25.39°.
  • Conditions of, and data collected from the single-crystal X-ray diffraction are shown in Table 1.
  • the hemifumarate form of tenofovir alafenamide from Synthetic Example 8 has an exceptional capability to purge this diastereomeric impurity, as compared with the capability of the monofumarate form (described in, e.g., U.S. Patent
  • Stable form screening of tenofovir alafenamide hemifumarate showed that it is thermodynamically stable in most solvents, such as ACN, toluene, ethyl acetate, methyl tert-butyl ether (MTBE), acetone, THF, and 2-methyl THF.
  • solvents such as ACN, toluene, ethyl acetate, methyl tert-butyl ether (MTBE), acetone, THF, and 2-methyl THF.
  • a similar stable form screening of the monofumarate form showed that this form is not thermodynamically stable in the above-listed solvents.
  • the monofumarate form of tenofovir alafenamide When suspended in these solvents, the monofumarate form of tenofovir alafenamide fully converts to the hemifumarate form in THF and 2-methyl THF, and partially converts to the hemifumarate form in ACN, ethyl acetate, MTBE, and acetone, as well as at ambient temperatures.
  • the hemifumarate form of tenofovir alafenamide has a melting point that is about 10 °C higher than that of the monofumarate form, indicating that the hemifumarate form has improved thermal stability as compared with the monofumarate form.
  • Test compounds were diluted in cell culture medium containing 10 ⁇ Hoechst 33342 and incubated for 3 hours with MDCKII-BCRP and nontransfected cells. Following removal of media containing Hoechst 33342 and test compound, cells were washed twice with warm medium and lysed at room temperature for 5-10 minutes in a buffer containing 20 mM Tris-HCl pH 9.0 and 0.4% Triton X-100. Wells were analyzed for Hoechst 33342 fluorescence at an excitation of 353 nm and an emission of 460 nm.
  • MDCKII cells were grown to confluence over 4 to 6-days on 24-well PET transwell plates (BD Biosciences). The same buffers were used in the donor and receiver wells as described above for caco-2 studies. Experiments were conducted as described above for caco-2 transepithelial transport studies and samples analyzed by LC/MS/MS. Similar quality control and acceptance criteria were used as those described above for caco-2 studies. TEER values and the permeability of lucifer yellow, atenolol, and propranolol were determined to meet acceptance criteria for each batch of assay plates. Efflux ratios were determined to be at least 3-fold higher in transfected versus nontransfected monolayers for the model Pgp substrate vinblastine and BCRP substrate prazosin.
  • Pgp and BCRP substrate assays in transfected MDCKII cells were completed in cells transfected with the genes for the human transport proteins to determine if cobicistat is a substrate for these efflux transporters ( Figure 10).
  • the overexpression of Pgp or BCRP in MDCKII cells increased the efflux ratios of cobicistat. These increased efflux ratios reflected a decrease in the forward permeability and an increase in the reverse permeability of cobicistat.
  • cobicistat efflux was decreased in the presence of the Pgp inhibitor CSA and the BCRP inhibitor Kol34.
  • Coadministration of cobicistat and ritonavir comparably reduced the efflux ratios by increasing the A-B flux and decreasing the B-A flux of the protease inhibitors (Figure 12A-D).
  • the effect of cobicistat on GS-7340 permeability across caco-2 monolayers was monitored over 2 hours.
  • GS-7340 Pharmacokinetic studies were done in humans to determine exposure to GS-7340 at three dose levels. Eligible subjects were randomized to receive either GS-7340 dose of 8 mg, GS-7340 dose of 25 mg, GS-7340 dose of 40 mg, tenofovir 300 mg or placebo-to-match GS-7340 for 10 days. (Note: Doses of GS-7340 are given as the mass of free base of GS-7340, even where other forms of GS-7340 were dosed.) GS-7340 was administered in a blinded fashion, unless a subject was randomized to receive tenofovir which was given on an open-label basis.
  • Figure 1 shows tenofovir plasma concentrations in patients on Day 1 of the study.
  • the top line (dashed line) shows the concentration of tenofovir in patients dosed with 300 mg tenofovir.
  • the next line down (triangles pointed down) shows the concentration of tenofovir in patients dosed with 40 mg GS-7340.
  • the next line down (triangles pointed up) shows the concentration of tenofovir in patients dosed with 25 mg GS-7340.
  • the bottom line (squares) shows the concentration of tenofovir in patients dosed with 8 mg GS-7340.
  • the table below the chart shows Cmax and AUC values obtained.
  • Figure 2 shows tenofovir plasma concentrations in patients on Day 10 of the study.
  • the top line shows the concentration of tenofovir in patients dosed with 300 mg tenofovir.
  • the next line down shows the concentration of tenofovir in patients dosed with 40 mg GS-7340.
  • the next line down shows the concentration of tenofovir in patients dosed with 25 mg GS-7340.
  • the bottom line shows the concentration of tenofovir in patients dosed with 8 mg GS-7340.
  • the table below the chart shows Cmax and AUC values obtained.
  • Table 4 show the dosing regimen and schedule for the study.
  • Tenofovir (TFV) concentrations are shown in the graph below for doses of emtricitabine and GS-7340 (upper line, right side up triangles) and emtricitabine, GS-7340 and efavirenz (lower line, upside down triangles).
  • Cmax and AUC results are displayed in the tables to the right for GS-7340 and tenofovir exposure.
  • Figure 4 shows GS-7340 concentrations (ng/ml) for doses of emtricitabine and GS-7340 (upper line, right side up triangles) and emtricitabine, GS-7340, darunavir and cobicistat (lower line, upside down triangles) in patients from Cohort 2.
  • Tenofovir (TFV) concentrations are shown in the graph below for doses of emtricitabine and GS-7340 (upper line, right side up triangles) and emtricitabine, GS-7340, darunavir, and cobicistat (lower line, upside down triangles).
  • Cmax and AUC results are displayed in the tables to the right for GS-7340 and tenofovir exposure.
  • Figure 5 shows GS-7340 concentrations (ng/ml) for doses of GS-7340 alone and GS-7340 and cobicistat (upper line, right side up triangles) in patients from Cohort 1.
  • Tenofovir (TFV) concentrations are shown in the graph below for doses of GS-7340 alone and GS-7340 and cobicistat (upper line, right side up triangles).
  • Cmax and AUC results are displayed in the tables to the right for GS-7340 and tenofovir exposure.
  • GS-7340 and cobicistat were administered in conjunction with elvitegravir and emtricitabine in a clinical trial to determine the relative bioavailability of these compounds.
  • the compounds were administered using a 25 mg or 40 mg dose of GS-7340 (test) relative to exposures (elvitegravir, cobicistat, emtricitabine) from elvitegravir/cobicistat/emtricitabine/tenofovir (reference) or GS-7340 (TFV) (reference).
  • GS-7340 GS-7340
  • a second cohort with a similar design will evaluate an alternate formulation of elvitegravir/cobicistat/emtricitabine/GS-7340 STR.
  • Elvitegravir/cobicistat/emtricitabine/GS-7340 (monolayer) tablets were manufactured by blending of emtricitabine/GS-7340 granulation with elvitegravir granulation and cobicistat, tablet compression, tablet film-coating, and packaging.
  • Elvitegravir/cobicistat/emtricitabine/GS-7340 bilayer tablets are manufactured by compression of the elvitegravir/cobicistat layer and emtricitabine/GS-7340 layer, tablet film-coating, and packaging.
  • a balanced Williams 4 x 4 design is being used in each cohort.
  • elvitegravir/cobicistat/emtricitabine/GS-7340 represent current investigational doses (elvitegravir, cobicistat) or marketed dose (emtricitabine) with demonstrated durable efficacy and long-term safety in HIV-infected patients.
  • Treatment A 1 x Single Tablet Regimen (STR) of Formulation 1
  • Treatment B 1 x STR Formulation 1 (150 mg elvitegravir plus 150 mg cobicistat plus 200 mg emtricitabine plus 40 mg GS-7340 (as 49.7 mg of the fumarate salt GS-7340-02)) QD, administered in A.M. for 12 days.
  • Treatment C 1 x STR (150 mg elvitegravir plus 150 mg cobicistat plus 200 mg emtricitabine plus 300 mg tenofovir (as tenofovir disoproxil fumarate) QD, administered in A.M. for 12 days.
  • Treatment D 1 x 25 mg GS-7340 tablet QD, administered in A.M. for 12 days.
  • Formulation 1 (mono-layer) was prepared by blending of
  • EVG/COBI/FTC/GS-7340 STR tablet cores contain colloidal silicon dioxide, croscarmellose sodium, hydroxypropyl cellulose, lactose monohydrate, microcrystalline cellulose, sodium lauryl sulfate and magnesium stearate as inactive ingredients and are film-coated with polyvinyl alcohol, polyethylene glycol, talc, and titanium dioxide.
  • Treatment E 1 x STR Formulation 2 (150 mg elvitegravir plus 150 mg cobicistat plus 200 mg emtricitabine plus 25 mg GS-7340 (as 31.1 mg of the fumarate salt GS-7340-02)) QD, administered in A.M. for 12 days.
  • Treatment F 1 x STR Formulation 2 (150 mg elvitegravir plus 150 mg cobicistat plus 200 mg emtricitabine plus 40 mg GS-7340 (as 49.7 mg of the fumarate salt GS-7340-02)) QD, administered in A.M. for 12 days.
  • Treatment C 1 x STR (150 mg elvitegravir plus 150 mg cobicistat plus
  • Treatment D 1 x 25 mg GS-7340 tablet QD, administered in A.M. for 12 days.
  • Formulation 2 was prepared as bilayer tablets that were manufactured by compression of the elvitegravir/cobicistat layer and emtricitabine/GS-7340 layer, tablet film-coating, and packaging.
  • the EVG/COBI/FTC/GS-7340 STR tablet cores contain colloidal silicon dioxide, croscarmellose sodium, hydroxypropyl cellulose, lactose monohydrate, microcrystalline cellulose, sodium lauryl sulfate and magnesium stearate as inactive ingredients and are film-coated with polyvinyl alcohol, polyethylene glycol, talc, and titanium dioxide.
  • Figure 6 shows pharmacokinetic data for GS-7340 from patients treated in Cohort 1 (Formulation 1, mono-layer).
  • the top line (upside down triangles) shows GS-7340 concentration (ng/ml) when 40 mg GS-7340 is administered with cobicistat.
  • the middle line (right side up triangles) shows GS-7340 concentration (ng/ml) when 25 mg GS-7340 is administered with cobicistat.
  • the bottom line (squares) shows GS-7340 concentration (ng/ml) when 25 mg GS-7340 is administered alone.
  • Figure 7 shows pharmacokinetic data for GS-7340 from patients treated in Cohort 2 (Formulation 2, bi-layer).
  • the top line (upside down triangles) shows GS-7340 concentration (ng/ml) when 40 mg GS-7340 is administered with cobicistat.
  • the middle line (right side up triangles) shows GS-7340 concentration (ng/ml) when 25 mg GS-7340 is administered with cobicistat.
  • the bottom line (squares) shows GS-7340 concentration (ng/ml) when 25 mg GS-7340 is administered alone.
  • Figure 8 shows pharmacokinetic data for tenofovir from patients treated in Cohort 1 (Formulation I, mono-layer).
  • the top line shows tenofovir concentration (ng/ml) when 300 mg tenofovir is administered with cobicistat.
  • the next line down shows tenofovir concentration (ng/ml) when 40 mg GS-7340 is administered with cobicistat.
  • the next line down shows tenofovir concentration (ng/ml) when 25 mg GS-7340 is administered with cobicistat.
  • the bottom line upside down triangles) shows tenofovir concentration (ng/ml) when 25 mg GS-7340 is administered alone.
  • Figure 9 shows pharmacokinetic data for tenofovir from patients treated in Cohort 2 (Formulation 2, bi-layer).
  • the top line shows tenofovir concentration (ng/ml) when 300 mg tenofovir is administered with cobicistat.
  • the next line down shows tenofovir concentration (ng/ml) when 40 mg GS-7340 is administered with cobicistat.
  • the next line down shows tenofovir concentration (ng/ml) when 25 mg GS-7340 is administered with cobicistat.
  • the bottom line upside down triangles) shows tenofovir concentration (ng/ml) when 25 mg GS-7340 is administered alone.
  • GS-7340 was coformulated with elvitegravir (EVG), cobicistat (COBI), and emtricitabine (FTC) into a single tablet regimen (STR).
  • EDG elvitegravir
  • COBI cobicistat
  • FTC emtricitabine
  • EVG/COBI/FTC/GS-7340 STR and/or interaction potential between GS-7340 and COBI were evaluated to facilitate GS-7340 dose selection for STR clinical development.
  • EVG/COBI/FTC/GS-7340 STR provided similar EVG, COBI, and FTC exposures versus reference treatments and historical data.
  • EVG/COBI/FTC/GS-7340 coadministration with COBI or as EVG/COBI/FTC/GS-7340 dosing, which may be due to COBI inhibition of intestinal Pgp-mediated intestinal secretion of GS-7340.
  • EVG/COBI/FTC/GS-7340 provided comparable GS-7340 and TFV exposures as GS-7340 at 25 mg and -90% lower TFV exposure versus EVG/COBI/FTC/TDF.

Abstract

La présente invention concerne l'utilisation de la {9-[(R)-2-[[(S)-[[(S)-1-(isopropoxycarbonyl)éthyl]amino]phénoxyphosphinyl]-méthoxy]propyl]adénine} et de la forme hémifumarate (hémifumarate du ténofovir alafénamide) en association avec le cobicistat. De plus, l'invention concerne l'association de l'hémifumarate du ténofovir alafénamide, du cobicistat, de l'emtricitabine, et de l'elvitegravir.
PCT/US2012/069853 2012-02-03 2012-12-14 Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d'infections virales WO2013115916A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2012327170A AU2012327170A1 (en) 2012-02-03 2012-12-14 Therapeutic compounds

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201261594894P 2012-02-03 2012-02-03
US61/594,894 2012-02-03
US201261618411P 2012-03-30 2012-03-30
US61/618,411 2012-03-30
US201261624676P 2012-04-16 2012-04-16
US61/624,676 2012-04-16
US201261692392P 2012-08-23 2012-08-23
US61/692,392 2012-08-23

Publications (1)

Publication Number Publication Date
WO2013115916A1 true WO2013115916A1 (fr) 2013-08-08

Family

ID=47436276

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/069853 WO2013115916A1 (fr) 2012-02-03 2012-12-14 Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d'infections virales

Country Status (2)

Country Link
AU (1) AU2012327170A1 (fr)
WO (1) WO2013115916A1 (fr)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015120057A1 (fr) * 2014-02-05 2015-08-13 Gilead Sciences, Inc. Combinaisons pharmaceutiques contre une co-infection par le vih et la tuberculose
WO2015161781A1 (fr) * 2014-04-21 2015-10-29 四川海思科制药有限公司 Procédé pour la préparation d'un analogue de nucléoside et intermédiaire correspondant
WO2015107451A3 (fr) * 2014-01-14 2015-12-10 Mylan Laboratories Ltd. Purification de ténofovir alafénamide et de ses intermédiaires
CN105237571A (zh) * 2014-11-28 2016-01-13 成都苑东药业有限公司 9-[(r)-2-[[(s)-[[(s)-1-(异丙氧基羰基)乙基]氨基]苯氧基氧膦基]甲氧基]丙基]腺嘌呤的盐
CN105330700A (zh) * 2015-12-17 2016-02-17 中国药科大学 富马酸替诺福韦艾拉酚胺杂质的制备方法
CN105518012A (zh) * 2014-06-25 2016-04-20 四川海思科制药有限公司 一种取代的氨基酸硫酯类化合物、其组合物及应用
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
WO2018153977A1 (fr) * 2017-02-24 2018-08-30 Hexal Ag Composition stable de ténofovir alafénamide
WO2019018676A1 (fr) * 2017-07-20 2019-01-24 Janssen Sciences Ireland Unlimited Company Compositions comprenant du darunavir, du cobicistat, de l'emtricitabine et du ténofovir alafénamide pour une utilisation dans le traitement du vih
WO2019113462A1 (fr) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine et dérivés et leurs utilisations anti-virales
EP3346995B1 (fr) 2015-11-09 2019-08-28 Gilead Sciences, Inc. Compositions thérapeutiques pour le traitement du virus de l'immunodéficience humaine
CN110305163A (zh) * 2018-03-27 2019-10-08 北京济美堂医药研究有限公司 替诺福韦艾拉酚胺半富马酸盐的制备方法
CN111686082A (zh) * 2019-03-11 2020-09-22 苏州特瑞药业有限公司 一种富马酸磷丙替诺福韦制剂及其制备方法
CN113072583A (zh) * 2016-11-28 2021-07-06 正大天晴药业集团股份有限公司 一种替诺福韦艾拉酚胺半富马酸盐的结晶及其制备方法
CN113777186A (zh) * 2021-08-12 2021-12-10 北京鑫开元医药科技有限公司 一种富马酸丙酚替诺福韦中杂质的检测方法
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11654150B2 (en) 2011-07-07 2023-05-23 Janssen Sciences Ireland Uc Darunavir combination formulations
US11981699B2 (en) 2021-08-02 2024-05-14 Emory University Phosphoramidates for the treatment of hepatitis B virus

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6043230A (en) 1996-07-26 2000-03-28 Gilead Sciences, Inc. Antiviral phosphonomethoxy nucleotide analogs having increased oral bioavailability
WO2003037379A1 (fr) 2001-10-30 2003-05-08 Degussa Ag Utlisation de materiaux granuleux a base de dioxyde de silicium produit de façon pyrogenique dans des compositions pharmaceutiques
WO2004064846A1 (fr) * 2003-01-14 2004-08-05 Gilead Sciences, Inc. Compositions et methodes de polytherapie antivirale
US7390791B2 (en) 2000-07-21 2008-06-24 Gilead Sciences, Inc. Prodrugs of phosphonate nucleotide analogues
WO2008103949A1 (fr) * 2007-02-23 2008-08-28 Gilead Sciences, Inc. Modulateurs de propriétés pharmacocinétiques de produits thérapeutiques
WO2010091197A2 (fr) * 2009-02-06 2010-08-12 Gilead Sciences, Inc. Pastilles destinées à une thérapie combinée

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6043230A (en) 1996-07-26 2000-03-28 Gilead Sciences, Inc. Antiviral phosphonomethoxy nucleotide analogs having increased oral bioavailability
US7390791B2 (en) 2000-07-21 2008-06-24 Gilead Sciences, Inc. Prodrugs of phosphonate nucleotide analogues
US7803788B2 (en) 2000-07-21 2010-09-28 Gilead Sciences, Inc. Prodrugs of phosphonate nucoleotide analogues
WO2003037379A1 (fr) 2001-10-30 2003-05-08 Degussa Ag Utlisation de materiaux granuleux a base de dioxyde de silicium produit de façon pyrogenique dans des compositions pharmaceutiques
WO2004064846A1 (fr) * 2003-01-14 2004-08-05 Gilead Sciences, Inc. Compositions et methodes de polytherapie antivirale
WO2008103949A1 (fr) * 2007-02-23 2008-08-28 Gilead Sciences, Inc. Modulateurs de propriétés pharmacocinétiques de produits thérapeutiques
WO2010091197A2 (fr) * 2009-02-06 2010-08-12 Gilead Sciences, Inc. Pastilles destinées à une thérapie combinée

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
"Gilead and Tibotec finalize agreement on single-tablet HIV regimen", THOMSON REUTERS DRUG NEWS, 17 November 2011 (2011-11-17), XP055052007, Retrieved from the Internet <URL:http://integrity.thomson-pharma.com/integrity/xmlxsl/pk_ref_list.xml_related_ref_to?p_id=315936&p_origen=PRO&p_tsearch=#link> [retrieved on 20130201] *
"Phase II trial investigates GS-7340 as part of a new treatment regimen", THOMSON REUTERS DRUG NEWS, 26 January 2012 (2012-01-26), XP055051645, Retrieved from the Internet <URL:http://integrity.thomson-pharma.com/integrity/xmlxsl/pk_ref_list.xml_related_ref_to?p_id=315936&p_origen=PRO&p_tsearch=#link> [retrieved on 20130129] *
CHAPMAN H ET AL: "Practical synthesis, separation, and stereochemical assignment of the PMPA pro-drug GS-7340", NUCLEOSIDES, NUCLEOTIDES AND NUCLEIC ACIDS, vol. 20, no. 4-7, 1 January 2001 (2001-01-01), TAYLOR & FRANCIS, PHILADELPHIA, PA, USA, pages 621 - 628, XP002202043, ISSN: 1525-7770, DOI: 10.1081/NCN-100002338 *
GENNARO ET AL.: "Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
LEPIST EI ET AL: "Cobicistat Boosts the Intestinal Absorption of Transport Substrates, Including HIV Protease Inhibitors and GS-7340, In Vitro", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, WASHINGTON, DC, US, vol. 56, no. 10, 1 October 2012 (2012-10-01), pages 5409 - 5413, XP009166632, ISSN: 0066-4804 *
LIZ HIGHLEYMAN: "ICAAC 2012: New Tenofovir Pro-drug Matches Potency of Original Version but with Less Toxicity", HIVANDHEPATITIS.COM, 6 December 2012 (2012-12-06), XP055052016, Retrieved from the Internet <URL:http://www.hivandhepatitis.com/hiv-aids/hiv-aids-topics/hiv-treatment/3853-new-tenofovir-pro-drug-matches-potency-of-original-version-but-with-less-toxicity> [retrieved on 20130201] *
MARK MASCOLINI: "Cobicistat Doubles to Triples Exposure of GS-7340 (a Tenofovir Prodrug) and Resulting Tenofovir", 16 April 2012 (2012-04-16) - 18 April 2012 (2012-04-18), 13th International Workshop on Clinical Pharmacology of HIV Therapy, Barcelona, Spain, XP002691442, Retrieved from the Internet <URL:http://www.natap.org/2012/pharm/Pharm_13.htm> [retrieved on 20130201] *
RICHARD ELION ET AL: "Phase 2 study of cobicistat versus ritonavir each with once-daily atazanavir and fixed-dose emtricitabine/tenofovir df in the initial treatment of HIV infection", AIDS, vol. 25, no. 15, 1 September 2011 (2011-09-01), pages 1881 - 1886, XP055051636, ISSN: 0269-9370, DOI: 10.1097/QAD.0b013e32834b4d48 *
TONG ET AL., ANTIMICROB AGENTS CHEMOTHER, vol. 51, 2007, pages 3498 - 504

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11654150B2 (en) 2011-07-07 2023-05-23 Janssen Sciences Ireland Uc Darunavir combination formulations
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
WO2015107451A3 (fr) * 2014-01-14 2015-12-10 Mylan Laboratories Ltd. Purification de ténofovir alafénamide et de ses intermédiaires
WO2015120057A1 (fr) * 2014-02-05 2015-08-13 Gilead Sciences, Inc. Combinaisons pharmaceutiques contre une co-infection par le vih et la tuberculose
US9463194B2 (en) 2014-02-05 2016-10-11 Gilead Sciences, Inc. Methods of treating patients co-infected with HIV and tuberculosis
WO2015161781A1 (fr) * 2014-04-21 2015-10-29 四川海思科制药有限公司 Procédé pour la préparation d'un analogue de nucléoside et intermédiaire correspondant
CN105518012A (zh) * 2014-06-25 2016-04-20 四川海思科制药有限公司 一种取代的氨基酸硫酯类化合物、其组合物及应用
CN105237571A (zh) * 2014-11-28 2016-01-13 成都苑东药业有限公司 9-[(r)-2-[[(s)-[[(s)-1-(异丙氧基羰基)乙基]氨基]苯氧基氧膦基]甲氧基]丙基]腺嘌呤的盐
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11744802B2 (en) 2015-11-09 2023-09-05 Gilead Sciences, Inc. Therapeutic compositions for treatment of human immunodeficiency virus
EP3346995B1 (fr) 2015-11-09 2019-08-28 Gilead Sciences, Inc. Compositions thérapeutiques pour le traitement du virus de l'immunodéficience humaine
CN105330700A (zh) * 2015-12-17 2016-02-17 中国药科大学 富马酸替诺福韦艾拉酚胺杂质的制备方法
CN113072583A (zh) * 2016-11-28 2021-07-06 正大天晴药业集团股份有限公司 一种替诺福韦艾拉酚胺半富马酸盐的结晶及其制备方法
WO2018153977A1 (fr) * 2017-02-24 2018-08-30 Hexal Ag Composition stable de ténofovir alafénamide
US10786518B2 (en) 2017-07-20 2020-09-29 Janssen Sciences Ireland Uc Compositions and methods of treating HIV
WO2019018676A1 (fr) * 2017-07-20 2019-01-24 Janssen Sciences Ireland Unlimited Company Compositions comprenant du darunavir, du cobicistat, de l'emtricitabine et du ténofovir alafénamide pour une utilisation dans le traitement du vih
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2019113462A1 (fr) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine et dérivés et leurs utilisations anti-virales
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
CN110305163A (zh) * 2018-03-27 2019-10-08 北京济美堂医药研究有限公司 替诺福韦艾拉酚胺半富马酸盐的制备方法
CN111686082A (zh) * 2019-03-11 2020-09-22 苏州特瑞药业有限公司 一种富马酸磷丙替诺福韦制剂及其制备方法
US11981699B2 (en) 2021-08-02 2024-05-14 Emory University Phosphoramidates for the treatment of hepatitis B virus
CN113777186A (zh) * 2021-08-12 2021-12-10 北京鑫开元医药科技有限公司 一种富马酸丙酚替诺福韦中杂质的检测方法
CN113777186B (zh) * 2021-08-12 2023-06-13 北京鑫开元医药科技有限公司 一种富马酸丙酚替诺福韦中杂质的检测方法

Also Published As

Publication number Publication date
AU2012327170A1 (en) 2013-08-22

Similar Documents

Publication Publication Date Title
AU2013204731C1 (en) Therapeutic compounds
WO2013115916A1 (fr) Polythérapie comprenant le gs-7340 et le cobicistat destinée à être utilisée dans le traitement d&#39;infections virales
ES2602784T3 (es) Moduladores de las propiedades farmacocinéticas de los agentes terapéuticos
TWI411613B (zh) 用於改善被細胞色素p450單氧合酶所代謝之治療劑的藥物動力性質之調節劑
JP5629585B2 (ja) シトクロムp450のインヒビター
AU2012250890B2 (en) Amorphous solid salts
US20150004239A1 (en) Pharmaceutical compositions and methods for their preparation
US11338007B2 (en) Combination formulation of three antiviral compounds
NO20200578A1 (no) Modulatorer av farmakokinetikkegenskaper til terapeutika
AU2019216697A1 (en) Modulators of pharmacokinetic properties of therapeutics

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2012327170

Country of ref document: AU

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

Ref document number: 12806843

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: 12806843

Country of ref document: EP

Kind code of ref document: A1