WO2008023273A2 - Method for improving pharmacokinetics - Google Patents

Method for improving pharmacokinetics Download PDF

Info

Publication number
WO2008023273A2
WO2008023273A2 PCT/IB2007/003593 IB2007003593W WO2008023273A2 WO 2008023273 A2 WO2008023273 A2 WO 2008023273A2 IB 2007003593 W IB2007003593 W IB 2007003593W WO 2008023273 A2 WO2008023273 A2 WO 2008023273A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
carbon atoms
formula
pyridyl
och
Prior art date
Application number
PCT/IB2007/003593
Other languages
French (fr)
Other versions
WO2008023273A3 (en
WO2008023273A8 (en
Inventor
Jinzi J. Wu
Michael Ge
Chandra J. Panchal
Original Assignee
Ambrilia Biopharma 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 Ambrilia Biopharma Inc. filed Critical Ambrilia Biopharma Inc.
Priority to JP2009520074A priority Critical patent/JP2010502569A/en
Priority to AU2007287353A priority patent/AU2007287353A1/en
Priority to CA002657316A priority patent/CA2657316A1/en
Priority to EP07825714A priority patent/EP2109461A2/en
Publication of WO2008023273A2 publication Critical patent/WO2008023273A2/en
Publication of WO2008023273A8 publication Critical patent/WO2008023273A8/en
Publication of WO2008023273A3 publication Critical patent/WO2008023273A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to methods for improving the pharmacokinetics of drugs which are metabolized by cytochrome P450 monooxygenase using PPL-100 and related pharmaceutical compositions.
  • the present invention relates to methods for improving the pharmacokinetics of HIV protease inhibitors by co-administering PPL-100 with such protease inhibitors.
  • proteases inhibitors of the HIV viral protease have been developed relatively recently and their use began only in 1996. Currently, they are considered the most effective drugs against HIV infection. Unfortunately, most current proteases inhibitors are relatively large hydrophobic molecules that possess rather low bioavailability. A high pill burden is therefore required to attain the therapeutic dose in a patient. This is a deterrent, which too often results in patient non-compliance and inadequate treatment results. This situation leads to sub- optimal therapeutic drug concentration that in turns leads to the development of HIV resistant strains. Consequently, there is an urgent need to improve the solubility and bioavailability of proteases inhibitors.
  • Lysine-based compounds with increased solubility and improved oral bioavailability are described herein and have been described in United States patent application No. 10/902,935 filed on August 2, 2004 and published on February 2, 2006 under No. 2006/0025592A1 , the entire content of which is incorporated herein by reference. These compounds may readily be cleaved in vivo to release an active ingredient which has an affinity for aspartyl proteases and which may act as a protease inhibitor. More particularly, the active ingredient may bind, for example, to an HIV aspartyl protease (U.S. patent no. 6,632,816) and may inhibit this enzyme.
  • the Lysine-based compounds are also referred herein as a protease inhibitor precursor.
  • the Lysine-based compounds Upon in vivo physiological conditions (e.g., metabolic, enteric and/or gastrointestinal conditions, etc.) the Lysine-based compounds, allow for the release of a protease inhibitor (e.g., aspartyl protease inhibitor).
  • a protease inhibitor e.g., aspartyl protease inhibitor
  • the Lysine-based compounds may thus serve as means for improving the solubility and/or bioavailability of protease inhibitors and therefore may reduce the pill burden and/or reduce dosages needed for inhibition. Improved treatment of HIV-infected patients and favourable patient's compliance may consequently occur.
  • the compounds described herein may be used to inhibit the activity of cytochrome P450 monooxygenase (CYP450). More particularly, these compounds may be used to inhibit the activity of CYP3A4, including for example, CYP3A4/5.
  • CYP450 cytochrome P450 monooxygenase
  • the compounds described herein may thus be used with drugs which are metabolized by cytochrome P450 monooxygenase in order to improve their pharmacokinetics. More particularly, these compounds may be used to improve the pharmacokinetics of drugs which are metabolized by CYP3A4, including CYP3A4/5.
  • drugs which may benefit from inhibition of CYP450 may include, for example, the immunosupressants cyclosporine, FK-506 and rapamycin, the chemotherapeutic agents taxol and taxotere, the antibiotic clarithromycin and the HIV protease inhibitors, A-77003, A-80987, MK-639.
  • Exemplary embodiments of compounds encompassed by the present invention and which may be used for inhibiting CYP450 may include, for example, a compound of formula I:
  • pharmaceutically acceptable salts and derivatives thereof e.g., for example, when the compound of the present invention comprises an amino group, the pharmaceutically acceptable salt may be an ammonium salt,
  • n may be, for example, 3 or 4,
  • X and Y may be selected, for example, from the group consisting of H 1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -OCF 3 , - CN, -NO 2 , -NR 4 R 5 , -NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 , and -CH 2 OH or X and Y together define an alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH 2 O- and an ethylenedioxy group of formula - OCH 2 CH 2 O-,
  • R 6 may be selected, for example, from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
  • R 3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R 3A -CO-, wherein R 3A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms (e.g.
  • a cycloalkyl group having 3 to 6 carbon atoms e.g. cyclopropyl-, cyclohexyl- etc.
  • a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof e.g.
  • a picolyl group selected from the group consisting of
  • a picolyloxy group selected from the group consisting of
  • a substituted pyridyl group selected from the group consisting of
  • X' and Y' may be selected, for example, from the group consisting of H 1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -NO 2 , - NR 4 R 5 , -NHCOR 4 . -OR 4 , -SR 4 , -COOR 4 , -COR 4 and -CH 2 OH,
  • R 4 and R 5 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms, wherein R 2 may be selected, for example, from the group consisting of a diphenylmethyl group of formula IV a naphthyl-1 -CH 2 - group of formula V
  • R 1 may be H or a physiologically cleavable unit, whereby upon ⁇ in vivo) physiological conditions the compound may be converted into an active protease inhibitor.
  • the compound upon cleavage of the physiologically cleavable unit, the compound may be able to release a protease inhibitor.
  • Ri may be selected, for example, from the group consisting of (HO) 2 P(O) and (MO) 2 P(O), wherein M is an alkali metal (e.g. Na, K, Cs 1 etc) or alkaline earth metal (Ca, Mg, etc.).
  • M is an alkali metal (e.g. Na, K, Cs 1 etc) or alkaline earth metal (Ca, Mg, etc.).
  • R 1 may be a group of formula R 1A -CO-, wherein R 1A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms (e.g. methyl, ethyl, propyl, /so-propyl, butyl, /so-butyl, terf-butyl, tert-butyl-CH 2 -, etc.), a cycloalkyl group having 3 to 6 carbon atoms (e.g.
  • cyclopropyl-, cyclohexyl- etc. a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, (e.g. cyclopropyl-CH 2 -, cyclohexyl-CH 2 -, etc.), an alkyloxy group of 1 to 6 carbon atoms (e.g.
  • HI a picolyl group selected from the group consisting of
  • a picolyloxy group selected from the group consisting of
  • a substituted pyridyl group selected from the group consisting of
  • a pharmaceutical composition which may comprise: a) a compound of formula I as described herein or a pharmaceutically acceptable salt thereof, b) a drug which may be metabolized by a cytochrome P450 monooxigenase (e.g., CYP3A4), and; c) a pharmaceutically acceptable carrier; where the compound of formula I is represented by;
  • n may be 3 or 4,
  • X and Y may be selected, for example, from the group consisting of H 1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -OCF 3 , - CN, -NO 2 , -NR 4 R 5 .
  • -NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 , and -CH 2 OH or X and Y together may define an alkylenedioxy group which may be selected, for example, from the group consisting of a methylenedioxy group of formula -OCH 2 O- and an ethylenedioxy group of formula -OCH 2 CH 2 O-,
  • R 8 may be selected from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
  • R 3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R 3A -CO-, where R 3A may be selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, -CH 2 OH 1 -CF 3 , -CH 2 CF 3 , -CH 2 CH 2 CF 3 .
  • i a picolyloxy group which may be selected from the group consisting of
  • a substituted pyridyl group which may be selected from the group consisting of
  • X 1 and Y may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -NO 2 , - NR 4 R 5 , -NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 and -CH 2 OH 1
  • R 4 and R 5 may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
  • R 2 may be selected from the group consisting of a diphenylmethyl group of formula IV
  • R 1 may be H or a physiologically cleavable unit, whereby upon (in vivo) physiological conditions the compound may be converted into an active protease inhibitor. For example, upon cleavage of the physiologically cleavable unit, the compound may be able to release a protease inhibitor.
  • Ri may be selected, for example, from the group consisting of H 1 (HO) 2 P(O) and (MO) 2 P(O) (wherein M may be, for example, an alkali metal or alkaline earth metal) and a group of formula Ri A -CO-, where Ri A rnay be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH 2 OH, CH 3 O 2 C-, CH 3 O 2 CCH 2 -, Acetyl-OCH 2 CH 2 -, HO 2 CCH 2 -, 2-hydroxyphenyl, 3-hydroxyphenyl, 4- hydroxy
  • a picolyl group selected from the group consisting of
  • a picolyloxy group selected from the group consisting of
  • a substituted pyridyl group selected from the group consisting of
  • a pharmaceutical composition which may comprise; a) a compound of formula Il and pharmaceutically acceptable salts thereof, b) a drug which may be metabolized by a cytochrome P450 monooxigenase (e.g., CYP3A4), and; c) a pharmaceutically acceptable carrier; where the compound of formula Il is represented by;
  • n may be 3 or 4,
  • X and Y may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F 1 Cl, Br, I, -CF 3 , -OCF 3 , -CN, -NO 2 , -NR 4 Rs, - NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 , and -CH 2 OH or X and Y together may define, for example, an alkylenedioxy group which may be selected from the group consisting of a methylenedioxy group of formula -OCH 2 O- and an ethylenedioxy group of formula - OCH 2 CH 2 O-,
  • R 6 may be selected, for example, from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
  • R 3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R 3 A-CO-, where R 3 A rnay be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, -
  • a picolyl group which may be selected from the group consisting of
  • a picolyloxy group which may be selected from the group consisting of
  • a substituted pyridyl group which may be selected from the group consisting of
  • X' and Y' may be selected from the group consisting of H, a straight a Iky I group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F 1 Cl, Br, I, -CF 3 , -NO 2 , -NR 4 Rs, - NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 and -CH 2 OH,
  • R 4 and R 5 may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
  • R 2 may be selected from the group consisting of a diphenylmethyl group of formula IV a naphthyl-1-CH 2 - group of formula V
  • R 1 may be H or a physiologically cleavable unit, whereby upon physiological conditions (in vivo) the compound may be converted into an active protease inhibitor.
  • R 1 may be selected, for example, from the group consisting of H, (HO) 2 P(O) and (MO) 2 P(O) (wherein M may be an alkali metal or alkaline earth metal), and a group of formula R 1A -CO-, where R 1A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, -CH 2 OH, CH 3 O 2 C-, CH 3 O 2 CCH 2 -, Acetyl- OCH 2 CH 2 -, HO 2 CCH 2 -, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxy phenyl,
  • a picolyl group which may be selected from the group consisting of
  • a picolyloxy group which may be selected from the group consisting of
  • a substituted pyridyl group which may be selected from the group consisting of
  • compositions which comprise compounds of formula Il wherein R 6 may be, for example, /so-butyl and n may be 3 are encompassed herewith.
  • pharmaceutical compositions which comprise compounds of formula Il wherein R 8 may be, for example, /so-butyl and n may be 4 are also encompassed herewith.
  • Ri may be selected, for example, from the group consisting of H, (HO) 2 P(O) and (NaO) 2 P(O).
  • Ri may be selected, for example, from the group consisting of CH 3 CO, 3-pyridyl-CO, (CH 3 ) 2 NCH 2 CO and
  • compositions which comprise compounds of formula Il wherein R 3 may be selected, for example, from the group consisting of CH 3 CO, CH 3 O-CO, (CH 3 ) 2 N-CO, 3- pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO are encompassed by the present invention.
  • X may be 4-NH 2 and Y may be H or F.
  • X' and Y' may both be H.
  • compositions which comprise compounds of formula Il wherein R 2 may be selected, for example, from the group consisting of a diphenylmethyl group of formula IV 1 a naphthyl-1- CH 2 - group of formula V, a naphthyl-2-CH 2 - group of formula Vl, a biphenylmethyl group of formula VII and an anthryl-9-CH 2 - group of formula VIII are encompassed herewith.
  • R 2 may, more particularly, be selected from the group consisting of a diphenylmethyl group of formula IV, a naphthyl-1-CH 2 - group of formula V, and a naphthyl- 2-CH 2 - group of formula Vl.
  • the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R 6 is /so-butyl, n is 4 and R 2 is a diphenylmethyl group of formula IV.
  • R 1 may be selected from the group consisting of H, (HO) 2 P(O) and (NaO) 2 P(O).
  • R 1 may be selected from the group consisting of CH 3 CO, 3-pyridyl-CO, (CH 3 J 2 NCH 2 CO and (CH 3 ) 2 CHCH(NH 2 )CO.
  • R 3 may be selected, for example, from the group consisting of CH 3 CO, CH 3 O-CO 1 (CH 3 ) 2 N-CO, 3- pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO.
  • X may be 4-NH 2 and Y may be H or F.
  • the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X may be, for example, 4-NH 2 , Y may be H, X' may be H, Y' may be H and R 3 may be CH 3 O-CO.
  • Ri may be (HO) 2 P(O).
  • R 1 may be (NaO) 2 P(O).
  • R 1 may be H.
  • the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X may be 4-NH 2 , Y may be 3-F, X' may be H, Y' may be H and R 3 may be CH 3 O- CO.
  • R 1 may be (HO) 2 P(O). In accordance with another particular embodiment of the present invention, R 1 may be (NaO) 2 P(O).
  • Ri may be H.
  • the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X is 4-NH 2 , Y is H or 3-F, X 1 is H, Y' is H and R 3 is CH 3 CO.
  • Ri may be (HO) 2 P(O).
  • Ri may be (NaO) 2 P(O).
  • R 1 may be H.
  • the present invention further provides a pharmaceutical composition which may comprise a compound of formula II, X is 4-NH 2 , Y is H or 3-F, X * is H 1 Y' is H and R 3 is 4-morphoHne-CO.
  • X may be 4-NH 2
  • Y may be H
  • X 1 may be H
  • Y * may be H
  • R 3 may be CH 3 O-CO.
  • R 1 may be 3-pyridyl- CO.
  • R 1 may be (CH 3 ) 2 NCH 2 CO.
  • R 1 may be (CHa) 2 CHCH(NH 2 )CO.
  • R 1 may be CH 3 CO.
  • the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R 8 is /sobutyl, n is 4, X' and Y" are both H, R 2 is Naphtyl-1-CH 2 -, X is 4-NH 2 , Y is H, R 3 is 4-morpholine-CO and R 1 may be selected, for example, from the group consisting of H, (HO) 2 P(O) and (NaO) 2 P(O).
  • the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R 6 is /so-butyl, n is 4, X 1 and Y' are both H, R 2 is Naphtyl-2-CH 2 -, X is 4-NH 2 , Y is H 1 R 3 is CH 3 O-CO and Ri may be selected, for example, from the group consisting of H, (HO) 2 P(O) and (NaO) 2 P(O).
  • the present invention provides a pharmaceutical composition which may comprise; a) a compound of formula Ha
  • X and Y may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -OCF 3 , - CN, -NO 2 , -NR 4 R 5 , -NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 .
  • alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH 2 O- and an ethylenedioxy group of formula - OCH 2 CH 2 O-,
  • X' and Y' may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF 3 , -NO 2 , - NR 4 R 5 , -NHCOR 4 , -OR 4 , -SR 4 , -COOR 4 , -COR 4 and -CH 2 OH 1 wherein n, R 1 , R 3 , R 4 , R 5 and R ⁇ are as defined herein;
  • R 1 may be selected from the group consisting of H, (HO) 2 P(O) and (MO) 2 P(O), wherein M may be an alkali metal or alkaline earth metal and a group of formula R 1A -CO-, R 1A which may be selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH 2 OH, CH 3 O 2 C-, CH 3 O 2 CCH 2 -, ACeIyI-OCH 2 CH 2 -, HO 2 CCH 2 -, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, (CH 3 ) 2 NCH
  • a picolyl group selected from the group consisting of
  • a picolyloxy group selected from the group consisting of
  • R 6 may be feo-b ⁇ tyl.
  • n may be 4.
  • R 1 may be selected from the group consisting of H, (HO) 2 P(O) and (NaO) 2 P(O).
  • Ri may be selected, for example, from the group consisting of CH 3 CO, 3-pyridyl-CO, (CH 3 J 2 NCH 2 CO and (CH 3 ) 2 CHCH(NH 2 )CO.
  • R 3 may be selected from the group consisting of CH 3 CO, CH 3 O-CO 1 (CH 3 ) 2 N-CO, 3-pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO.
  • R 3 may be selected from the group consisting of CH 3 CO, CH 3 O-CO, (CH 3 ) 2 N-CO, 3-pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO.
  • X may be 4-NH 2 and Y may be H or F.
  • X may be 4-NH 2 and Y may be H or F.
  • X may be 4-NH 2
  • Y may be H or 3-F
  • X 1 may be H
  • Y' may be H
  • R 3 may be CH 3 CO.
  • X may be 4-NH 2
  • Y may be H or 3-F
  • X' may be H
  • Y' may be H
  • R 3 may be 4-morpholine-CO.
  • X may be 4-NH 2
  • Y may be H 1
  • X' may be H
  • Y" may be H
  • R 3 may be CH 3 O-CO and R 1 may be (HO) 2 P(O), (NaO) 2 P(O) or H.
  • X may be 4- NH 2
  • Y may be 3-F
  • X' may be H
  • Y' may be H 1
  • R 3 may be CH 3 O-CO
  • Ri may be (HO) 2 P(O). (NaO) 2 P(O) or H.
  • X may be 4- NH 2
  • Y may be H or 3-F
  • X * may be H
  • Y' may be H 1
  • R 3 may be CH 3 CO
  • Ri may be (HO) 2 P(O) 1 (NaO) 2 P(O) or H.
  • X may be 4- NH 2
  • Y may be H or 3-F
  • X 1 may be H
  • Y' may be H
  • R 3 may be 4-morpholine-CO.
  • X may be 4-NH 2 , Y may be H 1 X" may be H, Y' may be H 1 R 3 may be CH 3 O-CO and R 1 may be 3-pyridyl-CO, (CH 3 ) 2 NCH 2 CO, (CH 3 ) 2 CHCH(NH 2 )CO or CH 3 CO.
  • X may be 4-NH 2 , Y may be 3-F, X' may be H, Y' may be H, R 3 may be CH 3 O-CO and Ri may be 3-pyridyl-CO, (CHg) 2 NCH 2 CO or (CH 3 ) 2 CHCH(NH 2 )CO.
  • R 1 may be, for example, H 1 (HO) 2 P(O) or (NaO) 2 P(O). Further in accordance with the present invention, n may be 4. Y may be, for example, H. R 3 may be, for example. CH 3 O-CO. R 2 may be, for example, a diphenylmethyl group of formula IV, where X' and Y 1 may be, for example H 1
  • compositions, methods, uses and kits encompassed by the present invention may comprise one or more of the following compounds and combination thereof; I O
  • n 4, R 1 is (CHa) 2 NCH 2 CO 1 X is 4-NH 2 , Y is H, X' is 30 H 1 Y * is H 1 R 6 is /so-butyl and R 3 is CH 3 O-CO 1 - a compound of formula Ha wherein n is 4, R 1 is (CHa) 2 CHCH(NH 2 )CO, X is 4-NH 2 , Y is H, X' is H, Y" is H, R 6 is /so-butyl and R 3 is CH 3 O-CO,
  • Any compound which is a precursor of an active ingredient described herein may be used to carry out the present invention and is also encompassed by the present invention.
  • the above identified active ingredient is identified herein as PL-100 and it is to be understood herein that any precursor able to release or generate the above mentioned exemplary compound either in vivo or in vitro is encompassed by the present invention and may be used to carry out methods, pharmaceutical compositions, kits and uses described herein.
  • the above identified active ingredient is identified herein as PL-337 and it is to be understood herein that any precursor able to release or generate the above mentioned exemplary compound either in vivo or in vitro may be used to carry out the present invention and is encompassed by the present invention.
  • the present invention thus relates to the use of a compound described herein for improving the pharmacokinetics of a drug which may affected by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.).
  • CYP3A4 e.g., CYP3A4/5 etc.
  • the present invention also relates to methods of treatment which may comprise admininistering to an individual in need, a compound described herein with one or more drugs which may be affected (metabolized) by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.). As indicated herein, administration may be done at the same time or at different time intervals.
  • the compound(s) and drug(s) may be mixed together or not.
  • the present invention further relates to the use of a compound described herein in the manufacture of a medicament for improving the pharmacokinetics of a drug which may affected by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.).
  • CYP3A4 e.g., CYP3A4/5 etc.
  • the present invention further relates to the use of a compound described herein and a drug which may affected by CYP450 in the manufacture of a medicament for treating a patient in need.
  • pharmaceutically effective amount refers to an amount effective in treating, preventing or reducing the risk or probability of HlV infection or of reducing HIV burden.
  • pharmaceutically effective amount also refers to an amount effective in treating, preventing or reducing the risk or probability of developing acquired immunodeficiency syndrome (AIDS), for delaying the apparition of AIDS, or reducing AIDS symptoms.
  • AIDS acquired immunodeficiency syndrome
  • a "pharmaceutically effective amount” may be construed as an amount giving a desired therapeutic effect, either taken into a single or multiple doses or in any dosage or route or taken alone or in combination with other therapeutic agents.
  • a "pharmaceutically effective amount” may be understood as an amount having an inhibitory effect (partial or complete) on HIV (HIV-1 and HIV-2 as well as related viruses (e.g., HTLV-I and HTLV-II, and simian immunodeficiency virus (SIV))) infection cycle (e.g., inhibition of replication, reinfection, maturation, budding etc.) and on any organism which rely on aspartyl proteases for its life cycle.
  • An inhibitory effect is to be understood herein as an effect such as a reduction in the capacity of an organism (e.g. HIV) to reproduce itself (replicate), to re-infect surrounding cells, etc, or even a complete inhibition (or elimination) of an organism.
  • HIV protease and “HIV aspartyl protease” are used interchangeably and includes, for example, the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2.
  • pharmaceutically acceptable carrier refers to a non-toxic carrier or adjuvant that may be administered to a patient, together with one or more compounds of the present invention, and which does not destroy the pharmacological activity thereof.
  • the term "precursor” refers to a compound, such as a Lysine-based compound which is able to be converted into an active ingredient in vitro or in vivo.
  • the compound PL-461 is a precursor of compound PL-100 as when administered to an individual, PL-461 is converted into PL-100 in vivo (e.g., under physiological conditions).
  • derivative refers to a compound which has been chemically synthesized from an original compound.
  • PL-461 is a derivative of PL-100.
  • composition includes the specified materials and may include other material that does not materially affect the basic characteristics of the pharmaceutical composition.
  • compositions of formula I such as compounds of formulae I, II, Ha, lib, Uc, MA and HA 1
  • pharmaceutically acceptable salts thereof such as, for example, ammonium salts are described herein.
  • a "pharmaceutically acceptable derivative” means any pharmaceutically acceptable salt, ester, or salt of such ester, of a compound of this invention or any other compound which, upon administration to a recipient (a mammal), is capable of providing
  • a "straight alkyl group of 1 to 6 carbon atoms” includes for example, methyl, ethyl, propyl, butyl, pentyl, hexyl.
  • branched alkyl group of 3 to 6 carbon atoms includes for example, without limitation, /so-butyl, tert-butyl, 2-pentyl, 3-pentyl, etc.
  • a "cycloalkyl group having 3 to 6 carbon” includes for example, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclocyclohexyl (i.e., C 6 H 11 ).
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N - (Ci -4 alkyl)/ salts.
  • the compounds described herein contain one or more asymmetric carbon atoms and thus may occur as racemates and racemic mixtures, single enantiomer, diastereomeric mixtures and individual diastereoisomers. All such isomeric forms of these compounds are expressly included in the present invention.
  • Each stereogenic carbon may be of the R or S configuration.
  • Pharmaceutically acceptable salts of the compounds described herein include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • acid salts include: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylhydrogensulfate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycollate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2- naphthylsulfonate, nicotinate, nit
  • any basic nitrogen containing groups of the compounds disclosed herein may be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chlorides, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • lower alkyl halides such as methyl, ethyl, propyl and butyl chlorides, bromides and iodides
  • dialkyl sulfates including dimethyl, diethyl, dibutyl and di
  • any specified range or group is to be understood as a shorthand way of referring to each and every member of a range or group individually as well as each and every possible sub-ranges or sub-groups encompassed therein; and similarly with respect to any sub-ranges or sub-groups therein.
  • the mention of the range of 1 to 6 carbon atoms is to be understood herein as incorporating each and every individual number of carbon atoms as well as sub-ranges such as, for example, 1 carbon atoms, 3 carbon atoms, 4 to 6 carbon atoms, etc. with respect to reaction time, a time of 1 minute or more is to be understood as specifically incorporating herein each and every individual time, as well as sub-range, above 1 minute, such as for example 1 minute, 3 to 15 minutes, 1 minute to 20 hours, 1 to 3 hours, 16 hours, 3 hours to 20 hours etc.;
  • the compound formulae each include each and every individual compound described thereby as well as each and every possible class or sub-group or sub-class of compounds whether such class or sub-class is defined as positively including particular compounds, as excluding particular compounds or a combination thereof; for example an exclusionary definition for the formula (e.g. I) may read as follows: "provided that when one of A and B is -COOH and the other is H, -COOH may not occupy the 4' position".
  • Scheme 1 illustrates a generic example for the preparation of the phosphate monoester /// derived from a primary alcohol (see I), a compound of HIV protease inhibitors (see example 1 (step G and H) in the experimental portion of this document for a specific example of this synthesis).
  • R 2 and R 3 are as defined herein.
  • phosphate monoester /// may use a HIV aspartyl protease inhibitor (/, see U.S. patent no. 6,632,816) as the starting material.
  • the diethyl phosphotriester // was obtained in good yield upon treatment with diethyl chlorophosphate and sodium hydride in a mixture of tetrahydrofuran and triethylphosphate. Then, addition of trimethysilyl bromide in dichloromethane (DCM) gave compound /// in good to excellent yields.
  • DCM dichloromethane
  • Scheme 1A represents another generic example for the preparation of the phosphate monoester IHA derived from a primary alcohol (see IA), a compound of HIV protease inhibitors.
  • n X, Y 1 R 2 , R 3 and Re are as defined herein.
  • Scheme 2 illustrates a generic example for the preparation of the phosphate monoester ///, a compound of HIV protease inhibitors, with a different approach starting from (3S)-3- isobutylamino-azepan-2-one (IV).
  • R 2 and R 3 are as defined herein.
  • the phosphate monoester derivative /// was obtained from (3S)-3- isobutylamino-azepan-2-one (/V) in a seven-step reaction sequence.
  • (2S)-3- isobutylamino-azepan-2-one (/V) was sulfonated with 4-acetamidobenzenesulfonyl chloride in the presence of triethylamine in dichloromethane to give compound V in excellent yields.
  • the derivative Vl was obtained quantitatively upon treatment of V with di- tert-butyl pyrocarbonate and DMAP in acetonitrile.
  • the reductive ring opening with sodium borohydride in ethanol lead to key intermediates VII in good yield.
  • the diethyl phosphotriester VIII was obtained in good yield upon treatment with diethyl chlorophosphate and sodium hydride in a mixture of tetrahydrofuran and triethylphosphate.
  • the Boc protective groups were removed upon treatment with HCI in ethanol to give compound IX quantitatively (T. W. Greene and P. G. M. Wuts, Protective groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, Inc. 1999).
  • Scheme 3 presents the transformation of a diphenylmethyl derivative; (1S,5S)-(1- ⁇ 5-[(4- am ino-benzenesulf onyl)-isobutyl-am ino]-6-hyd roxy-hexy lcarbam oyl ⁇ -2 , 2-diphenyl-ethy I)- carbamic acid methyl ester (PL -100) into its fluorinated phosphate mo ⁇ oester sodium salt analog Xl.
  • This reaction sequence may be used to produce any other similar compounds (compounds) made of unsubstituted (or substituted) diphenylmethyl, 1- naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
  • Scheme 4 illustrates a generic example for the transformation of a phosphotriester // into its fluorinated analog XIII in a two-step reaction sequence.
  • This generic example represents a second approach for the synthesis of fluorinated compounds described herein.
  • the fluorine atom is added to the phosphotriester // instead of the primary alcohol derivative of general formula / or, more specifically, PL-100 as shown on scheme 3.
  • This alternate reaction sequence may be used to produce any other similar compounds made of unsubstituted (or substituted) diphenylmethyl, 1-naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
  • R 2 and R 3 are as defined herein.
  • Scheme 5 illustrates exemplary synthesis of various ester compounds XVI described herein.
  • the ester compounds are known to be easily cleaved in vivo by esterase enzymes and, as a result, may release the active ingredient.
  • R 2 is set as a diphenylmethyl group.
  • this reaction sequence may be used to produce any other similar compounds made of unsubstituted (or substituted) diphenylmethyl, 1- naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
  • R 1A represents the "residue" of the acid molecule that is linked to the free primary alcohol group present on intermediate XV and is as defined herein.
  • the compounds XVI are generally obtained in a three-step reaction sequence in high yields.
  • HABt 1-hydroxybenzotriazole
  • EDAC 1-[3-(dimethylamino)propyl]-3- ethylcarbodiimide hydrochloride
  • acetyl ester was obtained quantitatively using acetic anhydride in the presence of ⁇ /, ⁇ /-dimethylaminopyridine (DMAP) in dichloromethane (DCM). Cleavage of the Boc protective group was achieved quantitatively upon treatment with trifluoroacetic acid (TFA) in DCM.
  • TFA trifluoroacetic acid
  • a second coupling with (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid is performed on the primary amino group of intermediate XV with HOBt and EDAC to give the desired compounds XVI in good to excellent yields.
  • catalytic hydrogenation of a benzyloxycarbonyl group is performed using 10% palladium on carbon to give the final compound XVII.
  • the compounds described herein may be modified by appending appropriate functionalities to enhance selective biological properties.
  • modifications may include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethyleneglycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • compositions of this invention may contain any conventional non-toxic pharmaceutically acceptable carriers, adjuvants or vehicles.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the compounds described herein When the compounds described herein are administered in combination with a drug which may be metabolized by CYP450, they may be administered sequentially or concurrently to the patient. Administration of the compound and drug may also be separated by a suitable time interval.
  • HIV-1 HIV-1 , -2 Human immunodeficiency virus type 1 , type 2
  • analytical plates may be treated with a 0.3% ninhydrin solution in ethanol containing 3% acetic acid and/or a CAM solution made of 20 g (NH ⁇ 6 Mo 7 O 24 and 8.3 g Ce(SO 4 J 2 polyhydrate in water (750 mL) containing concentrated sulfuric acid (90 mL).
  • Preparative HPLC were performed on a Gilson apparatus equipped with a C18 column, a 215 liquid handler module and 25 mL/min capacity head pumps. The HPLC is operated with a Gilson UniPoint System Software.
  • HPLC system 2 Gilson #305-25 mL pumps, Gilson #215 liquid handler for injection and collection and a Gilson #155 UV-Vis absorbance detector, all controlled from a Gilson Unipoint V1.91 software
  • Melting points (mp) were determined on a B ⁇ chi 530 melting point apparatus in capillary tubes and were uncorrected.
  • Mass spectra were recorded on a Hewlett Packard LC/MSD 1100 system using APCI or electrospray sources either in negative mode or positive mode.
  • ⁇ /o-isobutyl-L- ⁇ -amino-caprolactam (IV) (4.1 g free base) was dissolved in DCM (200 mL) and treated with 4.0 g triethylamine, followed by 4-acetamidobenzenesulfonyl chloride (5.2 g). A 0.1 g portion of dimethylaminopyridine was added and the mixture was stirred 5 h. The resulting thick slurry was poured into 500 mL 0.5 M HCI and shaken vigorously. The solid in the biphasic solution was filtered out and washed with cold acetone to give 7.3 g (87%) of clean product.
  • Step C Preparation of (3S)-3- ⁇ [4-(acetyl-tert-butoxycarbonyl-amino)- benzenesulfonyl]-isobutyl-amino ⁇ -2-oxo-azepane-1-carboxylic acid tert-butyl ester (Boc activation) (V/)
  • Step D Preparation of (1S)-4-amino-/V-(5-amino-1-hydroxymethyl-pentyl)- ⁇ /- isobutyl-benzenesulfonamide (V7/-de protected) (reductive ring opening and deprotection)
  • V7/-de protected reductive ring opening and deprotection
  • a 3.0 g portion of (SSJ-S-IH-t ⁇ cetyl-tert-butoxycarbonyl-aminoJ-benzenesulfonyll-isobutyl- amino ⁇ -2-oxo-azepane-1-carboxylic acid tert-butyl ester (W 1 step C) is dissolved in 40 mL EtOH followed by 750 mg NaBH 4 . Brief heating with a heat gun gives a clear solution.
  • Step F Preparation of (1S,5S)-(1- ⁇ 5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- hydroxy-hexylcarbamoyl ⁇ -2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-100)
  • step D The title compound was prepared from (1 S)-4-amino- ⁇ /-(5-amino-1-hydroxymethyl-pentyl)- ⁇ /-isobutyl-benzenesulfonamide (W/-deprotected) (step D) and (2S)-2- methoxycarbonylamino-S ⁇ -diphenyl-propionic acid (step E) using the coupling procedure with HOBt and EDAC described in example 3 (step D). The final product was obtained in 67% yield (121 mg).
  • Step G Preparation of (1S,5S)- ⁇ 1-[5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6-
  • the PL-100 compound (product of step F, 203 mg, 0.325 mmol) was dissolved in dry tetrahydrofuran (3 ml.) and 0.2 mL triethylphosphate under N 2 atmosphere. The mixture was stirred at this temperature for 15 min, followed by the addition of diethyl chlorophosphate (0.061 mL, 0.423 mmol). Sodium hydride (60% in mineral oil) (17 mg,
  • Step H Preparation of (1 S,5S)-(1- ⁇ 5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- phosphonooxy-hexylcarbamoyl ⁇ -2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-461)
  • step G The product of step G prepared above (152 mg) was dissolved in anhydrous dichloromethane (3.0 ml_). Trimethylsilyl bromide (0.5 ml_) was added at 0 0 C. The mixture was stirred during 1h at this temperature and overnight at room temperature. The solvent was evaporated and 0.2 ml_ water was added to the residue. 3 ml_ EtOH was added mixed and evaporated. This step was repeated three times and the residue dried in vacuo. Yields 98 mg 70% of the title derivatives of this first example.
  • Step B Preparation of (2S)-phosphoric acid 6-amino-2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-hexyl ester diethyl ester (IX)
  • the crude product obtained in the previous step (WW 1 2.66 g) is dissolved in 12 ml_ EtOH. 4 mL of HCI co n e- is added and heated briefly to 70 0 C then left at room temperature for 3h. The solvent is evacuated and the residue triturated with 50 m L ether. The thick residue is S then dissolved in 3 mL ice water and the pH adjusted to 12 with 50% NaOH.
  • Step C Preparation of (2S)-2-methoxycarbonylamino-3-naphthalen-2-yl-0 propionic acid (or L-Moc-2-naphthylalanine)
  • Step D Preparation of (1 S,5S)-(1 - ⁇ 5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- phosphonooxy-hexylcarbamoyl ⁇ -naphthalen ⁇ -yl-ethyO-carbamic acid methyl ester (PL-5 507) 100 mg L-Moc-2-naphthylalanine (step C) was activated with 100 mg EDAC and 57 mg HOBt in 1.5 mL DMF for 30 minutes.
  • step B 100 mg of phosphoric acid 6-amino-2-[(4- amino-benzenesulfonyl)-isobutyl-amino]-hexyl ester diethyl ester (step B) was added and left to stir at room temperature for 1 h. 40 mL of 1M K 2 CO 3 was added to the DMF solution and left for 10 min. 50 mL of EtOAc was then added and the mixture was then agitated vigorously. Separation of the EtOAc phase was effected, followed by extraction with 5% citric acid (50 mL) once, then water (50 mL) 3 times and finally brine. The organic phase was the separated and evaporated.
  • Step B Preparation of (2S.2S) Phosphoric acid mono-(2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6- ⁇ 2-[(morpholine-4-carbonyl)-amino]-3-naphthalen-1-yl- propionylamino ⁇ -hexyl) ester (PL-498) 0
  • Step A Preparation of (1- ⁇ 5-[(4-amino-3-fluoro-benzenesulfonyl)-isobutyl-amino]-6- hydroxy-hexylcarbamoyl ⁇ -2,2-diphenyl-ethyl)-carbamic acid methyl ester (X) (PL-337)
  • step F (0.624 g, 1 mmol) is dissolved in 5 mL MeCN at 24 0 C.
  • SelectFluor 0.35 g (1 mmol) is added in one portion and stirred for 1 h.
  • 1 mL of water is added and the solution was injected directly into a preparative reverse-phase HPLC.
  • the product was collected and lyophilized to give 250 mg (38%) yield of a white solid.
  • Step B Preparation of (1S,5S)- ⁇ 1-[5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-amino]-6-(diethoxy-phosphoryloxy)-hexylcarbamoyl]-2,2-diphenyl-ethyl ⁇ -carbamic acid methyl ester
  • step A The product of step A was phosphorylated with chlorodiethylphosphate following the procedure described in example 1, step G. Yields 157 mg, 68%.
  • Step C Preparation of (1S,5S)-(1- ⁇ 5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol- ⁇ -phosphonooxy-hexylcarbamoyl ⁇ . ⁇ -diphenyl-ethylj-carbamic acid methyl ester (XQ (PL-515)
  • Step A Preparation (1S,5S)-(1- ⁇ 5-[(4-amino-benzenesulfonyI)-isobutyl-amino]-6- phosphonooxy-hexylwrbamoyl ⁇ -2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-461)
  • (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid ((example 1 , step E) 0.9 g, 3 mmol) was activated in DMF (5 ml_) with EDAC (1.7 g, 9 mmol) and HOBt (1.2 g, 9 mmol).
  • EDAC 1.7 g, 9 mmol
  • HOBt 1.2 g, 9 mmol
  • Step B Preparation (1 S,5S)- ⁇ 1-[5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol- ⁇ -tdiethoxy-phosphoryloxyj-hexylcarbamoyll ⁇ -diphenyl-ethyl ⁇ carbamic acid methyl ester (XII)
  • Step C Preparation (1 S,5S)-(1- ⁇ 5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol- ⁇ -phosphonooxy-hexylcarbamoyl ⁇ -diphenyl-ethyO-carbamic acid methyl ester (XIlI, in this specific case is compound Xl) (PL-515)
  • This derivative was prepared from (2S)-acetic acid 6-tert-butoxycarbonylamino-2-[(4-tert- butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester as described in example 15, step B.
  • the yellow solid (66 mg) was used for the next reaction without purification.
  • This derivative was prepared from (2S)-acetic acid 6-amino-2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-hexyl ester (product of step B) as described in example 15, step B.
  • the final product was purified by flash chromatography with a mixture of eluents hexane/ethyl acetate (2/8). A yellow solid was obtained in 70% yield (70 mg).
  • Step C Preparation of (2S,2S)-nicotinic acid 2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl-propionylamino)-hexyl ester (PL-520)
  • step B (2S)-nicotinic acid 6-amino-2-[(4-amino-benzenesulfonyl)-isobutyl- amino]-hexyl ester (100 mg, 0.22 mmol) was dissolved in anhydrous DMF (2 mL) and treated with 0.062 mL (0.45 mmol) of triethylamine followed by EDC (100 mg, 0.56 mmol), HOBt (75 mg, 0.56 mmol) and (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid (56 mg, 0.19 mmol). The mixture was stirred overnight at room temperature.
  • Step C Preparation of (2S,2S)-dimethylamino-acetic acid 2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl- propionylamino)-hexyl ester (PL-534)
  • This title compound was obtained from (1S)- ⁇ 4-[(5-tert-butoxycarbonylamino-1- hydroxymethyl-pentyl)-isobutyl-sulfamoyl)-phenyl ⁇ -carbamic acid tert-butyl ester (intermediate product (VW) of example 1 , step D) as described in example 15, step A using (2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid.
  • the crude product was purified by S flash chromatography eluting with a mixture of
  • Step C Preparation of (2S,2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid 2-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3- diphenyl-propionylamino)-hexyl ester 0
  • Stock solution of PL-100 was prepared in methanol at 25 mM.
  • the 25 mM stock solution was diluted with methanol to 0.25 and 0.125 mM.
  • the 25 mM stock solution and the 0.25 and 0.125 mM solutions were diluted 250 fold for microsomal incubation to a final concentration of 200, 500 and 1000 nM.
  • a sub-stock solution of PL- 100 was prepared in methanol at 25 ⁇ M and diluted in methanol to 2.5 and 0.25 ⁇ M and further diluted 250 fold for microsomal incubation giving final concentrations of 100, 10 and 1nM.
  • Control incubations were performed in the presence of an equal volume of methanol. The final methanol concentration in incubation mixtures was ⁇ 1%. Selective inhibitors of CYP450 were tested in parallel as a positive control.
  • the TESH activity was determined by HPLC-UV analysis following incubation of microsomes with testosterone as. a substrate.
  • Liver microsomes (at a final concentration of 0.30 mg/ml protein) were incubated for 10 minutes at 37°C with various concentrations of testosterone in 0.5 ml reaction mixture, containing 0.1 M phosphate buffer (pH 7.4), 1 mM EDTA, and 3mM magnesium chloride with a NADPH-generating system, PL-100, Ritonavir or Ketoconazole, at appropriate concentrations, were present in the incubation medium with the probe substrate before the reaction was started by addition of NADPH- generating system.
  • the reaction was terminated with 500 ⁇ L ice-cold acetonitrile and the 6 ⁇ -hydroxytestosterone formation was quantified using 6 point standard curve (range 2 to 20 nmol/mL).
  • the analysis of 3 quality controls (LQC, MQC and HQC) was also performed.
  • the associated enzyme activity was expressed as nmol of 6 ⁇ - hydroxytestosterone per mg of protein per min. Data were captured with Millenium chromatographic data management and storage system (version 4.0).
  • EnzFitter software (Biosoft) was used for the calculation of Km, Vmax of probe substrates and apparent Km', Vmax' in the presence of various concentration of PL-100, Ritonavir or selective inhibitors and Ki of PL-100, Ritonavir or selective inhibitors.
  • the cytochrome P450 activities were plotted against substrate concentrations.
  • a nonlinear regression analysis was generated for Michaelis-Menten models in the presence and absence of PL- 100, Ritonavir or selective inhibitors. Lineweaver-Burk Plots
  • Ki was calculated using the equation corresponding to the type of inhibition:
  • V Vmax*[S] / (Km (1 +[I] / Ki) +[S])
  • Km' Km* (1+[I] / Ki )
  • Vmax' Vmax / (1+[l] / Ki)
  • Km' Km / (1+[I] / Ki) 25
  • Vmax' Vmax / (1+[I] / Ki)
  • V Velocity of the enzyme reaction in the presence of inhibitor
  • inhibitor Ki dissociation constant of the enzyme-inhibitor complex
  • Enzyme kinetic parameters Km and Vm ax
  • Ki values of test articles and selective inhibitors were calculated by nonlinear regression analysis using EnzFitter software.
  • EnzFitter software was utilized also for the fitting of the Michaelis-Menten curves and for the generation of the Lineweaver-Burk plots, from the fitted values of Michaelis-Menten curves, allowing the identification of the type of inhibition were appropriate.
  • the testosterone 6 ⁇ -hydrolase activity a selective marker for CYP3A4/5, was assessed in the presence of PL-100.
  • PL-100 could be considered as a strong competitive inhibitor of human CYP3A4 although this compound was approximately 13 fold less potent than Ritonavir.
  • PL-100 is thus considered a strong inhibitor of CYP3A4/5, but with a Ki value 13 fold higher than Ritonavir and 8.5 fold higher than ketoconazole.
  • Ki value 13 fold higher than Ritonavir and 8.5 fold higher than ketoconazole.
  • Example 12 In vivo inhibition of CYP450
  • test article PPL-100
  • concentration 40 mg/mL.
  • Purity of the test article was taken into account for calculation of the concentration.
  • Atazanavir mixture (10 mg/mL) Atazanavir was dissolved in a mixture of 20% ethanol, 50% propylene glycol, 0.1% Tween and 30% water (v/v/v/v), to obtain a concentration of 10 mg/mL.
  • Dosing solutions were prepared shortly prior to dosing as follows: Briefly 4 mL of PPL- 100 mixture (40 mg/mL) was mixed with 4 mL of Atazanavir mixture (10 mg/mL).
  • Sprague Dawley rats (Rattus ⁇ orv ⁇ gicus) aged 7 — 8 weeks at start of dosing were used for the study (Charles River Canada Inc., Montreal, PQ). Animals received Atazanavir (25 mg/kg) and PPL-100 (100 mg/kg). Six female rats were used on each time point.
  • protein was extracted from the plasma samples and PL-100 (the active ingredient released rom PPL-100 under physiological conditions) or atazanavir were detected by HPLC-UV-MSD (1100 series, Hewlett Packard).
  • the concentration of the protease inhibitors and PL-100 was 10 ⁇ M, except for the experiment involving Atazanavir, in which Atazanavir was at concentration of 1.0 ⁇ M and PL-100 was at a concentration of 2.0 ⁇ M.
  • the percent of parent (original protease inhibitor) remaining was measured. The measurements were conducted at time point 60 minutes. The percent of parent remaining quantifies the extent of stability of protease inhibitors, as protease inhibitors are susceptible to being metabolized into daughter molecules in microsomes, decreasing the amount of the original protease inhibitor. The greater the percent of parent remaining, the greater the stability of the protease inhibitor in microsomes.
  • the percent of parent remaining was measured for each protease inhibitor in the presence or absence of PL-100. As a control, the percent of protease inhibitor remaining was also measured in the presence of Ritonavir (RTV) to compare the boosting effect between PL-100 and RTV.
  • RTV Ritonavir

Abstract

The present invention provides pharmaceutical compositions and methods for improving the pharmacokinetics of drugs which may be metabolized by cytochrome P450 monooxigenase, the method may comprise co-administering a compound of formula I, with the drug; when the compound of formula I comprises an amino group, pharmaceutically acceptable ammonium salts thereof, wherein R1 may be, for example, H1 (HO)2P(O)-, (NaO)2P(O)-, alkyl-CO- or cycloalkyl-CO-, wherein X may be, for example, F, Cl, and Br, and wherein R2 and R3 are as defined herein.

Description

METHOD FOR IMPROVING PHARMACOKINETICS
TECHNICAL FIELD OF THE INVENTION
This invention relates to methods for improving the pharmacokinetics of drugs which are metabolized by cytochrome P450 monooxygenase using PPL-100 and related pharmaceutical compositions. In particular, the present invention relates to methods for improving the pharmacokinetics of HIV protease inhibitors by co-administering PPL-100 with such protease inhibitors.
BACKGROUND OF THE INVENTION
inhibitors of the HIV viral protease have been developed relatively recently and their use began only in 1996. Currently, they are considered the most effective drugs against HIV infection. Unfortunately, most current proteases inhibitors are relatively large hydrophobic molecules that possess rather low bioavailability. A high pill burden is therefore required to attain the therapeutic dose in a patient. This is a deterrent, which too often results in patient non-compliance and inadequate treatment results. This situation leads to sub- optimal therapeutic drug concentration that in turns leads to the development of HIV resistant strains. Consequently, there is an urgent need to improve the solubility and bioavailability of proteases inhibitors.
Examples of improved compounds have been developed in the form of prodrugs of aspartyl protease inhibitors such as described, for example, in U.S. patent no. 6,436,989 to Hale et al, the entire content of which is incorporated herein by reference. This patent shows a novel class of molecules characterized by favourable aqueous solubility, high oral bioavailability and facile in vivo generation of the active ingredient. However, it is well known that HIV has the ability to develop resistance to the currently available drugs. Thus, there is a need for alternative HIV protease inhibitors active towards wild-type and resistant viral strains. Thus, molecules derived from current HIV protease inhibitors showing enhanced solubility and bioavailability is desirable to fight resistant viral strains.
A unique class of aromatic derivatives which are inhibitors of aspartyl proteases is described in U.S. patent no. 6,632,816 to Stranix et al, the entire content of which is incorporated herein by reference. This patent includes, more particularly, /V-synthetic amino acid substituted L-lysine derivatives possessing potent aspartyl protease inhibitory properties. However, it would be advantageous to improve these derivatives by enhancing aqueous solubility and bioavailability in order to reduce the pill burden and to favour patient's compliance. Since it is challenging to generate active protease inhibitors, specifically toward wild-type and resistant strains, the formation of derivatives of original HIV protease inhibitors such as inhibitors described in U.S. patent no. 6,632,816 to Stranix βt al, known to be active toward resistant strains represents a viable route with considerable advantages. More particularly, generation of compounds and formulations with enhanced aqueous solubility, oral bioavailability, time of duration and formulation properties along with other advantages is desirable in the development of an effective drug. Protease inhibitors with improved pharmacokinetics are therefore desirable.
SUMMARY OF THE INVENTION
Lysine-based compounds with increased solubility and improved oral bioavailability are described herein and have been described in United States patent application No. 10/902,935 filed on August 2, 2004 and published on February 2, 2006 under No. 2006/0025592A1 , the entire content of which is incorporated herein by reference. These compounds may readily be cleaved in vivo to release an active ingredient which has an affinity for aspartyl proteases and which may act as a protease inhibitor. More particularly, the active ingredient may bind, for example, to an HIV aspartyl protease (U.S. patent no. 6,632,816) and may inhibit this enzyme. The Lysine-based compounds are also referred herein as a protease inhibitor precursor.
Upon in vivo physiological conditions (e.g., metabolic, enteric and/or gastrointestinal conditions, etc.) the Lysine-based compounds, allow for the release of a protease inhibitor (e.g., aspartyl protease inhibitor). The Lysine-based compounds may thus serve as means for improving the solubility and/or bioavailability of protease inhibitors and therefore may reduce the pill burden and/or reduce dosages needed for inhibition. Improved treatment of HIV-infected patients and favourable patient's compliance may consequently occur.
The compounds described herein may be used to inhibit the activity of cytochrome P450 monooxygenase (CYP450). More particularly, these compounds may be used to inhibit the activity of CYP3A4, including for example, CYP3A4/5. The compounds described herein may thus be used with drugs which are metabolized by cytochrome P450 monooxygenase in order to improve their pharmacokinetics. More particularly, these compounds may be used to improve the pharmacokinetics of drugs which are metabolized by CYP3A4, including CYP3A4/5.
Some of the drugs which may benefit from inhibition of CYP450, may include, for example, the immunosupressants cyclosporine, FK-506 and rapamycin, the chemotherapeutic agents taxol and taxotere, the antibiotic clarithromycin and the HIV protease inhibitors, A-77003, A-80987, MK-639. VX-478, AG1343, DMP-323, XM-450, BILA2011 BS, BILA 1096 BS, BILA 2185 BS1 BMS 186,318, LB71262, SC-52151, SC-629 (N,N-dimethylglycyl-N-(2-hydroxy-3-(((4-methoxyphenyl)sulfonyl)(2-methylpropyl) amino)- 1-(phenylmethyl)propyl)-3-methyl-L-valinamide), KNI-272, CGP 53437, CGP 57813 and U-103017).
Exemplary embodiments of compounds encompassed by the present invention and which may be used for inhibiting CYP450 (e.g., CYP3A4) may include, for example, a compound of formula I:
Figure imgf000004_0001
pharmaceutically acceptable salts and derivatives thereof (e.g., for example, when the compound of the present invention comprises an amino group, the pharmaceutically acceptable salt may be an ammonium salt),
wherein n may be, for example, 3 or 4,
wherein X and Y, the same or different, may be selected, for example, from the group consisting of H1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -OCF3, - CN, -NO2, -NR4R5, -NHCOR4, -OR4, -SR4, -COOR4, -COR4, and -CH2OH or X and Y together define an alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula - OCH2CH2O-,
wherein R6 may be selected, for example, from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
wherein R3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R3A-CO-, wherein R3A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms (e.g. methyl, ethyl-, propyl, /so-propyl, butyl, /so-butyl, te/t-butyl, tert- butyl-CH2-, etc.), a cycloalkyl group having 3 to 6 carbon atoms (e.g. cyclopropyl-, cyclohexyl- etc.), a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, (e.g. cyclopropyl-CH2-, cyclohexyl-CH2-, etc.), an alkyloxy group of 1 to 6 carbon atoms (e.g. CH3O-, CH3CH2O-, /so-butylO-, terf-butylO- (Boc), etc.), tetrahydro-3-furanyloxy, -CH2OH, -CF3, -CH2CF3, - CH2CH2CF3, pyrrolidinyl, piperidinyl, 4-morpholinyl, CH3O2C-, CH3O2CCH2-, Acetyl- OCH2CH2-, HO2CCH2-, 3-hydroxyphenyl, 4-hydroxyphenyl, 4-CH3OC6H4CH2-, CH3NH-, (CH3)2N-, (CH3CH2)2N-, (CH3CH2CHz)2N-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, C6H5CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl-, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4- quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000005_0001
a picolyl group selected from the group consisting of
Figure imgf000005_0002
a picolyloxy group selected from the group consisting of
Figure imgf000006_0001
a substituted pyridyl group selected from the group consisting of
Figure imgf000006_0002
and a group of formula,
Figure imgf000006_0003
wherein X' and Y', the same or different, may be selected, for example, from the group consisting of H1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -NO2, - NR4R5, -NHCOR4. -OR4, -SR4, -COOR4, -COR4 and -CH2OH,
wherein R4 and R5, the same or different, may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms, wherein R2 may be selected, for example, from the group consisting of a diphenylmethyl group of formula IV
Figure imgf000007_0001
a naphthyl-1 -CH2- group of formula V
Figure imgf000007_0002
a naphthyl-2-CH2- group of formula Vl
Figure imgf000007_0003
VI a biphenylmethyl group of formula VII
Figure imgf000007_0004
and an anthryl-9-CH2- group of formula VIII
Figure imgf000008_0001
VIII
wherein R1 may be H or a physiologically cleavable unit, whereby upon {in vivo) physiological conditions the compound may be converted into an active protease inhibitor. For example, upon cleavage of the physiologically cleavable unit, the compound may be able to release a protease inhibitor.
In accordance with the present invention, Ri may be selected, for example, from the group consisting of (HO)2P(O) and (MO)2P(O), wherein M is an alkali metal (e.g. Na, K, Cs1 etc) or alkaline earth metal (Ca, Mg, etc.).
Further in accordance with the present invention, R1 may be a group of formula R1A-CO-, wherein R1A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms (e.g. methyl, ethyl, propyl, /so-propyl, butyl, /so-butyl, terf-butyl, tert-butyl-CH2-, etc.), a cycloalkyl group having 3 to 6 carbon atoms (e.g. cyclopropyl-, cyclohexyl- etc.), a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, (e.g. cyclopropyl-CH2-, cyclohexyl-CH2-, etc.), an alkyloxy group of 1 to 6 carbon atoms (e.g. CH3O-, CH3CH2O-, /so-butylO-, tert-butylO- (Boc), etc.), -CH2OH, CH3O2C-, CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, (CHa)2NCH2-, (CHa)2CHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, 2- pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 1-methyl-1 ,4-dihydro-3-pyridyl, 2-pyrazinyl, 2- quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyI, a phenyl group of formula
Figure imgf000008_0002
HI a picolyl group selected from the group consisting of
Figure imgf000009_0001
Figure imgf000009_0002
(4-picolyl)
a picolyloxy group selected from the group consisting of
Figure imgf000009_0003
(2-picolyloxy) (3-picolyloxy) (4-picolyloxy)
a substituted pyridyl group selected from the group consisting of
Figure imgf000009_0004
(substituted 2-pyridyl) (substituted 3-pyridyl) (substituted 4-pyridyl)
and a group of formula,
Figure imgf000009_0005
wherein X1, Y1, R4 and R5 are as defined herein.
The compounds mentioned herein may thus be used for improving the pharmacokinetics of drugs in the methods, pharmaceutical compositions, kits and uses described herein, More particularly, the present invention relates to a pharmaceutical composition which may comprise: a) a compound of formula I as described herein or a pharmaceutically acceptable salt thereof, b) a drug which may be metabolized by a cytochrome P450 monooxigenase (e.g., CYP3A4), and; c) a pharmaceutically acceptable carrier; where the compound of formula I is represented by;
Figure imgf000010_0001
wherein n may be 3 or 4,
wherein X and Y, the same or different, may be selected, for example, from the group consisting of H1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -OCF3, - CN, -NO2, -NR4R5. -NHCOR4, -OR4, -SR4, -COOR4, -COR4, and -CH2OH or X and Y together may define an alkylenedioxy group which may be selected, for example, from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula -OCH2CH2O-,
wherein R8 may be selected from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
wherein R3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R3A-CO-, where R3A may be selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, -CH2OH1 -CF3, -CH2CF3, -CH2CH2CF3. pyrrolidinyl, piperidinyl, 4-morpholinyl, CH3O2C-, CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 3-hydroxyphenyl, 4-hydroxyphenyl, 4-CH3OC6H4CH2-, CH3NH-, (CHa)2N-, (CH3CH2)2N-, (CH3CH2CH2)2N-. HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, C6H5CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl-, 2-pyrazinyl, 2- quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000011_0001
10 a picolyl group which may be selected from the group consisting of
Figure imgf000011_0002
i s a picolyloxy group which may be selected from the group consisting of
Figure imgf000011_0003
a substituted pyridyl group which may be selected from the group consisting of
20
and
Figure imgf000011_0005
Figure imgf000011_0004
a group of formula
Figure imgf000012_0001
wherein X1 and Y", the same or different, may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -NO2, - NR4R5, -NHCOR4, -OR4, -SR4, -COOR4, -COR4 and -CH2OH1
wherein R4 and R5, the same or different, may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
wherein R2 may be selected from the group consisting of a diphenylmethyl group of formula IV
Figure imgf000012_0002
a naphthyl-1 -CH2- group of formula V
Figure imgf000012_0003
a naphthyl-2-CH2- group of formula Vl
Figure imgf000013_0001
a biphenylmethyl group of formula VII
Figure imgf000013_0002
and an anthryl-9-CH2- group of formula VIII
Figure imgf000013_0003
VIII wherein R1 may be H or a physiologically cleavable unit, whereby upon (in vivo) physiological conditions the compound may be converted into an active protease inhibitor. For example, upon cleavage of the physiologically cleavable unit, the compound may be able to release a protease inhibitor.
In accordance with the present invention, Ri may be selected, for example, from the group consisting of H1 (HO)2P(O) and (MO)2P(O) (wherein M may be, for example, an alkali metal or alkaline earth metal) and a group of formula RiA-CO-, where RiA rnay be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH2OH, CH3O2C-, CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 2-hydroxyphenyl, 3-hydroxyphenyl, 4- hydroxyphenyl, (CHa)2NCH2-, (CH3)2CHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 1-methyl-1 ,4-dihydro-3-pyridyl, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000014_0001
III
a picolyl group selected from the group consisting of
Figure imgf000014_0002
(2-picolyl) (3-picoIyl) (4-picolyl)
a picolyloxy group selected from the group consisting of
Figure imgf000014_0003
(2-picolyloxy) (3-picolyloxy) (4-picolyloxy)
a substituted pyridyl group selected from the group consisting of
Figure imgf000015_0001
(substituted 2-pyiϊdyl) (substituted 3-pyridyl) (substituted 4-pyridyl)
and a group of formula,
Figure imgf000015_0002
wherein X", Y1, R4 and R5 are as defined herein.
More particularly, the present invention provides in one aspect thereof, a pharmaceutical composition which may comprise; a) a compound of formula Il and pharmaceutically acceptable salts thereof, b) a drug which may be metabolized by a cytochrome P450 monooxigenase (e.g., CYP3A4), and; c) a pharmaceutically acceptable carrier; where the compound of formula Il is represented by;
Figure imgf000015_0003
wherein n may be 3 or 4,
wherein X and Y, the same or different, may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F1 Cl, Br, I, -CF3, -OCF3, -CN, -NO2, -NR4Rs, - NHCOR4, -OR4, -SR4, -COOR4, -COR4, and -CH2OH or X and Y together may define, for example, an alkylenedioxy group which may be selected from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula - OCH2CH2O-,
wherein R6 may be selected, for example, from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
wherein R3 may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R3A-CO-, where R3A rnay be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, -
CH2OH, -CF3, -CH2CF3, -CH2CH2CF3, pyrrolidinyl, piperidinyl, 4-morpholinyl, CH3O2C-,
CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 3-hydroxyphenyl, 4-hydroxyphenyl, 4-
CH3OCeH4CH2-, CH3NH-, (CH3)2N-, (CH3CH2)2N-, (CH3CH2CH2)2N-, HOCH2CH2NH-,
CH3OCH2O-, CH3OCH2CH2O-, C6H5CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl-, 2- pyrazinyl, 2-quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000016_0001
a picolyl group which may be selected from the group consisting of
Figure imgf000016_0002
a picolyloxy group which may be selected from the group consisting of
Figure imgf000017_0001
a substituted pyridyl group which may be selected from the group consisting of
Figure imgf000017_0002
a group of formula
Figure imgf000017_0003
wherein X' and Y', the same or different, may be selected from the group consisting of H, a straight a Iky I group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F1 Cl, Br, I, -CF3, -NO2, -NR4Rs, - NHCOR4, -OR4, -SR4, -COOR4, -COR4 and -CH2OH,
wherein R4 and R5, the same or different, may be selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
wherein R2 may be selected from the group consisting of a diphenylmethyl group of formula IV
Figure imgf000018_0001
a naphthyl-1-CH2- group of formula V
Figure imgf000018_0002
a naphthyl-2-CH2- group of formula Vl
Figure imgf000018_0003
VI a biphenylmethyl group of formula VII
Figure imgf000018_0004
and an anthryl-9-CH2- group of formula VIII
Figure imgf000019_0001
VIII
wherein R1 may be H or a physiologically cleavable unit, whereby upon physiological conditions (in vivo) the compound may be converted into an active protease inhibitor.
In accordance with the present invention, R1 may be selected, for example, from the group consisting of H, (HO)2P(O) and (MO)2P(O) (wherein M may be an alkali metal or alkaline earth metal), and a group of formula R1A-CO-, where R1A may be selected, for example, from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, -CH2OH, CH3O2C-, CH3O2CCH2-, Acetyl- OCH2CH2-, HO2CCH2-, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxy phenyl, (CHs)2NCH2- , (CH3)ZCHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-. CH3OCH2CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 1-methyl-1 ,4-dihydro-3-pyridyl, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4- quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000019_0002
III
a picolyl group which may be selected from the group consisting of
Figure imgf000020_0002
(2-picolyl) (3-picolyl)
Figure imgf000020_0001
a picolyloxy group which may be selected from the group consisting of
Figure imgf000020_0003
(2-picolyloxy) (3-picolyl oxy) (4-picolyloxy)
a substituted pyridyl group which may be selected from the group consisting of
Figure imgf000020_0004
Figure imgf000020_0005
(substituted 4-pyridyl)
and a group of formula,
Figure imgf000020_0006
wherein X', Y1, R4 and R5 are as defined herein.
In accordance with an embodiment of the present invention, pharmaceutical compositions which comprise compounds of formula Il wherein R6 may be, for example, /so-butyl and n may be 3 are encompassed herewith. In accordance with an additional embodiment of the present invention, pharmaceutical compositions which comprise compounds of formula Il wherein R8 may be, for example, /so-butyl and n may be 4 are also encompassed herewith.
In a particular embodiment of the present invention, Ri may be selected, for example, from the group consisting of H, (HO)2P(O) and (NaO)2P(O).
In another particular embodiment of the present invention, Ri may be selected, for example, from the group consisting of CH3CO, 3-pyridyl-CO, (CH3)2NCH2CO and
Figure imgf000021_0001
In accordance with another particular embodiment of the present invention, pharmaceutical compositions which comprise compounds of formula Il wherein R3 may be selected, for example, from the group consisting of CH3CO, CH3O-CO, (CH3)2N-CO, 3- pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO are encompassed by the present invention.
In accordance with an embodiment of the present invention, X may be 4-NH2 and Y may be H or F.
In accordance with an embodiment of the present invention, X' and Y' may both be H.
In accordance with a particular embodiment of the present invention, pharmaceutical compositions which comprise compounds of formula Il wherein R2 may be selected, for example, from the group consisting of a diphenylmethyl group of formula IV1 a naphthyl-1- CH2- group of formula V, a naphthyl-2-CH2- group of formula Vl, a biphenylmethyl group of formula VII and an anthryl-9-CH2- group of formula VIII are encompassed herewith.
For example, R2 may, more particularly, be selected from the group consisting of a diphenylmethyl group of formula IV, a naphthyl-1-CH2- group of formula V, and a naphthyl- 2-CH2- group of formula Vl.
In a further aspect, the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R6 is /so-butyl, n is 4 and R2 is a diphenylmethyl group of formula IV. In accordance with an embodiment of the present invention, R1 may be selected from the group consisting of H, (HO)2P(O) and (NaO)2P(O).
In accordance with a further embodiment of the present invention, R1 may be selected from the group consisting of CH3CO, 3-pyridyl-CO, (CH3J2NCH2CO and (CH3)2CHCH(NH2)CO.
In accordance with an additional embodiment of the present invention, R3 may be selected, for example, from the group consisting of CH3CO, CH3O-CO1 (CH3)2N-CO, 3- pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO.
In accordance with an embodiment of the present invention, X may be 4-NH2 and Y may be H or F.
More particularly, in accordance with an embodiment thereof, the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X may be, for example, 4-NH2, Y may be H, X' may be H, Y' may be H and R3 may be CH3O-CO.
In accordance with a particular embodiment of the present invention, Ri may be (HO)2P(O).
In accordance with another particular embodiment of the present invention, R1 may be (NaO)2P(O).
In accordance with a further particular embodiment of the present invention, R1 may be H.
More particularly, in accordance with a further embodiment thereof, the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X may be 4-NH2, Y may be 3-F, X' may be H, Y' may be H and R3 may be CH3O- CO.
In accordance with a particular embodiment of the present invention, R1 may be (HO)2P(O). In accordance with another particular embodiment of the present invention, R1 may be (NaO)2P(O).
In accordance with a further particular embodiment of the present invention, Ri may be H.
More particularly, in accordance with an additional embodiment thereof, the present invention provides a pharmaceutical composition which may comprise a compound of formula II, wherein X is 4-NH2, Y is H or 3-F, X1 is H, Y' is H and R3 is CH3CO.
In accordance with a particular embodiment of the present invention, Ri may be (HO)2P(O).
In accordance with another particular embodiment of the present invention, Ri may be (NaO)2P(O).
In accordance with a further particular embodiment of the present invention, R1 may be H.
More particularly, in accordance with an embodiment thereof, the present invention further provides a pharmaceutical composition which may comprise a compound of formula II, X is 4-NH2, Y is H or 3-F, X* is H1 Y' is H and R3 is 4-morphoHne-CO.
In accordance with an embodiment of the present invention X may be 4-NH2, Y may be H, X1 may be H, Y* may be H and R3 may be CH3O-CO.
In accordance with a particular embodiment of the present invention R1 may be 3-pyridyl- CO.
In accordance with another particular embodiment of the present invention R1 may be (CH3)2NCH2CO.
In accordance with yet another particular embodiment of the present invention R1 may be (CHa)2CHCH(NH2)CO.
In accordance with an additional particular embodiment of the present invention R1 may be CH3CO. In an additional embodiment the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R8 is /sobutyl, n is 4, X' and Y" are both H, R2 is Naphtyl-1-CH2-, X is 4-NH2, Y is H, R3 is 4-morpholine-CO and R1 may be selected, for example, from the group consisting of H, (HO)2P(O) and (NaO)2P(O).
In a further embodiment, the present invention provides pharmaceutical compositions comprising a compound of formula II, wherein R6 is /so-butyl, n is 4, X1 and Y' are both H, R2 is Naphtyl-2-CH2-, X is 4-NH2, Y is H1 R3 is CH3O-CO and Ri may be selected, for example, from the group consisting of H, (HO)2P(O) and (NaO)2P(O).
More particularly, the present invention provides a pharmaceutical composition which may comprise; a) a compound of formula Ha
Figure imgf000024_0001
and pharmaceutically acceptable salts thereof,
wherein X and Y, the same or different, may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -OCF3, - CN, -NO2, -NR4R5, -NHCOR4, -OR4, -SR4, -COOR4, -COR4. and -CH2OH or X and Y together define an alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula - OCH2CH2O-,
wherein X' and Y', the same or different, may be selected, for example, from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -NO2, - NR4R5, -NHCOR4, -OR4, -SR4, -COOR4, -COR4 and -CH2OH1 wherein n, R1, R3, R4, R5 and Rβ are as defined herein;
b) a drug which may be metabolized by a cytochrome P450 monooxigenase, and;
c) a pharmaceutically acceptable carrier.
In accordance with an embodiment of the present invention, R1 may be selected from the group consisting of H, (HO)2P(O) and (MO)2P(O), wherein M may be an alkali metal or alkaline earth metal and a group of formula R1A-CO-, R1A which may be selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH2OH, CH3O2C-, CH3O2CCH2-, ACeIyI-OCH2CH2-, HO2CCH2-, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, (CH3)2NCH2-, (CHa)2CHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, 2-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl. 1 -methyl- 1 ,4-dihydro-3-pyridyl, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4- quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000025_0001
a picolyl group selected from the group consisting of
Figure imgf000025_0002
(2-picolyl) (3-picolyl) (4-picoIyl)
a picolyloxy group selected from the group consisting of
Figure imgf000026_0001
(2-picolyloxy) (3-picolyloxy) (4-picolyloxy)
a substituted pyτidyl group selected from the group consisting of
Figure imgf000026_0002
(substituted 2-pyridyl) (substituted 3-pyridyl) (substituted 4-pyridyl)
and a group of formula,
Figure imgf000026_0003
wherein X', Y', R4 and R5 are as defined herein.
In accordance with another embodiment of the present invention, R6 may be feo-bυtyl.
In accordance with yet another embodiment of the present invention, n may be 4.
In accordance with a further embodiment of the present invention, R1 may be selected from the group consisting of H, (HO)2P(O) and (NaO)2P(O).
Further in accordance with an embodiment of the present invention, Ri may be selected, for example, from the group consisting of CH3CO, 3-pyridyl-CO, (CH3J2NCH2CO and (CH3)2CHCH(NH2)CO.
Also in accordance with an embodiment of the present invention, R3 may be selected from the group consisting of CH3CO, CH3O-CO1 (CH3)2N-CO, 3-pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO. In accordance with another embodiment of the present invention, R3 may be selected from the group consisting of CH3CO, CH3O-CO, (CH3)2N-CO, 3-pyridyl-CO, 4-pyridyl-CO and 4-morpholine-CO.
In accordance with an embodiment of the present invention, X may be 4-NH2 and Y may be H or F.
In accordance with another embodiment of the present invention, X may be 4-NH2 and Y may be H or F.
Also in accordance with an embodiment of the present invention, X may be 4-NH2, Y may be H or 3-F, X1 may be H, Y' may be H and R3 may be CH3CO.
In accordance with another embodiment of the present invention, X may be 4-NH2, Y may be H or 3-F, X' may be H, Y' may be H and R3 may be 4-morpholine-CO.
In accordance with a particular embodiment of the present invention, X may be 4-NH2, Y may be H1 X' may be H, Y" may be H, R3 may be CH3O-CO and R1 may be (HO)2P(O), (NaO)2P(O) or H.
In accordance with another particular embodiment of the present invention, X may be 4- NH2, Y may be 3-F, X' may be H, Y' may be H1 R3 may be CH3O-CO and Ri may be (HO)2P(O). (NaO)2P(O) or H.
In accordance with a further particular embodiment of the present invention, X may be 4- NH2, Y may be H or 3-F, X* may be H, Y' may be H1 R3 may be CH3CO and Ri may be (HO)2P(O)1 (NaO)2P(O) or H.
In accordance with another particular embodiment of the present invention, X may be 4- NH2, Y may be H or 3-F, X1 may be H, Y' may be H and R3 may be 4-morpholine-CO.
In accordance with an additional embodiment of the present invention, X may be 4-NH2, Y may be H1 X" may be H, Y' may be H1 R3 may be CH3O-CO and R1 may be 3-pyridyl-CO, (CH3)2NCH2CO, (CH3)2CHCH(NH2)CO or CH3CO. In accordance with another embodiment of the present invention, X may be 4-NH2, Y may be 3-F, X' may be H, Y' may be H, R3 may be CH3O-CO and Ri may be 3-pyridyl-CO, (CHg)2NCH2CO or (CH3)2CHCH(NH2)CO.
Other compounds which may be used to carry out the present invention may include, for example, a compound of formula MA;
Figure imgf000028_0001
IIA
wherein Y, π, R1, R2, R3, X" and Y' are as defined herein.
In accordance with the present invention, R1 may be, for example, H1 (HO)2P(O) or (NaO)2P(O). Further in accordance with the present invention, n may be 4. Y may be, for example, H. R3 may be, for example. CH3O-CO. R2 may be, for example, a diphenylmethyl group of formula IV, where X' and Y1 may be, for example H1
Figure imgf000028_0002
Compounds of formula HA' as well as pharmaceutically acceptable salts and derivatives thereof may be used to carry out the present invention,
Figure imgf000029_0001
ILV
such as, for example, compound of formula HA' wherein R1 is H, or (HO)2P(O) or, 5 compound of formula MA' wherein Ri is (NaO)2P(O).
For example, pharmaceutical compositions, methods, uses and kits encompassed by the present invention may comprise one or more of the following compounds and combination thereof; I O
-a compound of formula Ha wherein n is 4, R1 is (HO)2P(O), X is 4-NH2, Y is H, X' is H, Y1 is H, R6 is /so-butyl and R3 is CH3O-CO,
- a compound of formula Ha wherein n is 4, R1 is (NaO)2P(O), X is 4-NH2, Y is H, X' is H, 15 Y" is H, Rβ is /so-butyl and R3 is CH3O-CO,
- a compound of formula Ua wherein n is 4, R1 is (HO)2P(O), X is 4-NH2, Y is H, X' is H, Y' is H, R6 is /so-butyl and R3 is CH3CO, 0 - a compound of formula Ha wherein n is 4, R1 is (HO)2P(O), X is 4-NH2, Y is 3-F, X' is H, Y1 is H, R6 is /so-butyl and R3 is CH3O-CO,
- a compound of formula Ha wherein n is 4, Ri is CH3CO1 X is 4-NH2, Y is H, X' is H, Y' is H, R6 is /so-butyl and R3 is CH3O-CO,
25
- a compound of formula Ha wherein n is 4, R1 is 3-pyridyl-CO, X is 4-NH2, Y is H, X' is H, Y' is H, R6 is /so-butyl and R3 is CH3O-CO1
- a compound of formula Ha wherein n is 4, R1 is (CHa)2NCH2CO1 X is 4-NH2, Y is H, X' is 30 H1 Y* is H1 R6 is /so-butyl and R3 is CH3O-CO1 - a compound of formula Ha wherein n is 4, R1 is (CHa)2CHCH(NH2)CO, X is 4-NH2, Y is H, X' is H, Y" is H, R6 is /so-butyl and R3 is CH3O-CO,
- a compound of formula lib wherein n is 4, R1 is (HO)2P(O), X is 4-NH2, Y is H, X" is H, Y' is H, R8 is /so-butyl and R3 is CH3O-CO and wherein the naphthyl group is a naphthyl-2- CH2 group,
- a compound of formula Hb wherein n is 4, R1 is (HO)2P(O), X is 4-NH2, Y is H1 X" is H, Y' is H, R6 is /so-butyl and R3 is 4-morpholine-CO and wherein the naphthyl group is a naphthyl-1-CH2 group, or
- a combination of any of the above mentioned compounds.
Any compound which is a precursor of an active ingredient described herein may be used to carry out the present invention and is also encompassed by the present invention. For example, compounds being able to release or generate (either in vivo or in vitro) an active ingredient of the following formula;
Figure imgf000030_0001
are encompassed by the present invention.
The above identified active ingredient is identified herein as PL-100 and it is to be understood herein that any precursor able to release or generate the above mentioned exemplary compound either in vivo or in vitro is encompassed by the present invention and may be used to carry out methods, pharmaceutical compositions, kits and uses described herein.
In addition, compounds which able to release (either in vivo or in vitro) an active ingredient of the following formula;
Figure imgf000031_0001
may be used to carry out the present invention and are therefore encompassed by the present invention.
The above identified active ingredient is identified herein as PL-337 and it is to be understood herein that any precursor able to release or generate the above mentioned exemplary compound either in vivo or in vitro may be used to carry out the present invention and is encompassed by the present invention.
The present invention thus relates to the use of a compound described herein for improving the pharmacokinetics of a drug which may affected by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.).
The present invention also relates to methods of treatment which may comprise admininistering to an individual in need, a compound described herein with one or more drugs which may be affected (metabolized) by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.). As indicated herein, administration may be done at the same time or at different time intervals. The compound(s) and drug(s) may be mixed together or not.
The present invention further relates to the use of a compound described herein in the manufacture of a medicament for improving the pharmacokinetics of a drug which may affected by CYP450 and more particularly, CYP3A4 (e.g., CYP3A4/5 etc.).
The present invention further relates to the use of a compound described herein and a drug which may affected by CYP450 in the manufacture of a medicament for treating a patient in need.
The term "pharmaceutically effective amount" refers to an amount effective in treating, preventing or reducing the risk or probability of HlV infection or of reducing HIV burden. The term "pharmaceutically effective amount" also refers to an amount effective in treating, preventing or reducing the risk or probability of developing acquired immunodeficiency syndrome (AIDS), for delaying the apparition of AIDS, or reducing AIDS symptoms. It is also to be understood herein that a "pharmaceutically effective amount" may be construed as an amount giving a desired therapeutic effect, either taken into a single or multiple doses or in any dosage or route or taken alone or in combination with other therapeutic agents. In the case of the present invention, a "pharmaceutically effective amount" may be understood as an amount having an inhibitory effect (partial or complete) on HIV (HIV-1 and HIV-2 as well as related viruses (e.g., HTLV-I and HTLV-II, and simian immunodeficiency virus (SIV))) infection cycle (e.g., inhibition of replication, reinfection, maturation, budding etc.) and on any organism which rely on aspartyl proteases for its life cycle. An inhibitory effect is to be understood herein as an effect such as a reduction in the capacity of an organism (e.g. HIV) to reproduce itself (replicate), to re-infect surrounding cells, etc, or even a complete inhibition (or elimination) of an organism.
The terms "HIV protease" and "HIV aspartyl protease" are used interchangeably and includes, for example, the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2.
The terms "pharmaceutically acceptable carrier", "pharmaceutically acceptable adjuvant" and "physiologically acceptable vehicle" refer to a non-toxic carrier or adjuvant that may be administered to a patient, together with one or more compounds of the present invention, and which does not destroy the pharmacological activity thereof.
The term "precursor" refers to a compound, such as a Lysine-based compound which is able to be converted into an active ingredient in vitro or in vivo. For example, the compound PL-461 is a precursor of compound PL-100 as when administered to an individual, PL-461 is converted into PL-100 in vivo (e.g., under physiological conditions).
The term "derivative" refers to a compound which has been chemically synthesized from an original compound. For example, when considering the chemical synthesis of PL-461 , PL-461 is a derivative of PL-100.
The term "consisting essentially of means that the pharmaceutical composition includes the specified materials and may include other material that does not materially affect the basic characteristics of the pharmaceutical composition.
Pharmaceutically acceptable derivatives of the compounds of formula I (such as compounds of formulae I, II, Ha, lib, Uc, MA and HA1) and where applicable pharmaceutically acceptable salts thereof such as, for example, ammonium salts are described herein. A "pharmaceutically acceptable derivative" means any pharmaceutically acceptable salt, ester, or salt of such ester, of a compound of this invention or any other compound which, upon administration to a recipient (a mammal), is capable of providing
(directly or indirectly) a an active compound or an antivirally active metabolite or residue thereof.
It is to be understood herein that a "straight alkyl group of 1 to 6 carbon atoms" includes for example, methyl, ethyl, propyl, butyl, pentyl, hexyl.
It is to be understood herein that a "branched alkyl group of 3 to 6 carbon atoms" includes for example, without limitation, /so-butyl, tert-butyl, 2-pentyl, 3-pentyl, etc.
It is to be understood herein, that a "cycloalkyl group having 3 to 6 carbon" includes for example, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclocyclohexyl (i.e., C6H11).
Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N - (Ci-4 alkyl)/ salts.
The compounds described herein contain one or more asymmetric carbon atoms and thus may occur as racemates and racemic mixtures, single enantiomer, diastereomeric mixtures and individual diastereoisomers. All such isomeric forms of these compounds are expressly included in the present invention. Each stereogenic carbon may be of the R or S configuration.
Pharmaceutically acceptable salts of the compounds described herein include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of such acid salts include: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylhydrogensulfate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycollate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2- naphthylsulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, perchlorate. persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, thiocyanate, tosylate, and undecanoate.
Compounds which are encompassed by the present invention also envisions the quaternization of any basic nitrogen containing groups of the compounds disclosed herein. The basic nitrogen may be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chlorides, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
It is to be understood herein, that if a "range" or "group of substances" is mentioned with respect to a particular characteristic (e.g., temperature, concentration, time and the like) of the present invention, the present invention relates to and explicitly incorporates herein each and every specific member and combination of sub-ranges or sub-groups therein whatsoever. Thus, any specified range or group is to be understood as a shorthand way of referring to each and every member of a range or group individually as well as each and every possible sub-ranges or sub-groups encompassed therein; and similarly with respect to any sub-ranges or sub-groups therein. Thus, for example, - with respect to the number of carbon atoms, the mention of the range of 1 to 6 carbon atoms is to be understood herein as incorporating each and every individual number of carbon atoms as well as sub-ranges such as, for example, 1 carbon atoms, 3 carbon atoms, 4 to 6 carbon atoms, etc. with respect to reaction time, a time of 1 minute or more is to be understood as specifically incorporating herein each and every individual time, as well as sub-range, above 1 minute, such as for example 1 minute, 3 to 15 minutes, 1 minute to 20 hours, 1 to 3 hours, 16 hours, 3 hours to 20 hours etc.;
- and similarly with respect to other parameters such as concentrations, elements, etc... It is in particular to be understood herein that the compound formulae each include each and every individual compound described thereby as well as each and every possible class or sub-group or sub-class of compounds whether such class or sub-class is defined as positively including particular compounds, as excluding particular compounds or a combination thereof; for example an exclusionary definition for the formula (e.g. I) may read as follows: "provided that when one of A and B is -COOH and the other is H, -COOH may not occupy the 4' position".
It is also to be understood herein that "g" or "gm" is a reference to the gram weight unit and "C", or " 0C " is a reference to the Celsius temperature unit.
The compounds described herein may easily be prepared using conventional techniques from readily available starting materials. The detailed descriptions of these approaches are presented, for example, in schemes 1 to 5 discussed below.
Scheme 1 illustrates a generic example for the preparation of the phosphate monoester /// derived from a primary alcohol (see I), a compound of HIV protease inhibitors (see example 1 (step G and H) in the experimental portion of this document for a specific example of this synthesis).
Note: a) R2 and R3 are as defined herein.
The synthesis of phosphate monoester /// may use a HIV aspartyl protease inhibitor (/, see U.S. patent no. 6,632,816) as the starting material. The diethyl phosphotriester // was obtained in good yield upon treatment with diethyl chlorophosphate and sodium hydride in a mixture of tetrahydrofuran and triethylphosphate. Then, addition of trimethysilyl bromide in dichloromethane (DCM) gave compound /// in good to excellent yields.
Figure imgf000036_0001
III
Scheme 1A represents another generic example for the preparation of the phosphate monoester IHA derived from a primary alcohol (see IA), a compound of HIV protease inhibitors.
Note: a) n, X, Y1 R2, R3 and Re are as defined herein.
Sc
Figure imgf000037_0001
UA
IA
1) TMS-Br DCM
2) H2O
Figure imgf000037_0002
IUA
The synthesis of phosphate monoester IHA is performed as described for the preparation of /// (scheme 1).
Scheme 2 illustrates a generic example for the preparation of the phosphate monoester ///, a compound of HIV protease inhibitors, with a different approach starting from (3S)-3- isobutylamino-azepan-2-one (IV).
Note: a) R2 and R3 are as defined herein.
As shown in scheme 2, the phosphate monoester derivative /// was obtained from (3S)-3- isobutylamino-azepan-2-one (/V) in a seven-step reaction sequence. Initially, (2S)-3- isobutylamino-azepan-2-one (/V) was sulfonated with 4-acetamidobenzenesulfonyl chloride in the presence of triethylamine in dichloromethane to give compound V in excellent yields. The derivative Vl was obtained quantitatively upon treatment of V with di- tert-butyl pyrocarbonate and DMAP in acetonitrile. The reductive ring opening with sodium borohydride in ethanol lead to key intermediates VII in good yield. The diethyl phosphotriester VIII was obtained in good yield upon treatment with diethyl chlorophosphate and sodium hydride in a mixture of tetrahydrofuran and triethylphosphate. The Boc protective groups were removed upon treatment with HCI in ethanol to give compound IX quantitatively (T. W. Greene and P. G. M. Wuts, Protective groups in Organic Synthesis, 3rd Edition, John Wiley & Sons, Inc. 1999). Then, coupling of the free amino group present on intermediate IX with a variety of synthetic amino acid in the presence of 1-hydroxybenzotriazole (HOBt) and 1-[3-(dimethylamino)propyl]-3- ethylcarbodiimide hydrochloride (EDAC) led to derivative // in good to excellent yields. Finally, addition of trimethysilyl bromide in dichloromethane (DCM) gave compound /// in good to excellent yields.
Scheme 2
Figure imgf000039_0001
Figure imgf000039_0002
BocHN
Figure imgf000039_0003
Figure imgf000039_0004
Scheme 3 presents the transformation of a diphenylmethyl derivative; (1S,5S)-(1-{5-[(4- am ino-benzenesulf onyl)-isobutyl-am ino]-6-hyd roxy-hexy lcarbam oyl}-2 , 2-diphenyl-ethy I)- carbamic acid methyl ester (PL -100) into its fluorinated phosphate moπoester sodium salt analog Xl. This reaction sequence may be used to produce any other similar compounds (compounds) made of unsubstituted (or substituted) diphenylmethyl, 1- naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
Thus, the treatment of PL-100 with Selectfluor™ in acetonitrile gave derivative X in 38% yield. The introduction of the phosphate monoester group was performed as described previously in scheme 1 and 2. First, the diethyl phosphotriester intermediate was obtained in good yield upon treatment with diethyl chlorophosphate and sodium hydride in a mixture of tetrahydrofuran and triethylphosphate. Secondly, addition of trimethysilyl bromide in dichloromethane (DCM) gave the phosphate monoester compound in good to excellent yields. The final product X/ was easily obtained upon treatment of the phosphate monoester with a solution of sodium hydroxide with good yields.
Scheme 3
Figure imgf000040_0001
PL-100 (example 1 , step F)
1) Diethylchlorophospphnaatiee,,
Figure imgf000040_0002
Xl
Scheme 4 illustrates a generic example for the transformation of a phosphotriester // into its fluorinated analog XIII in a two-step reaction sequence. This generic example represents a second approach for the synthesis of fluorinated compounds described herein. In this case, the fluorine atom is added to the phosphotriester // instead of the primary alcohol derivative of general formula / or, more specifically, PL-100 as shown on scheme 3. This alternate reaction sequence may be used to produce any other similar compounds made of unsubstituted (or substituted) diphenylmethyl, 1-naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
Note: a) R2 and R3 are as defined herein.
Briefly, treatment of derivative // with Selectfluor™ in acetonitrile gave derivative XII in good yields. Then, addition of trimethysilyl bromide in dichloromethane (DCM) gave the phosphate monoester compound XIII in good to excellent yields. If desired, the final product XIII may be easily transformed into the phosphate monoester sodium salt analog as described before in scheme 3.
Sc
Figure imgf000041_0001
Scheme 5 illustrates exemplary synthesis of various ester compounds XVI described herein. The ester compounds are known to be easily cleaved in vivo by esterase enzymes and, as a result, may release the active ingredient. In this scheme R2 is set as a diphenylmethyl group. However, this reaction sequence may be used to produce any other similar compounds made of unsubstituted (or substituted) diphenylmethyl, 1- naphthyl, 2-naphthyl, biphenyl and 9-anthryl groups described herein.
Note: a) R1A represents the "residue" of the acid molecule that is linked to the free primary alcohol group present on intermediate XV and is as defined herein.
The compounds XVI are generally obtained in a three-step reaction sequence in high yields. Esterification of (1S)-{4-[(5-tert-butoxycarbonylamino-1-hydroxymethyl-pentyl)- isobutyl-sulfamoyl]-phenyl}-carbamic acid tert-butyl ester (VII) with a variety of acid in the presence of 1-hydroxybenzotriazole (HOBt) and 1-[3-(dimethylamino)propyl]-3- ethylcarbodiimide hydrochloride (EDAC) led to the desired esters XIV in excellent yields. The acetyl ester was obtained quantitatively using acetic anhydride in the presence of Λ/,Λ/-dimethylaminopyridine (DMAP) in dichloromethane (DCM). Cleavage of the Boc protective group was achieved quantitatively upon treatment with trifluoroacetic acid (TFA) in DCM. A second coupling with (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid is performed on the primary amino group of intermediate XV with HOBt and EDAC to give the desired compounds XVI in good to excellent yields. If necessary, catalytic hydrogenation of a benzyloxycarbonyl group is performed using 10% palladium on carbon to give the final compound XVII.
Scheme 5
Figure imgf000043_0002
Figure imgf000043_0001
VII (R1A = CH3) XIV
TFA, DCM, 99%
Figure imgf000043_0003
XVII
As it may be appreciated by the person skilled in the art, the above synthetic schemes are not intended to be a comprehensive list of all means by which the compound described in this application may be synthesized but only represent exemplification of synthesis methods among others. Further methods will be evident to those of ordinary skill in the art.
The compounds described herein may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications may include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
Pharmaceutical compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethyleneglycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
It is therefore understood herein that oral administration or administration by injection are encompassed by the present invention. For example, compounds of the present invention, may, for example, be orally administered in an aqueous solution. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically acceptable carriers, adjuvants or vehicles. The term "parenteral" as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
When the compounds described herein are administered in combination with a drug which may be metabolized by CYP450, they may be administered sequentially or concurrently to the patient. Administration of the compound and drug may also be separated by a suitable time interval.
In the description herein, the following abbreviations are used:
Abbreviation Meaning
Ac Acetyl
AcOH Acetic acid APCI Atmospheric pressure chemical ionization
AIDS Acquired Immunodeficiency Syndrome APV Amprenavir
ATV Atazanavir
AZT 3-Azido-3-deoxythymine (Zidovudine)
Boc Benzyloxycarbonyl t-Butyl tert-Butyl
CAM Cerium ammonium molybdate
DCM Dichloromethane
DMAP Λ/,Λ/-dimethylaminopyridine
DMSO Dimethylsulfoxide
DMF Dimethylformamide
DNA Deoxyribonucleic acid
EDAC 1-[3-(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride
EtOAc Ethyl acetate
EtOH Ethyl alcohol g Gram h hour
HIV-1 , -2 Human immunodeficiency virus type 1 , type 2
HOBt 1 -Hydroxybenzotriazole
HPLC High performance liquid chromatography
HTLV-I, -Il Human T-cell lymphotropic virus type I, type Il
IL-2 lnterleukin-2
IDV Indinavir
Kg Kilogram
L Liter
LC-MS Liquid chromatography-mass spectrometry
LPV Lopinavir
M Molar
MeOH Methyl alcohol mg Milligram mp Melting point min Minute
Moc Methoxycarbonyl mol Mole mL Milliliter mmol Millimole NFV Nelfinavir nm Nanometer nM Nanomolar po Orally rEPO Recombinant erythropoietin
RTV Ritonavir
SQV Saquinavir
TLC Thin layer chromatography
3TC 2\3'-Dideoxy-3-thiacytidine
TFA Trifluoroacetic acid
THF Tetrahydrofuran
EXAMPLES This section describes the synthesis of lysine based compounds able to release an HIV aspartyl protease inhibitors as described herein. These examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention in any way. This section presents the detailed synthesis of compounds no. 1 to 10 of this invention.
Exemplary synthesis schemes of the active ingredients have been disclosed in U.S. patent no. 6,632,816 to Stranix et a/,.
Materials and Methods-preparation of compounds
Analytical thin layer chromatography (TLC) was carried out with 0.25 mm silica gel E. Merck 60 F254 plates and eluted with the indicated solvent systems. Preparative chromatography was performed by flash chromatography, using silica gel 60 (EM Science) with the indicated solvent systems and positive air pressure to allow proper rate of elution. Detection of the compounds was carried out by exposing eluted plates (analytical or preparative) to iodine, UV light and/or treating analytical plates with a 2% solution of p-anisaldehyde in ethaπol containing 3% sulfuric acid and 1% acetic acid followed by heating. Alternatively, analytical plates may be treated with a 0.3% ninhydrin solution in ethanol containing 3% acetic acid and/or a CAM solution made of 20 g (NH^6Mo7O24 and 8.3 g Ce(SO4J2 polyhydrate in water (750 mL) containing concentrated sulfuric acid (90 mL). Preparative HPLC were performed on a Gilson apparatus equipped with a C18 column, a 215 liquid handler module and 25 mL/min capacity head pumps. The HPLC is operated with a Gilson UniPoint System Software.
Semi-preparative HPLC conditions for purification of test compounds: HPLC system: 2 Gilson #305-25 mL pumps, Gilson #215 liquid handler for injection and collection and a Gilson #155 UV-Vis absorbance detector, all controlled from a Gilson Unipoint V1.91 software Column: Alltech (#96053) Hyperprep PEP, C-18, 100 Aa, 8 μm, 22 x 250 mm Flow: 15 mL/min Solvents: A: H2O; B: CH3CN Gradient: 25% to 80% of B over 40 min Detector: absorbance; λ: 210 & 265 nm
The crude material dissolved in acetonitrile to a concentration of around 50 to 80 mg / 2 mL were injected in each run. Fractions were collected in amounts of 9 mL pertaining absorbance was detected at the UV detector.
Unless otherwise indicated, all starting materials were purchased from a commercial source such as Aldrich Co. or Sigma Co.
Melting points (mp) were determined on a Bϋchi 530 melting point apparatus in capillary tubes and were uncorrected.
Mass spectra were recorded on a Hewlett Packard LC/MSD 1100 system using APCI or electrospray sources either in negative mode or positive mode.
Nuclear magnetic resonance (NMR) spectra were recorded on a Bruker AMX-ll-500 equipped with a reversed or QNP probe. Samples were dissolved in deuterochloroform
(CDCI3), deuteroacetone (acetone-dβ), deuteromethanol (CD3OD) or deuterodimethylsulfoxide (DMSO-d8) for data acquisition using tetramethylsilane as internal standard. Chemical shifts () are expressed in parts per million (ppm), the coupling constants (J) are expressed in hertz (Hz) whereas multiplicities are denoted as s for singlet, d for doublet, 2d for two doublets, dd for doublet of doublets, t for triplet, q for quartet, quint, for quintet, m for multiplet, and br s for broad singlet. DETAILED DESCRIPTION OF THE INVENTION
EXAMPLES:
Specific examples for the preparation of derivatives of general formula I
The preparation of PL-100, PL-337 and other compounds of this class is presented in U.S. patent no. 6,632,816 to Stranix et al.
The following compounds were prepared from L-lysine derivatives using the procedures summarized in schemes 1 , 1A, 2, 3, 4 and 5 of this invention.
Example 1. Preparation of (1S,5S)-<1-{5-[(4-amino-benzenesulfonyl)-isobutyl- amino]-6-phosphonooxy-hexylcarbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-461)
The preparation of the title compound is based on schemes 1 and 2 of this invention.
Step A. Preparation of (3S)-3-isobutylamino-azepan-2-one (IV)
L-α-aminoτ-caprolactam (22.0 g) was dissolved in cold dichloroethane (DCM1 200 ml_). isobutyraldehyde (12.6 g) was added slowly and stirred until the heat evolved was dissipated (water forms at the surface). The cold solution was added to 46.5 g of powdered NaBH(OAc)3 in DCM (0.5 L). AcOH (70 mL) was added to the solution. The slightly turbid mixture was stirred at 20 0C for 4 h. A 500 mL solution of 2M NaOH was added slowly to the turbid mixture and the pH adjust to 11 using a concentrated NaOH solution, and then the mixture stirred for a further 20 min. After extraction, the DCM layer was dried with MgSO4, filtered and evaporated. The oil thus obtained crystallizes slowly on standing (27.8 g, 85%) and was used without further purification in the next step.
1H NMR (CDCI3): δ 0.93 (d, J = 6.5, 3H), 0.97 (d, J = 6.5. 3H), 1.39 (t, J = 9.8, 1H), 1.47 (m, 1H), 1.78-1.65 (m, 2H), 2.00-1.93 (m, 2H), 2.32-2.2 (m, 2H), 2.38 (t, J = 9.7, 1H), 3.16 (m, 3H), 6.62 (s, 1H (NH)). mp 52-54 0C (hexanes).
A small sample was converted to the S-methyl benzyl urea by adding the solid to a solution of S-methyl benzyl isocyanate in MeCN. NMR gives 98% ee Step B. Preparation of Mx-isobutyl-/Vα-(4-acetamidobenzenesulfonyl)-L-α- amino— caprolactam (V)
Λ/o-isobutyl-L-α-amino-caprolactam (IV) (4.1 g free base) was dissolved in DCM (200 mL) and treated with 4.0 g triethylamine, followed by 4-acetamidobenzenesulfonyl chloride (5.2 g). A 0.1 g portion of dimethylaminopyridine was added and the mixture was stirred 5 h. The resulting thick slurry was poured into 500 mL 0.5 M HCI and shaken vigorously. The solid in the biphasic solution was filtered out and washed with cold acetone to give 7.3 g (87%) of clean product.
1H NMR (DMSO-dβ): . 0.93 (d, J = 6.0, 3H), 0.96 (d, J = 6.0, 3H), 1.39 (t, J = 12.0, 1 H), 1.85-1.65 (m, 3H), 2.08-2.18 (m and s, 6H), 2.90-2.97 (m, 1H), 3.00-3.06 (m, 2H), 3.35 (dd, J = 14.2, 8.5. 1H), 4.65 (d, J = 8.7, 1H)1 6.3 (s, 1H), 7.42 (d, J = 8.8, 2H), 7.6 (d, J = 8.8, 2H). mp 230-233 0C (EtOH).
Step C. Preparation of (3S)-3-{[4-(acetyl-tert-butoxycarbonyl-amino)- benzenesulfonyl]-isobutyl-amino}-2-oxo-azepane-1-carboxylic acid tert-butyl ester (Boc activation) (V/)
4.2 g of Λ/α-isobutyl-Λ/α-(4-acetamidobenzenesulfonyl)-L-α-aminor-caprolactam (V) was suspended in 30 mL MeCN and briefly sonicated to break up any large chunks. To this white suspension was added 6.7 g (3 eq.) of di-tert-butyl pyrocarbonate in 10 mL MeCN. The suspension was stirred with a magnetic bar and a 120 mg portion of DMAP was added. The solution becomes a clear light yellow after a few minutes. TLC (EtOAc) reveals 1 product Rf 0.9 (starting material Rf at 0.4). The solution is poured in distilled water 20 mL and extracted with ether, dried with Na2SO4 and evaporated yielding 6.90 g. A sample was recrystallized from hexanes.
1H NMR (DMSO-Cf6): . 0.68 (d, J = 6.0, 3H), 0.85 (d, J = 6.0, 3H), 1.39 (s, 10H), 1.47 (s, 9H)1 1.85-1.65 (m. 3H). 2.15 (S1 3H)1 2.80 (q. J = 4, 1H)1 3.10-3.36 (m, 2H)1 4.01 (d, J = 8.0, 1 H), 4.85 (d, J = 8.7, 1 H)1 7.32 (d, J = 8.8, 2H)1 7.87 (d, J = 8.8, 2H). mp 123-124 0C
Step D. Preparation of (1S)-4-amino-/V-(5-amino-1-hydroxymethyl-pentyl)-Λ/- isobutyl-benzenesulfonamide (V7/-de protected) (reductive ring opening and deprotection) A 3.0 g portion of (SSJ-S-IH-tøcetyl-tert-butoxycarbonyl-aminoJ-benzenesulfonyll-isobutyl- amino}-2-oxo-azepane-1-carboxylic acid tert-butyl ester (W1 step C) is dissolved in 40 mL EtOH followed by 750 mg NaBH4. Brief heating with a heat gun gives a clear solution. TLC reveals one streaky spot after 20 min (EtOAc). The solution is concentrated to a paste, poured in 40 mL 1 N NaOH and extracted with ethyl acetate, the organic phase dried with NaSO4 and evaporated to give 2.8 g of product intermediate (W/); (1S)-{4-[(5-tert- butoxycarbonylamino-i-hydroxymethylφentyO-isobutyl-sulfamoylJ-phenyty-carbamic acid tert-butyl ester (W/).
The above product intermediate is dissolved in 5 mL EtOH and 5 mL 12 N HCI is added. Vigorous gas evolution is observed for a few minutes. After 2 h the solution is evaporated and rendered basic with concentrated KOH and extracted with EtOAc yielding 1.75 g of a white powder.
1H NMR (DMSO-cfe): . 0.82 (m, 6H), 0.97-1.12 (m, 2H)1 1.15-1.30 (m, 3H), 1.57 (m, 1H)1 1.84 (m, 1H)1 2.40 (t, J = 7.8, 2H)1 2.75 (m, 1H), 2.85 (m, 1H)1 3.21 (m, 1H)1 3.44 (d, J = 6.4, 2H), 5.92 (br s. 2H), 6.59 (d. J = 8.0, 2H), 7.39 (d, J = 8.0, 2H).
Step E. Preparation (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid
To a solution of L-diphenylalanine (241 mg, 1.0 mmol) (Peptech Corp.) in 5 mL 1N NaOH and 0.5 mL saturated Na2CO3 (resulting solution at pH 10) was added methoxycarbonyloxysuccinimide (carbonic acid 2,5-dioxo-pyrrolidin-i-yl ester methyl ester) (180 mg, 1.1 mmol) dissolved in 5 mL. Afterwards, the reaction mixture was stirred at room temperature for 2 h. The alkaline solution was extracted once with ether (10 mL) and the aqueous phase was acidified with 1N HCI. This was extracted twice with 20 mL EtOAc1 and the combined organic phases were washed with 50 mL 1 N HCI. The organic phase was dried over Na2SO4. filtered and evaporated to an oil, which solidifies to yields 250 mg (83%) of the desired material. This derivative was used as such in the next step.
Step F. Preparation of (1S,5S)-(1-{5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- hydroxy-hexylcarbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-100)
The title compound was prepared from (1 S)-4-amino-Λ/-(5-amino-1-hydroxymethyl-pentyl)- Λ/-isobutyl-benzenesulfonamide (W/-deprotected) (step D) and (2S)-2- methoxycarbonylamino-S^-diphenyl-propionic acid (step E) using the coupling procedure with HOBt and EDAC described in example 3 (step D). The final product was obtained in 67% yield (121 mg).
LC-MS : 625.3 (M+H)*, 95% pure
1H NMR (CD3OD): δ 0.71-0.85 (m, 2H), 0.88 (d, J = 6.3, 5H), 0.91-0.96 (m, 2H), 1.29- 1.34 (m, 1H), 1.41-1.52 (m, 1H) 1.82-1.92 (m, 1H)1 2.61-2.68 (m, 1H), 2.81-2.85 (m, 2H)1 2.94-3.05 (m, 2H), 3.38-3.40 (t, J = 5.0, 1H)1 3.50-3.51 (m, 1H), 3.52 (s, 3H), 4.28 (d, J = 11.0 1H), 4.87 (d, J = 11.0, 1H), 6.69 (d, J = 8.0, 2H), 7.15-718 (m, 2H), 7.20-7.31 (m, 6H)1 7.33 (d, J = 7.9, 2H), 7.47 (d, J = 7.5, 1H).
13C NMR (CD3OD): δ 20.0, 20.1, 23.3, 25.4, 28.1 , 28.5, 28.9, 38.1 , 40.0, 51.2, 51.6, 53.1 , 57.2. 57.4, 59.5, 61.9, 62.4, 112.6, 125.7, 126.2, 126.3, 127.9, 128.1 ,128.15, 128.2, 128.4, 128.7, 141.3, 141.9, 152.4, 155.9, 169.9, 172.5.
Step G. Preparation of (1S,5S)-{1-[5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6-
(diethoxy-phosphoryloxy)-hexylcarbamoyl]-2,2-diphenyl-ethyl}-carbamic acid methyl ester
The PL-100 compound (product of step F, 203 mg, 0.325 mmol) was dissolved in dry tetrahydrofuran (3 ml.) and 0.2 mL triethylphosphate under N2 atmosphere. The mixture was stirred at this temperature for 15 min, followed by the addition of diethyl chlorophosphate (0.061 mL, 0.423 mmol). Sodium hydride (60% in mineral oil) (17 mg,
0.423 mmol) was added at 0 0C. The solution was stirred for 1 h at 0 0C and 12 h at room temperature. 20 mL of Amberlite XAD-2 was added to the solution and the beads were thoroughly mixed with the solvent. To the mixture was added ice water 2 mL, and the THF evaporated off. The beads were then washed with distilled water 6 times 100 mL then extracted three times with ethyl acetate (30 mL). The combined phase was evaporated and the residue was dried under high vacuum. The crude product was purified by flash chromatography using ethyl acetate/hexane (8/2), then EtOAc 100% as eluent. The yield of this reaction is 152 mg 61%.
LC-MS: 761.2 (M+H)+, 90% pure
1H NMR (CD3OD): <5 0.68-0.75 (m, 1H), 0.75-0.84 (m, 1H), 0.84-1.10 (m, 9H), 1.21-1.50 (m, 8H), 1.88 (m. 1H), 2.58-2.71 (m, 1 H)1 2.80-2.89 (m, 1H)1 2.89-3.08 (m, 2H), 3.49-3.60
(S1 3H)1 3.65-3.74 (m, 1H)1 3.85-3.95 (m, 1H), 3.97-4.02 (m. 1H), 4.07-4.21 (m, 4H), 4.29 (d, J = 10.8, 1H), 6.71 (d, J = 8.0, 2H). 7.10-7.20 (m, 2H)1 7.20-7.32 (m, 5H), 7.32-7.45 (m, 3H), 7.50 (d, J = 7.5. 2H), 7.86 (br s, 1 H).
31P NMR (CD3OD): δ 1.62
Step H. Preparation of (1 S,5S)-(1-{5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- phosphonooxy-hexylcarbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-461)
The product of step G prepared above (152 mg) was dissolved in anhydrous dichloromethane (3.0 ml_). Trimethylsilyl bromide (0.5 ml_) was added at 0 0C. The mixture was stirred during 1h at this temperature and overnight at room temperature. The solvent was evaporated and 0.2 ml_ water was added to the residue. 3 ml_ EtOH was added mixed and evaporated. This step was repeated three times and the residue dried in vacuo. Yields 98 mg 70% of the title derivatives of this first example.
LC-MS: 705.2 (M+H)+, 95% pure
1H NMR (CD3OD): δ 0.65-0.73 (m, 1H), 0.75-0.83 (m, 1H), 0.89 (d, J = 5.6, 8H), 1.27- 1.38, (m, 1H). 1.42-4.55 (m, 1H), 1.82-1.94 (m, 1H), 2.57-2.68 (m, 1H), 2.78-2.90 (m, 1H), 2.91-3.09 (m, 2H), 3.54 (s, 3H), 3.60-3.72 (m, 1 H), 3.87-4.05 (m, 1H), 4.00 (m, 1H)1 4.29 (d, J = 11.3. 1H), 4.90 (d, J = 11.4, 1H)1 6.73 (d, J = 8.0, 2H), 7.13-7.22 (m, 2H), 7.22-7.33 (m, 6H), 7.33-7.45 (m, 2H), 7.51 (d, J = 7.5, 2H).
31P NMR (CD3OD): δ 2.80
Example 2. Preparation of (1 S,5S)-(1-{5-[(4-amino-benzenesulfonyl)-isobutyl- amino]-6-phosphonooxy-hexylcarbamoyJ}-2,2-diphenyl-ethyl)- carbamic acid methyl ester sodium salt (PL-462)
70.7 mg of the final product of example 1 is added to 1 ml_ 0.1 N NaOH and diluted with 1 mL of distilled water. The Solution is then frozen and lyophilized. Yields 67.2 mg (92%) of the desired material with 95% purity.
1H NMR (CD3OD): δ 0.72-0.83 (m, 1H), 0.90 (d, J = 5.8, 9H), 1.26-1.38 (m, 1H), 1.53- 1.65 (m, 1H), 1.88-2.00 (m, 1 H), 2.60-2.70 (m, 1H), 2.79-2.89 (m, 1 H), 2.98-3.00 (m, 1H),
3.00-3.08 (m, 1H), 3.54 (s. 3H), 3.58-3.71 (m, 1H)1 3.72-3.83 (m, 1 H)1 3.84-3.95 (m. 1H). 4.28 (d, J = 11.1, 1H), 4.91 (d, J = 11.0, 1H)1 6.70 (d, J = 7.6, 2H), 7.12-7.22 (m, 2H), 7.22-7.32 (m, 6H), 7.33-7.40 (m, 2H), 7.50 (d, J = 7.7. 2H).
31P NMR (CD3OD): δ 3.13
Example 3. Preparation of (1S,5S)-(1-{5-[(4-amino-benzenesulfonyl)-isobutyl- amino]-6-phosphonooxy-hexylcarbamoyl}-2-naphthalen-2-yl-ethyl)- carbamic acid methyl ester (PL-507)
The preparation of the title compound is based on scheme 2 of this invention.
Step A. Preparation of (1S)-(4-{[5-tert-butoxycarbonylamino-1-
(diethoxyphosphoryloxymethyl)-pentyl]-isobutyl-sulfamoyl}-phenyl)-carbamic acid tert- butyl ester (VIII)
2.00 g (3.7 mmol) (1 S)-{4-[(5-tert-butoxycarbonylamino-1-hydroxymethyl-pentyl)-isobutyl- sulfamoyl]-phenyl}-carbamic acid tert-butyl ester (VII) (example 1, step D) is dissolved in 0.63 ml. triethylphosphate and 10 mL THF at 0 0C under inert argon atmosphere. 0.63 mL (4.44 mmol) diethylchlorophosphate is added and then 0.25 g (6.2 mmol), NaH 60% in oil is added in port ion wise. The mixture is allowed to warm to room temperature and left to stir for 2 h (LC-MS showed completion after 1 h). To the solution is added 20 mL of Amberlite XAD-2 resin and the slurry thoroughly mixed and added to 200 mL ice water. After stirring for 15 min. the resin suspension is filtered and the resin washed several times with distilled water (500 mL). The desired product is desorbed from the resin with acetone (5 X 50 mL), EtOAc (5 X 50 mL), the organic phase is then dried over Na2SO4. After evaporation of the solvent 2.66 g (89%) of clear oil is obtained. The crude product contains a fraction with two diethyl phosphates and is used as is in the next step.
1H NMR (CD3OD): δ 0.91 (d, J = 6.3, 6H), 1.11-1.21 (m, 2H), 1.33 (t, J = 6.9, 10H), 1.43 (s, 9H), 1.53 (S, 10H), 1.90-1.97 (m, 1H), 2.88-2.96 (m, 3H), 2.96-3.04 (m, 1H), 3.81-3.90 (m, 1H), 3.91-3.99 (m, 1H), 4.01-4.14 (m, 4H), 7.61 (d, J = 8.3, 2H), 7.72 (d, J = 8.4, 2H).
31P NMR (CD3OD): δ 1.59
Step B. Preparation of (2S)-phosphoric acid 6-amino-2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-hexyl ester diethyl ester (IX) The crude product obtained in the previous step (WW1 2.66 g) is dissolved in 12 ml_ EtOH. 4 mL of HCI cone- is added and heated briefly to 70 0C then left at room temperature for 3h. The solvent is evacuated and the residue triturated with 50 m L ether. The thick residue is S then dissolved in 3 mL ice water and the pH adjusted to 12 with 50% NaOH. The thick slurry obtained is extracted with EtOAc (3 X 50 mL) and the organic phase dried over Na2SO4. After filtration of the drying agent the organic phase is evacuated to yield 1.84 g (98%) of the desired product (/X). 0 LC-MS: 480.2 (M+H)*, 95% pure.
1H NMR (CD3OD): δ 0.91 (s, 6H), 1.11-1.26 (m, 3H), 1.28-1.43 (m, 8H), 1.45-1.51 (m, 1H), 1.52-1.61 (m, 1H), 1.89-1.96 (m, 1H), 2.56 (t, J = 6.7, 2H), 2.85-2.91 (m, 1H), 2.98- 3.11 (m. 1H), 3.79-3.99 (m, 1H), 3.94 (d, J = 5.3, 1H)1 4.09-4.11 (m, 4H), 6.69 (d, J = 7.9,5 2H). 7.50 (d, J = 7.9, 2H).
31 P NMR (CD3OD): 5 1.61
Step C. Preparation of (2S)-2-methoxycarbonylamino-3-naphthalen-2-yl-0 propionic acid (or L-Moc-2-naphthylalanine)
To a solution of L-2-naphthylalanine (215 mg, 1 mmol) (Peptech Corp.) in 5 mL 1 N NaOH and 0.5 mL saturated Na2CO3 (resulting solution at pH 10) was added methoxycarbonyloxysuccinimide (187 mg, 1.1 mmol) dissolved in 5 mL. Afterwards, the5 reaction mixture was stirred at room temperature for 2 h. The alkaline solution was extracted once with ether (10 mL) and the aqueous phase was acidified with 1N HCI. This was extracted twice with 20 mL EtOAc1 and the combined organic phases were washed with 50 mL 1N HCI. The organic phase was dried over Na2SO4. filtered and evaporated to an oil, which solidifies to yields 200 mg (73%) of the desired material. This intermediate0 (referred as the Λ/-substituted amino acid) was used without further purification in the next step.
Step D. Preparation of (1 S,5S)-(1 -{5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- phosphonooxy-hexylcarbamoyl^-naphthalen^-yl-ethyO-carbamic acid methyl ester (PL-5 507) 100 mg L-Moc-2-naphthylalanine (step C) was activated with 100 mg EDAC and 57 mg HOBt in 1.5 mL DMF for 30 minutes. Then, 100 mg of phosphoric acid 6-amino-2-[(4- amino-benzenesulfonyl)-isobutyl-amino]-hexyl ester diethyl ester (step B) was added and left to stir at room temperature for 1 h. 40 mL of 1M K2CO3 was added to the DMF solution and left for 10 min. 50 mL of EtOAc was then added and the mixture was then agitated vigorously. Separation of the EtOAc phase was effected, followed by extraction with 5% citric acid (50 mL) once, then water (50 mL) 3 times and finally brine. The organic phase was the separated and evaporated. The residue was taken up in 50 mL DCM and re- evaporated. The residue was again taken up in 50 mL DCM and 0.5 mL of TMSBr was added. The solution was left overnight (16 h). The DCM was evacuated and a solution of ice cold MeOH: Water 1:1 was added, stirred briefly and evacuated. The residue was triturated with ether then dissolved in 1N NaOH. The clear solution was extracted with ether and the aqueous phase acidified with 6N HCI. The white precipitated was then collected by filtration and dried in vacuo overnight. Yields 88 mg of the title compound.
LC-MS: 679.8 (M+H)\ 95% pure.
1H NMR (CD3OD): δ 0.89-0.98 (m, 8H), 1.15 (m, 2H). 1.35 (m, 1H), 1.45 (m, 1H)1 1.88 (m, 1H), 2.84 (m, 2H), 2.98 (m, 1H), 3.01 (m, 2H), 3.24 (m, 1H), 3.56. (s, 3H), 3.60 (m, 1 H), 3.81 (m, 1H), 3.99 (m, 1 H), 4.39 (t, J = 6.8, 1H), 6.91 (d, J = 8.0, 2H), 7.34 (d, J= 8.0, 1H), 7.45 (m. 2H). 7.58 (d. J = 8.0, 2H). 7.66 (s, 1H)1 7.70-7.82 (m, 3H).
31P NMR (CD3OD): δ 2.56
Example 4. Preparation of (2S.2S) phosphoric acid mono-(2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-{2-[(morpholine-4-carbonyl)- amino]-3-naphthalen-1-yl-propionylamino}-hexyl) ester (PL-498)
Step A. Preparation of (2S)-2-[(morpholine-4-carbonyl)-amino]-3-naphthalen-1-yl- propionic acid
To a solution of L-1-naphthylalanine (215 mg, 1 mmol) (Peptech Corp.) in 5 mL 1N NaOH and 0.5 mL saturated Na2CO3 (resulting solution at pH 10) was added morpholine-4- carbonyl chloride (150 mg, 1.0 mmol) dissolved in 5 mL. Afterwards, the reaction mixture was stirred at room temperature for 2 h. The alkaline solution was extracted once with ether (10 mL) and the aqueous phase was acidified with 1N HCI. This was extracted twice with 20 mL EtOAc, and the combined organic phases were washed with 50 mL 1N HCI. The organic phase was dried over Na2SO4, filtered and evaporated to an oil, which solidifies to yields 125 mg (38%) of the desired material. This compound was used as S such in the next step.
Step B. Preparation of (2S.2S) Phosphoric acid mono-(2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-{2-[(morpholine-4-carbonyl)-amino]-3-naphthalen-1-yl- propionylamino}-hexyl) ester (PL-498) 0
This compound was made as for the preparation of the product of example 3 (step D) with 100 mg of (2S)-2-[(moφholine-4-carbonyl)-amino]-3-naphthalen-1-yl-propionic acid (step A of this example). The resulting precipitated residue was further purified by reverse phase preparative HPLC. Yields 41 mg of the final compound. 5
LC-MS: 734.8 (M+H)+, 95% pure.
1H NMR (CD3OD): δ 0.83-0.98 (m, 8H), 1.00-1.25 (m, 4H), 1.45-1.52 (m, 1H), 1.52-1.66 (m, 1H), 1.88-1.99 (m, 1H), 2.77-2.92 (m, 2H), 2.98-3.16 (m, 3H), 3.40-3.49 (m, 1H), 3.50-0 3.56 (m, 6H), 3.67-3.69 (m, 1H), 3.81-3.89 (m, 1H), 3.99-4.05 (m, 1H), 4.59 (t, J = 6.0, 1H)1 6.75 (d, J = 8.0, 2H), 7.30-7.60 (m, 7H), 7.75 (d, J = 8.0, 1H), 7.90 (d, J= 7.8. I H), 8.23 (d. J=7.8 2H).
31P NMR (CD3OD): δ 2.1 Λ 5
Example 5. Preparation of (2S,2S)-phosphoric acid mono-{6-(2-acetylamino-3,3- diphenyl-propionylamino)-2-[(4-amino-benzenesulfonyl)-isobutyl- amino]-hexyl> ester (PL-504) 0
Step A. Preparation (2S)-2-acetylamino-3,3-diphenyl-propionic acid
To a solution of L-diphenylalanine (100 mg, 0.4 mmol) (Peptech Corp.) in 5 mL 1 N NaOH and 0.5 mL saturated Na2CO3 (resulting solution at pH 10) was added acetyl chloride (0.55 mmol) dissolved in 5 mL. Afterwards, the reaction mixture was stirred at room temperature for 2 h. The alkaline solution was extracted once with ether (10 mL) and the aqueous phase was acidified with 1N HCI. This was extracted twice with 20 mL EtOAc, and the combined organic phases were washed with 50 mL 1N HCI. The organic phase was dried over Na2SO4. filtered and evaporated to an oil, which solidifies to yields 70 mg (60%) of the desired material. This crude intermediate was used as such in the next step.
Step B. Preparation of (2S,2S)-phosphoric acid mono-{6-(2-acetylamino-
3,3-diphenyl-propionylamino)-2-[(4-amino-benzenesulfonyl)-isobutyl-amino]-hexyl} ester (PL-504)
This compound was made as for the preparation of the product of example 3 (step D) with 100 mg of (2S)-2-acetylaminc~3,3-diphenyl-propionic acid (this example step A). The final product was obtained in 30% yield (30 mg).
LC-MS: 689.3 (M+H)+, 95% pure.
1H NMR (CD3OD): δ 0.77-1.04 (m, 9H), 1.10-1.17 (m, 1H), 1.23-1.49 (m, 1H), 1.46-1.57 (m, 1H), 1.78 (S, 3H)1 1.88-1.99 (m, 1H), 2.80-2.92 (m, 2H), 2.92-3.08 (m, 2H), 3.63-3.75 (m, 1H), 3.79-3.95 (m, 1H), 4.00 (m, 1H)1 4.34 (d. J = 11.3, 1H). 5.19-5.28 (m, 1H), 6.77- 6.85 (m, 2H), 7.10-7.20 (m, 2H), 7.27-7.33 (m, 6H), 7.32-7.41 (m, 2H)1 7.49-7.62 (m, 2H).
31P NMR (CD3OD): δ 2.70
Example 6. Preparation of (1 S,5S)-(1 -{5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-amino]-6-phosphonooxy-hexylcarbamoyl}-2,2-diphenyl- ethyl)-carbamic acid methyl ester (PL-515)
First methodology: The preparation of the title compound is based on scheme 3 of this invention.
Step A. Preparation of (1-{5-[(4-amino-3-fluoro-benzenesulfonyl)-isobutyl-amino]-6- hydroxy-hexylcarbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (X) (PL-337)
The product of example 1, step F (0.624 g, 1 mmol) is dissolved in 5 mL MeCN at 24 0C. SelectFluor 0.35 g (1 mmol) is added in one portion and stirred for 1 h. 1 mL of water is added and the solution was injected directly into a preparative reverse-phase HPLC. The product was collected and lyophilized to give 250 mg (38%) yield of a white solid. LC-MS: 643.3 (M+H)\ 99% pure.
1H NMR (MeOD): δ" 0.71-0.85 (m 2H), 0.88 (d, J = 6.3, 6H)1 0.91-0.96 (m, 2H), 1.21-1.29 (m, 1H), 1.41-1.52 (m, 1H) 1.82-1.92 (m, 1H), 2.61-2.68 (m, 1H), 2.81-2.85 (m, 2H), 2.94- 3.05 (m, 2H), 3.38-3.40 (t, J = 5. 1H), 3.49-3.52 (m, 5H), 4.28 (d, J = 10, 1H), 4.87 (d, J = 10, 1H) 6.90 (t, J = 8.3. 1H), 7.20 (m, 2H), 7.28 (m, 3H), 7.33 (m, 3H), 7.39 (mt 4H).
Step B. Preparation of (1S,5S)-{1-[5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-amino]-6-(diethoxy-phosphoryloxy)-hexylcarbamoyl]-2,2-diphenyl-ethyl}-carbamic acid methyl ester
The product of step A was phosphorylated with chlorodiethylphosphate following the procedure described in example 1, step G. Yields 157 mg, 68%.
LC-MS: 779.3 (M+H)\ 95% pure.
1H NMR (CD3OD): δ 0.82 (m, 1H)1 0.92 (d, J = 6.2, 8H), 0.96 (m, 3H). 1.36 (d, J = 3.7, 6H), 1.90 (m, 1H). 2.69 (m, 1H), 2.89 (iti, 1H), 2.98 (m, 2H), 3.56 (s, 3H), 3.74 (m, 1H)1 3.93 (m, 1H)1 4.03 (m, 1H)1 4.12 (q, J = 7.5 and 14.8. 4H). 4.32 (d. J = 11.4, 1H). 4.92 (d, J = 11.4, 1H), 6.90 (t, J = 8.3. 1H). 7.20 (m. 2H), 7.28 (m, 3H). 7.33 (m, 3H), 7.39 (m. 4H).
31P NMR (CD3OD): δ 1.65
Step C. Preparation of (1S,5S)-(1-{5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol-β-phosphonooxy-hexylcarbamoyl^.∑-diphenyl-ethylj-carbamic acid methyl ester (XQ (PL-515)
Deprotection was effected using the procedure described in example 1 , step G. Yields 101 mg.
LC-MS: 723.2 (M+H)\ 95% pure.
1H NMR (CD3OD): δ 0.65-0.77 (m. 1H). 0.77-0.85 (m, 1H), 0.85-1.05 (m, 9H), 1.25-1.39 (m, 1 H), 1.40-1.52 (m, 1H), 1.82-1.98 (m, 1H), 2.58-2.72 (m, 1 H)1 2.82-2.92 (m, 1H). 2.92-
3.05 (m, 2H)1 3.54 (s, 3H)1 3.64-3.75 (m, 1H)1 3.80-3.92 (m, 1H), 3.91-4.04 (m, 1H), 4.29 (d, J = 11.4, 1 H)1 7.19 (t, J = 6.6, 1H), 7.13-7.21 (m, 2H), 7.22-7.33 (m, 6H), 7.34-7.38 (m, 2H), 7.39-7.48 (m, 2H).
31P NMR (CD3OD): δ 2.74
Second methodology: The preparation of the title compound is based on scheme 4 of this invention.
Step A. Preparation (1S,5S)-(1-{5-[(4-amino-benzenesulfonyI)-isobutyl-amino]-6- phosphonooxy-hexylwrbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (PL-461)
(2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid ((example 1 , step E) 0.9 g, 3 mmol) was activated in DMF (5 ml_) with EDAC (1.7 g, 9 mmol) and HOBt (1.2 g, 9 mmol). To the solution was added 1.17 g of (2S)-phosphoric acid 6-amino-2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-hexyl ester diethyl ester (OQ (example 3, step B) and the mixture stirred for 3 h. 20 g of Amberlite XAD-2 resin was then added and the beads were left to soak for 10 min. The resin was transferred into a glass filter and washed thoroughly with distilled water (400 ml_) and 200 mL of 1M NaHCO3. The beads were then washed with 4 X 50 ml portions of MeOH then EtOAc 200 ml_. The organic phase was evaporated. The residue was adsorbed onto silica gel and passed through a short silica gel column (EtOAc) to yield 2.4 g (83%) of white solid after evaporation.
NMR identical as in example 1 , step H.
Step B. Preparation (1 S,5S)-{1-[5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol-β-tdiethoxy-phosphoryloxyj-hexylcarbamoyll^^-diphenyl-ethyl^carbamic acid methyl ester (XII)
The product of step A above, (1 S,5S)-(1-{5-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6- phosphonooxy hexylcarbamoyl}-2,2-diphenyl-ethyl)-carbamic acid methyl ester (0.555 g, 0.73 mmol) was dissolved in 5 ml_ MeCN. Selectfluor (0.26 g, 0.7 mmol) was added and the mixture stirred for 30 min. The mixture was purified by reverse phase preparative HPLC and lyophilized to yield 278 mg (48% yield) white solid.
1H NMR identical as previous entry, see first methodology above. Step C. Preparation (1 S,5S)-(1-{5-[(4-amino-3-fluoro-benzenesulfonyl)- isobutyl-aminol-β-phosphonooxy-hexylcarbamoyl^^-diphenyl-ethyO-carbamic acid methyl ester (XIlI, in this specific case is compound Xl) (PL-515)
The procedure make this derivative was as described in the deprotection step for the methodology above. Yields 139 mg 70% after reverse phase HPLC.
1H NMR identical as previous entry, see first methodology above.
Example 7. Preparation of (2S,2S)-acetic acid 2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-6-{2-methoxycarbonylamino-3,3-diphenyl- propionylamino)-hexyl ester (PL-S21)
The preparation of the title derivative is based on scheme 5 of this invention.
Step A. Preparation of (2S)-acetic acid 6-tert-butoxycarbonylamino-2-[(4-tert- butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester {XIV, R1A = CH3)
To a stirred solution of (1 S)-{4-[(5-tert-butoxycarbonylamino-1-hydroxymethyl-pentyl)- isobutyl-sulfamoyl]-phenyl}-carbamic acid tert-butyl ester (intermediate product (VII) of example 1 , step D, 97 mg, 0.18 mmol) in anhydrous CH2CI2 (3 ml_) was added N1N- dimethylaminopyridine (22 mg, 0.18 mmol) and acetic anhydride (0.014 ml_, 0.18 mmol).
The mixture was stirred at room temperature for 1 hour. The solvent was evaporated.
Ethyl acetate (50 ml_) was added and the organic layer was washed with water (30 mL), then dried with Na2SO4 and concentrated. The residue was purified by flash chromatography eluting with ethyl acetate. The yield obtained was quantitative (100 mg).
LC-MS: 586.2 (M+H)+, 95% pure
Step B. Preparation of (2S)-acetic acid 6-amino-2-[(4-amino- benzenesulfoπyl)-isobutyl-amino]-hexyl ester (XV, RiA = CH3)
This derivative was prepared from (2S)-acetic acid 6-tert-butoxycarbonylamino-2-[(4-tert- butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester as described in example 15, step B. The yellow solid (66 mg) was used for the next reaction without purification.
LC-MS: 386.2 (M+H)*, 95% pure
Step C. Preparation of (2S,2S)-acetic acid 2-[(4-amino-benzenesulfonyl-
)isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl-propionylamino)-hexyl ester (XW. R1A = CH3) (PL-521)
This derivative was prepared from (2S)-acetic acid 6-amino-2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-hexyl ester (product of step B) as described in example 15, step B. The final product was purified by flash chromatography with a mixture of eluents hexane/ethyl acetate (2/8). A yellow solid was obtained in 70% yield (70 mg).
LC-MS: 667.3 (M+H)\ 95% pure
1H NMR (acetone-dβ): δ 0.85-0.97 (m, 12H), 1.21-1.41 (m, 2H), 1.88-2.00 (s, 3H), 2.59- 2.69 (m, 1 H), 2.83-2.90 (m, 1H), 2.90-3.01 (m, 1 H), 3.01-3.10 (br s. 1 H), 3.45-3.60 (s, 3H), 3.70-3.80 (m. 1 H), 3.93-4.00 (m, 1 H), 4.00-4.11 (m, 1 H), 4.38-4.45 (d, J = 11.0, 1H), 4.89- 4.98 (t, J = 10.0, 1H), 5.43-5.58 (br s, 1 H), 6.28-6.48 (d, J = 8.9, 1H), 6.72-6.83 (d, J = 8.0, 2H), 6.85-6.93 (br s, 1 H), 7.12-7.22 (t, J = 7.4, 1 H), 7.21-7.31 (d, J = 7.0, 4H), 7.31-7.45 (m, 5H), 7.48-7.57 (d, J = 8.0. 2H).
Example 8. Preparation of (2S,2S)-nicotinic acid 2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-6-(2-rnethoxycarbonylamino-3,3-diphenyl- propionylamino)-hexyl ester (PL-520)
Step A. Preparation of (2S)-nicotinic acid 6-tert-butoxycarbonylamino-2-[(4-tert- butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester (XIV, RiA = 3-pyridyl)
(1 S)-{4-[(5-tert-butoxycarbonylamino-1-hydroxymethyl-pentyl)-isobutyl-sulfamoyl]-phenyl}- carbamic acid tert-butyl ester (intermediate product (VII) of example 1 , step D, 130 mg, 0.24 mmol) was dissolved in anhydrous DMF (1 mL) and treated with 0.066 mL (0.48 mmol) of triethylamine followed by EDC (120 mg, 0.65 mmol), HOBt (88 mg, 0.65 mmol) and nicotinic acid (27 mg, 0.22 mmol). The mixture was stirred overnight at room temperature. The product was extracted with ethyl acetate (40 mL) and water (40 ml_). The organic phase was separated and dried with Na2SO4, then evaporated to give 200 mg of crude product. This compound was purified by flash chromatography with ethyl acetate as the eluent. A clear oil was obtained in 100% yield (150 mg).
LC-MS: 649.3 (M+H)\ 95% pure
1H NMR (acetone-dβ): δ 0.90-1.14 (d, J = 5.9, 6H), 1.31-1.42 (m, 2H), 1.48 (s, 9H), 1.51- 1.55 (m, 2H), 1.59 (s, 9H), 1.62-1.69 (m, 1H), 1.72-1.83 (m, 1 H), 3.00-3.11 (m, 2H). 3.11- 3.17 (m, 1H), 3.19-3.27 (m, 1H), 4.15-4.24 (m, 1H), 4.35-4.44 (t, J = 9.1, 1H), 4.50-4.58 (dd, J = 4.4 and 11.5, 1H), 5.89-5.99 (br s, 1H), 7.53-7.60 (m, 1H), 7.70-7.77 (d, J = 8.2, 2H), 7.80-7.87 (d, J = 8.2, 2H)1 8.24-8.31 (d, J = 7.3, 1H), 8.75-8.82 (m, 1H), 8.82-8.88 (m, 1 H), 9.12-9.18 (br s, 1H).
Step B. Preparation of (2S)-nicotinic acid 6-amino-2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-hexyl ester (XV, RiA = 3-pyridyl)
The product of step A, (2S)-nicotinic acid 6-tert-butoxycarbonylamino-2-[(4-tert- butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester (150 mg, 0.23 mmol), was dissolved in CH2CI2 (5 mL) and trifluoroacetic acid (1 mL) was added. The mixture was stirred during 2 hours at room temperature. The solvent was evaporated and the residue was extracted with ethyl acetate (40 mL) and NaOH 1M (40 mL) (pH = 10). The organic portion was separated, dried with Na2SO4 and evaporated. The residue (100 mg) was used for the next reaction without further purification. The yield was quantitative.
LC-MS: 449.2 (M+H)*. 95% pure
Step C. Preparation of (2S,2S)-nicotinic acid 2-[(4-amino-benzenesulfonyl)- isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl-propionylamino)-hexyl ester (PL-520)
The product of step B, (2S)-nicotinic acid 6-amino-2-[(4-amino-benzenesulfonyl)-isobutyl- amino]-hexyl ester (100 mg, 0.22 mmol) was dissolved in anhydrous DMF (2 mL) and treated with 0.062 mL (0.45 mmol) of triethylamine followed by EDC (100 mg, 0.56 mmol), HOBt (75 mg, 0.56 mmol) and (2S)-2-methoxycarbonylamino-3,3-diphenyl-propionic acid (56 mg, 0.19 mmol). The mixture was stirred overnight at room temperature. The product was extracted with ethyl acetate (40 mL) and water (40 mL). The organic layer was separated and dried with Na2SO4, then evaporated to give 160 mg of crude oil. The residue was purified by flash chromatography with a mixture of eluents hexane/ethyl S acetate (2/8). The title compound was obtained as a clear oil in 20% yield (25 mg).
LC-MS: 730.2 (M+H)\ 95% pure
1H NMR (acetone-dβ): δ 0.80-0.97 (m, 9H), 0.97-1.13 (m, 2H), 1.26-1.40 (m, 1H), 1.40-0 1.57 (m, 1H), 2.61-2.73 (m, 1H), 2.86-2.98 (m, 2H), 3.00-3.17 (m, 2H). 3.45-3.59 (s, 3H). 3.91-4.00 (m, 1H), 4.24-4.34 (m. 1H), 4.34-4.47 (m. 2H). 4.90-4.99 (t, J = 9.7, 1 H), 6.35- 6.44 (m, 1H), 6.68-6.79 (d, J = 7.9, 1H)1 6.91-7.00 (br s, 1H), 7.13-7.22 (m, 2H), 7.22-7.31 (m. 3H). 7.35-7.48 (m. 4H), 7.49-7.64 (m, 2H). 7.75-7.84 (m, 1H), 8.25-8.36 (m, 1H), 8.76- 8.88 (br s, 1 H), 9.12-9.26 (br s, 1 H). 5
Example 9. Preparation of (2S,2S)-dimethylamino-acetic acid 2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3- diphenyl-propionylamino)-hexyl ester (PL-534) 0 Step A. Preparation of (2S)-dimethylamino-acetic acid 6-tert-butoxycarbonylamino-
2-[(4-tert-butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]-hexyl ester (XIV, R1A = (CH3J2NCH2-)
This title compound was obtained from (1 S)-{4-[(5-tert-butoxycarbonylamino-1-5 hydroxymethyl-pentylj-isobutyl-sulfamoylj-phenylj-carbamic acid tert-butyl ester (intermediate product (VW) of example 1, step D) as described example 15, step A using Λ/,Λ/-dimethylglycine. The clear oil was obtained in 100% yield (150 mg).
LC-MS: 629.3 (M+H)\ 95% pure 0
1H NMR (acetone-dβ): δ 0.81-0.95 (d, J = 6.1, 6H)1 1.18-1.30 (m, 2H), 1.32-1.43 (s, 9H), 1.43-1.52 (s, 8H), 1.52-1.62 (m, 1H)1 1.93-2.00 (m, 1H), 2.19-2.29 (s, 4H), 2.69-2.80 (m, 4H)1 2.90-3.05 (m, 6H)1 3.60-3.65 (m. 1H), 3.85-3.97 (m, 1H), 3.98-4.08 (m, 1H), 4.08- 4.14 (m, 1H)1 5.78-5.88 (m. 1 H)1 7.68-7.80 (m, 3H), 8.80-8.88 (br s, 1H). 5 Step B. Preparation of (2S)-dimethylamino-acetic acid 6-amino-2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-hexyl ester (XV1 R1A = (CH3^NCH2-)
The title derivative was prepared from (2S)-dimethylamino-acetic acid 6-tert- butoxycarbonylamino^-^-tert-butoxycarbonylamino-benzenesulfonyO-isobutyl-amino]- hexyl ester as described in example 15, step B. The final product (100 mg) was used as such in the next step.
LC-MS: 429.3 (M+H)*, 90% pure
Step C. Preparation of (2S,2S)-dimethylamino-acetic acid 2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl- propionylamino)-hexyl ester (PL-534)
This title compound was prepared from (2S)-dimethylamino-acetic acid 6-amino-2-l(4- amino-benzeπesulfonyl)-isobutyl-amino]-hexyl ester as described in example 15, step C. The crude product was purified by LC-preparative. The final compound was obtained in 10% yield (10 mg).
LC-MS: 710.3 (M+H)+, 92% pure
1H NMR (acetone-dβ): δ 0.81-0.98 (m, 12H), 1.14-1.30 (m, 2H), 1.31-1.45 (m, 1H), 2.58- 2.77 (m, 2H), 2.79-2.90 (m, 2H)1 3.42-3.56 (s, 3H)1 3.75-3.85 (m, 1H), 3.99-4.17 (m, 3H)1 4.23-4.35 (m, 1H). 4.36-4.45 (m, 1H), 4.86-4.96 (m, 1H). 6.33-6.42 (m, 1H), 6.74-6.83 (m. 1H)1 6.85-6.90 (m, 1 H)1 7.12-7.22 (m, 3H), 7.23-7.31 (m, 4H), 7.31-7.44 (m, 5H)1 7.47- 7.55 (m, 1H), 7.73-7.80 (m, 1H).
Example 10. Preparation of (2S,2S)-2-amino-3-methyl-butyric acid 2-[(4-amiπo- benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3- diphenyl-propionylamino)-hexyl ester (PL-530)
Step A. Preparation of (2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid 6-tert- butoxycarbonylamino-2-[(4-tert-butoxycarbonylamino-benzenesulfonyl)-isobutyl-amino]- hexyl ester (XIV, R1A = (CH3)2CHCH(NH2)-) This title compound was obtained from (1S)-{4-[(5-tert-butoxycarbonylamino-1- hydroxymethyl-pentyl)-isobutyl-sulfamoyl)-phenyl}-carbamic acid tert-butyl ester (intermediate product (VW) of example 1 , step D) as described in example 15, step A using (2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid. The crude product was purified by S flash chromatography eluting with a mixture of hexane/ethyi acetate (1/1). The yield obtained was 100% (150 mg).
LC-MS: 777.3 (M+H)\ 95% pure 0 1H NMR (acetone-dβ): δ 0.80-1.00 (m, 14), 1.13-1.28 (s, 2H), 1.30-1.44 (s, 11H), 1.45- 1.56 (S1 10), 1.58-1.67 (m, 1H), 2.87-3.04 (m, 4H), 3.84-3.97 (m, 1H), 3.97-4.12 (m, 2H), 4.12-4.21 (m, 1H), 4.99-5.14 (m, 2H), 5.78-5.89 (m, 1 H), 6.38-6.52 (m, 1H), 7.24-7.34 (m, 1H), 7.34-7.41 (m, 2H), 7.65-7.83 (m, 4H), 8.77-8.86 (m, 1H). 5 Step B. Preparation of (2S)-benzyloxycarbonylamino-3-methyl-butyric acid
6-amino-2-[(4-amino-benzenesulfonyl)-isobutyl-amino]-hexyl ester (XV, R1A = (CH3)2CHCH(NH2)-)
This derivative was prepared from (2S)-2-benzyloxycarbonylarnino-3-methyl-butyric acid0 6-tert-butoxycarbonylamino-2-[(4-tert-butoxycarbonylamino-benzenesulfonyl)-isobutyl- amino]-hexyl ester (product of step A) as described in example 15, step B. The final compound was obtained in quantitative yield (110 mg) and used for the next step without purification. 5 LC-MS: 577.3 (M+H)*, 90% pure
Step C. Preparation of (2S,2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid 2-[(4-amino-benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3- diphenyl-propionylamino)-hexyl ester 0
The title compound was obtained from (2S)-benzyloxycarbonylamino-3-methyl-butyric acid 6-amino-2-[(4-amino-benzenesulfonyl)-isobutyl-amino]-hexyl ester (product of step B) as described in example 15, step C. The clear oil was obtained in 86% yield (120 mg). 5 LC-MS: 858.3 (M+H)*, 95% pure Step D. Preparation of (2S,2S)-2-amino-3-methyl-butyric acid 2-[(4-amino- benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3.3-diphenyl- propionylamino)-hexyl ester (PL-530)
To a stirred solution of (2S,2S)-2-benzyloxycarbonylamino-3-methyl-butyric acid 2-[(4- amino-benzenesulfonyl)-isobutyl-amino]-6-(2-methoxycarbonylamino-3,3-diphenyl- propionylamino)-hexyl ester (step C. 120 mg, 0.14 mmol) in anhydrous THF (8 mL), under nitrogen atmosphere, was added palladium 10% wt. on activated carbon (160 mg). The mixture was reacted under hydrogen atmosphere overnight, at room temperature. The solution was filtered and the palladium on carbon was washed with THF (50 mL). The solvent was evaporated and the residue (110 mg) was purified by flash chromatography using ethyl acetate as the eluent. The clear oil was obtained in 47% yield (47 mg).
LC-MS: 796.4 (IvHH)+, 95% pure
1H NMR (acetone-dβ): δ 0.84-0.97 (m, 12H), 0.97-1.08 (m, 2H)1 1.27-1.43 (m, 3H), 1.49- 1.62 (m, 4H), 1.80-1.93 (m, 1H), 1.94-2.00 (m, 1H), 2.36-2.46 (m, 1H), 2.58-2.74 (m, 2H), 2.86-2.96 (m, 3H), 2.99-3.10 (m, 2H), 3.46-3.52 (s, 3H). 3.52-3.60 (m, 2H), 3.75-3.87 (m, 2H)1 3.95-4.04 (m, 1H), 4.10-4.18 (m, 1H), 4.37-4.44 (m, 1H), 4.89-4.97 (m, 1H), 5.40- 5.48 (m, 1H)1 6.30-6.40 (m, 1H), 6.76-6.83 (d, J = 8.2, 1 H), 6.87-7.03 (m, 2H)1 7.14-7.22 (m, 1H), 7.23-7.34 (m, 3H)1 7.35-7.45 (m, 4H)1 7.50-7.56 (m. 1H), 7.57-7.65 (m, 1H).
Example 11. In vitro inhibition of CYP450
Human Liver Microsomes
Pooled human liver microsomes (from 15 donors) were purchased from In vitro Technologies, Inc., Baltimore, Maryland, USA.
PL-100 and Ritonavir solutions preparation
Stock solution of PL-100 was prepared in methanol at 25 mM. For the CYP3A4 assay, the 25 mM stock solution was diluted with methanol to 0.25 and 0.125 mM. The 25 mM stock solution and the 0.25 and 0.125 mM solutions were diluted 250 fold for microsomal incubation to a final concentration of 200, 500 and 1000 nM. A sub-stock solution of PL- 100 was prepared in methanol at 25μM and diluted in methanol to 2.5 and 0.25μM and further diluted 250 fold for microsomal incubation giving final concentrations of 100, 10 and 1nM.
Stock solution of Ritonavir was prepared in methanol at 25 mM. A sub-stock solution was prepared in methanol at 25 μM. This sub-stock solution was diluted in methanol to 2.5 and 0.25 μM and further diluted 250 fold for microsomal incubation giving final concentrations of 100, 10 and 1nM.
Control incubations were performed in the presence of an equal volume of methanol. The final methanol concentration in incubation mixtures was < 1%. Selective inhibitors of CYP450 were tested in parallel as a positive control.
Testosterone 6β-Hydrolase (TESH) Activity
The TESH activity was determined by HPLC-UV analysis following incubation of microsomes with testosterone as. a substrate. Liver microsomes (at a final concentration of 0.30 mg/ml protein) were incubated for 10 minutes at 37°C with various concentrations of testosterone in 0.5 ml reaction mixture, containing 0.1 M phosphate buffer (pH 7.4), 1 mM EDTA, and 3mM magnesium chloride with a NADPH-generating system, PL-100, Ritonavir or Ketoconazole, at appropriate concentrations, were present in the incubation medium with the probe substrate before the reaction was started by addition of NADPH- generating system. The reaction was terminated with 500 μL ice-cold acetonitrile and the 6β -hydroxytestosterone formation was quantified using 6 point standard curve (range 2 to 20 nmol/mL). The analysis of 3 quality controls (LQC, MQC and HQC) was also performed. The associated enzyme activity was expressed as nmol of 6β- hydroxytestosterone per mg of protein per min. Data were captured with Millenium chromatographic data management and storage system (version 4.0).
Michaelis-Menten Kinetics
EnzFitter software (Biosoft) was used for the calculation of Km, Vmax of probe substrates and apparent Km', Vmax' in the presence of various concentration of PL-100, Ritonavir or selective inhibitors and Ki of PL-100, Ritonavir or selective inhibitors. The cytochrome P450 activities were plotted against substrate concentrations. A nonlinear regression analysis was generated for Michaelis-Menten models in the presence and absence of PL- 100, Ritonavir or selective inhibitors. Lineweaver-Burk Plots
To determine the mode of inhibition, a transformation of fitted 1/V values were employed to generate a linear regression of 1/V vs. 1/[S] using EnzFitter software. The nature of the curves was utilized as an indication of the type of inhibition. 5
Ki was calculated using the equation corresponding to the type of inhibition:
No inhibitor: V= Vmax*[S]/ (Km +[S])
l o Competitive inhibition: V= Vmax*[S] / (Km (1 +[I] / Ki) +[S])
Km'= Km* (1+[I] / Ki )
Non-competitive inhibition: V= Vmax*[S] / (Km (1 +[I] / Ki ) +[S] (1 +[I] / Ki )) = 15
= (Vmax / (1+[I] / Ki )* [S]) / (Km+[S])
Vmax'= Vmax / (1+[l] / Ki)
20 Uncompetitive inhibition: V= Vmax*[S] / (Km+[S](1 +[I] / Ki)) =
= (Vmax / (1+ [I] / Ki) *[S]) / (Km/(1+[l] / Ki) +[S])
Km' = Km / (1+[I] / Ki) 25
Vmax' = Vmax / (1+[I] / Ki)
V : Velocity of the enzyme reaction in the presence of inhibitor
Vmax : Maximal velocity
30 Vmax' : Maximal velocity in the presence of inhibitor
[S] : Substrate concentration
[I] : Inhibitor concentration
Km : dissociation constant of the enzyme-substrate complex
Km' : dissociaton constant of the enzyme-substrate complex in the presence of
35 inhibitor Ki : dissociation constant of the enzyme-inhibitor complex
HPLC methods 6β- hydroxytestosterone
Column : C18, 4μm, 3.9 x 150 mm
Detection: UV@ 254 nm
Mobile phase (gradient) : A: Methanol, Water, Acetonitrile (39:60:1)
B: Methanol, Water, Acetonitrile (80:18:2) Flow rate: 1m Um in.
Data calculation and Statistical analysis
Numerical data obtained during the study were subjected to calculation group mean values, standard deviations, where appropriate, using Excel software (Microsoft). Enzyme kinetic parameters (Km and Vm ax) and the Ki values of test articles and selective inhibitors were calculated by nonlinear regression analysis using EnzFitter software. EnzFitter software was utilized also for the fitting of the Michaelis-Menten curves and for the generation of the Lineweaver-Burk plots, from the fitted values of Michaelis-Menten curves, allowing the identification of the type of inhibition were appropriate.
The testosterone 6β-hydrolase activity, a selective marker for CYP3A4/5, was assessed in the presence of PL-100. With a Ki of 0.445 ± 0.050 μM (R2 = 0.98: Figs. 1 to 4), PL-100 could be considered as a strong competitive inhibitor of human CYP3A4 although this compound was approximately 13 fold less potent than Ritonavir.
In agreement with litterature data, Ritonavir showed a strong competitive inhibition of CYP3A4 activity with a Ki of 0.034 ± 0.005μM (R2 = 0.98: Figs. 1 and 2).
A Ki of 0.052 ± 0.012 μM was calculated for ketoconazole, a strong competitive inhibitor of CYP3A4 activity (R2 = 0.99: Figs. 1 to 3).
PL-100 is thus considered a strong inhibitor of CYP3A4/5, but with a Ki value 13 fold higher than Ritonavir and 8.5 fold higher than ketoconazole. Example 12. In vivo inhibition of CYP450
Preparation of PPL-100 mixture (40 mg/mL)
An appropriable amount of test article, PPL-100, was dissolved in a mixture of 20% ethanol, 50% propylene glycol, 0.1% Tween and 30% water (v/v/v/v), to obtain a concentration of 40 mg/mL. Purity of the test article was taken into account for calculation of the concentration.
Preparation of Atazanavir mixture (10 mg/mL) Atazanavir was dissolved in a mixture of 20% ethanol, 50% propylene glycol, 0.1% Tween and 30% water (v/v/v/v), to obtain a concentration of 10 mg/mL.
Preparation of Dosing Solutions
Dosing solutions were prepared shortly prior to dosing as follows: Briefly 4 mL of PPL- 100 mixture (40 mg/mL) was mixed with 4 mL of Atazanavir mixture (10 mg/mL).
Animals
Sprague Dawley rats (Rattus πorvβgicus) aged 7 — 8 weeks at start of dosing were used for the study (Charles River Canada Inc., Montreal, PQ). Animals received Atazanavir (25 mg/kg) and PPL-100 (100 mg/kg). Six female rats were used on each time point.
Animals were dosed with a mixture containing Atazanavir (5 mg/mL) and PPL-100 (20 mg/mL) at a dose volume 5 mL per kg body weight. A summary of treatment is illustrated in Table A below.
Table A: Treatment Groups
Figure imgf000070_0001
Blood samples were collected on 6 occasions. Rats were bled as per outline in Table B. For the purpose of collection of the samples indicated above, each rat were bled from the orbital sinus. Each blood sample (approximately 0.4 ml_) was collected into a tube containing the anticoagulant K2 EDTA. The time (actual time, in conjunction with the day and time of dosing) was recorded for each sample. Following collection, the samples was inverted for approximately 10 minutes and then, the samples were centrifuged under refrigeration (2 - 8βC) for 20 minutes at 2000 rpm. The plasma obtained from each sample was recovered and stored frozen (at approximately -800C) pending analysis.
A the time of analysis, protein was extracted from the plasma samples and PL-100 (the active ingredient released rom PPL-100 under physiological conditions) or atazanavir were detected by HPLC-UV-MSD (1100 series, Hewlett Packard).
Table B.
Figure imgf000071_0001
Results of this analysis presented in Fig. 5 and Fig. 6 indicate that PL-100 (PPL-100) decreases the metabolism of Atazanavir in vivo. PL-100 (PPL-100) succesfully increases the plasma concentration of Atazanavir at 24 hours.
Example 13. Microsomal Stability
In vitro studies were conducted to demonstrate that PL-100 enhances protease inhibitors' stability in microsomes.
The protease inhibitors which were tested were: Atazanavir (ATV), Lopinavir (LPV), Saquinavir (SQV), Amprenavir (APV), Nelfinavir (NFV) and Indinavir (IDV).
The concentration of the protease inhibitors and PL-100 was 10 μM, except for the experiment involving Atazanavir, in which Atazanavir was at concentration of 1.0 μM and PL-100 was at a concentration of 2.0 μM. After each protease inhibitor was incubated with microsomes, the percent of parent (original protease inhibitor) remaining was measured. The measurements were conducted at time point 60 minutes. The percent of parent remaining quantifies the extent of stability of protease inhibitors, as protease inhibitors are susceptible to being metabolized into daughter molecules in microsomes, decreasing the amount of the original protease inhibitor. The greater the percent of parent remaining, the greater the stability of the protease inhibitor in microsomes.
The percent of parent remaining was measured for each protease inhibitor in the presence or absence of PL-100. As a control, the percent of protease inhibitor remaining was also measured in the presence of Ritonavir (RTV) to compare the boosting effect between PL-100 and RTV.
The results of the experiment (Figure 7) demonstrate that PL-100 can enhance the stability of all tested protease inhibitors in microsomes. Thus, PPL- 100 is a potential booster of these protease inhibitors in vivo.

Claims

We claim:
1. A pharmaceutical composition comprising:
a) a lysine-based compound of formula I
Figure imgf000073_0001
or a pharmaceutically acceptable salt thereof, wherein n is 3 or 4, wherein X and Y, the same or different, are selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F1 Cl1 Br, I, -CF3, -OCF3, - CN. -NO2, -NR4R5, -NHCOR4. -OR4. -SR4, -COOR4, -COR4. and -CH2OH or X and Y together define an alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula -OCH2CH2O-, wherein R6 is selected from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, wherein R3 is selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R3A-CO-, R3A being selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, - CH2OH, -CF3, -CH2CF3, -CH2CH2CF3, pyrrolidinyl, piperidinyl, 4-morpholinyl, CH3O2C-, CH3O2CCH2-, ACeIyI-OCH2CH2-, HO2CCH2-, 3-hydroxyphenyl, 4- hydroxyphenyl, 4-CH3OC6H4CH2-, CH3NH-, (CH3)2N-, (CH3CH2)2N-, (CH3CH2CHz)2N-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, CβHsCH2O-, 2- pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl-, 2-pyraziπyl, 2-quinolyl, 3-quinolyl, A- quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000074_0001
a picolyl group selected from the group consisting of
Figure imgf000074_0002
a picolyloxy group selected from the group consisting of
Figure imgf000074_0003
a substituted pyridyl group selected from the group consisting of
Figure imgf000074_0004
a group of formula
Figure imgf000074_0005
wherein X1 and Y', the same or different, are selected from the group consisting of H1 a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I1 -CF3, -NO2, - NR4R5, -NHCOR4, -OR4, -SR4, -COOR4, -COR4 and -CH2OH.
wherein R4 and R5, the same or different, are selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
wherein R2 is selected from the group consisting of a diphenylmethyl group of formula IV
Figure imgf000075_0001
a naphthyl-1-CH2- group of formula V
Figure imgf000075_0002
V a naphthyl-2-CH2- group of formula Vl
Figure imgf000075_0003
VI
a biphenylmethyl group of formula VII
Figure imgf000076_0001
and an anthryl-9-CH2- group of formula VIII
Figure imgf000076_0002
VIII
wherein R1 is H or a physiologically cleavable unit;
b) a drug which is affected by cytochrome P450 monooxigenase; and
c) a pharmaceutically acceptable carrier;
wherein the ratio (w/w) of the lysine-based compound, or pharmaceutically acceptable salt thereof, to said drug which is affected by cytochrome P450 monooxigenase is between about 4:1 and about 1:1.
2. A pharmaceutical composition according to claim 1 wherein the lysiπe-based compound is a compound of formula II,
Figure imgf000077_0001
or a pharmaceutically acceptable salt thereof.
3. The pharmaceutical composition of claim 1 or claim 2, wherein R1 is selected from the group consisting of H, (HO)2P(O) and (MO)2P(O), and a group of formula R1A- CO-, R1A being selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH2OH, CH3O2C-, CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 2- hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, (CH3)2NCH2-, (CHs)2CHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, 2-pyrrolyl, 2- pyridyl, 3-pyridyl, 4-pyridyl, 1-methyl-1 ,4-dihydro-3-pyridyl, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000077_0002
a picolyl group selected from the group consisting of
Figure imgf000078_0001
(2-picolyl) (3-picolyl) (4-picolyl)
a picolyloxy group selected from the group consisting of
Figure imgf000078_0002
(2-picolyloxy) (3-picolyloxy) (4-picolyloxy)
a substituted pyridyl group selected from the group consisting of
Figure imgf000078_0003
Figure imgf000078_0004
(substituted 4-pyridyl)
and a group of formula,
Figure imgf000078_0005
wherein M is an alkali metal or alkaline earth metal and wherein X', Y', R4 and Rs are as defined in claim 1.
4. The pharmaceutical composition of claim 1 , wherein X, Y, n, R1 , R2, R3, R6, X' and Y' are set forth for each of the lysine-based compounds of formula I below:
Figure imgf000079_0002
5. The pharmaceutical composition of any one of claims 1 to 4, wherein said drug which is affected by cytochrome P450 monoxigenase is selected from the group consisting of Atazanavir (ATV), Lopinavir (LPV)1 Saquinavir (SQV), Amprenavir (APV)1 Nelfinavir (NFV) and Indinavir (IDV).
6. The pharmaceutical composition of any one of claims 1 to 5, wherein the ratio (w/w) of said lysine-based compound and said drug which is affected by cytochrome P450 monooxigenase is about 4:1.
7. The pharmaceutical composition of any one of claims 1 to 6, wherein said cytochrome P450 monoxignease is CYP3A4.
8. A pharmaceutical combination comprising:
a) a lysine-based compound of formula I
Figure imgf000079_0001
or a pharmaceutically acceptable salt thereof,
wherein n is 3 or 4,
wherein X and Y, the same or different, are selected from the group consisting of
H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F1 Cl1 Br1 11 -CF3, -OCF3, - CN, -NO2, -NR4R5, -NHCOR4, -OR4, -SR4, -COOR4. -COR4, and -CH2OH or X and Y together define an alkylenedioxy group selected from the group consisting of a methylenedioxy group of formula -OCH2O- and an ethylenedioxy group of formula
-OCH2CH2O-.
wherein R6 is selected from the group consisting of a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof,
wherein R3 is selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, and a group of formula R3A-CO-, R3A being selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atoms, tetrahydro-3-furanyloxy, - CH2OH, -CF3, -CH2CF3, -CH2CH2CF3, pyrrolidinyl, piperidinyl. 4-morpholinyl,
CH3O2C-, CH3O2CCH2-, Acetyl-OCH2CH2-, HO2CCH2-, 3-hydroxyphenyl, 4- hydroxyphenyl, 4-CH3OC6H4CH2-, CH3NH-, (CH3)2N-, (CH3CH2)2N-, (CH3CH2CH2)2N-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, C6H5CH2O-, 2- pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl-, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, A- quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000080_0001
a picolyl group selected from the group consisting of
Figure imgf000081_0001
a picolyloxy group selected from the group consisting of
Figure imgf000081_0002
a substituted pyridyl group selected from the group consisting of
Figure imgf000081_0003
a group of formula
Figure imgf000081_0004
wherein X' and V1 the same or different, are selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, a cycloalkyl group of 3 to 6 carbon atoms, F, Cl, Br, I, -CF3, -NO2, - NR4R5, -NHCOR4. -OR4, -SR4, -COOR4, -COR4 and -CH2OH,
wherein R4 and R5, the same or different, are selected from the group consisting of H, a straight alkyl group of 1 to 6 carbon atoms, a branched alkyl group of 3 to 6 carbon atoms, and a cycloalkyl group of 3 to 6 carbon atoms,
wherein R2 is selected from the group consisting of a diphenylmethyl group of formula IV
Figure imgf000082_0001
a naphthyl-1-CH2- group of formula V
Figure imgf000082_0002
a naphthyl-2-CH2- group of formula Vl
Figure imgf000082_0003
VI
a biphenylmethyl group of formula VII
Figure imgf000083_0001
and an anthryl-9-CH2- group of formula VIII
Figure imgf000083_0002
VIII
wherein Ri is H or a physiologically cleavable unit; and
b) a drug which is affected by cytochrome P450 monooxigenase;
wherein the ratio (w/w) of said lysine-based compound, or pharmaceutically acceptable salt thereof, to said drug which is affected by cytochrome P450 monooxigenase is between about 4:1 and about 1 :1.
9. A pharmaceutical combination of claim 8 wherein the lysine-based compound is a compound of formula II,
Figure imgf000083_0003
or a pharmaceutically acceptable salt thereof.
10. The pharmaceutical combination of claim 8 or claim 9, wherein R1 is selected from the group consisting of H1 (HO)2P(O) and (MO)2P(O), and a group of formula R1A- CO-, R1A being selected from the group consisting of a straight or branched alkyl group of 1 to 6 carbon atoms, a cycloalkyl group having 3 to 6 carbon atoms, a cycloalkylalkyl group having 3 to 6 carbon atoms in the cycloalkyl part thereof and 1 to 3 carbon atoms in the alkyl part thereof, an alkyloxy group of 1 to 6 carbon atom, -CH2OH, CH3O2C-, CH3O2CCH2-, Acetyl-OCHzCHz-, HO2CCH2-. 2- hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, (CH3)2NCH2-, (CH3)2CHCH(NH2)-, HOCH2CH2NH-, CH3OCH2O-, CH3OCH2CH2O-, 2-pyrrolyl, 2- pyridyl, 3-pyridyl, 4-pyridyl, 1-methyl-1,4-dihydro-3-pyridy1, 2-pyrazinyl, 2-quinolyl, 3-quinolyl, 4-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 2-quinoxalinyl, a phenyl group of formula
Figure imgf000084_0001
III
a picolyl group selected from the group consisting of
Figure imgf000084_0003
(2-picolyl) (3-picolyl)
Figure imgf000084_0002
a picolyloxy group selected from the group consisting of
Figure imgf000085_0001
(2-picolyloxy) (3-picolyloxy) (4-picolyloxy)
a substituted pyridyl group selected from the group consisting of
Figure imgf000085_0002
(substituted 2-pyridyl) (substituted 3-pyridyl) (substituted 4-pyridyl)
and a group of formula,
Figure imgf000085_0003
wherein M is an alkali metal or alkaline earth metal and wherein X', Y', R4 and Rs are as defined in claim 8.
11. The pharmaceutical combination of claim 8, wherein X, Y1 n, R1 , R2, R3, R6, X' and Y' are set forth for each of the lysine-based compounds of formula I below:
Figure imgf000086_0002
12. The pharmaceutical combination of any one of claims 8 to 11 , wherein said drug which is affected by cytochrome P450 monoxigenase is selected from the group consisting of Atazanavir (ATV). Lopinavir (LPV)1 Saquinavir (SQV), Amprenavir
(APV), Nelfinavir (NFV) and Indinavir (IDV).
13. The pharmaceutical combination of any one of claims 8 to 12, wherein the ratio (w/w) of said lysine-based compound and said drug which is affected by cytochrome P450 monooxigenase is about 4: 1.
14. The pharmaceutical combination of any one of claims 8 to 13, wherein said cytochrome P450 monoxignease is CYP3A4.
15. The use of at least one lysine-based compound of formula I
Figure imgf000086_0001
or a pharmaceutically acceptable salt thereof and a drug which is affected by cytochrome P450 monooxigenase in the manufacture of a drug for the treatment or prevention of an HIV infection or for treatment or prevention of AIDS, wherein n, X, Y, X', Y', R1, R2, R3, R4, R5 and R6 are as defined in claim 1;
and wherein the ratio (w/w) of the lysine-based compound and the drug which is affected by cytochrome P450 monooxigenase is between about 4:1 and about 1:1.
16. A method of treating or preventing an HIV infection or of treating or preventing
AIDS, the method comprising administering either the pharmaceutical composition as defined in claim 1, or the pharmaceutical combination as defined in claim 8, to a mammal in need thereof.
17. A method of treating or preventing an HIV infection or of treating or preventing AIDS, which comprises administering
a) a lysine-based compound of formula I
Figure imgf000087_0001
wherein n, X, Y, X', Y", R1, R2, R3, R4, Rs and R8 are as defined in claim 1 or a pharmaceutically acceptable salt thereof, and
b) one or more drugs which are affected by cytochrome P450 monoxigenase,
wherein said lysine-based compound is in an amount which is sufficient to reduce the metabolism of said one or more drugs which are affected by cytochrome P450 monoxigenase, and wherein the ratio (w/w) of said lysine based compound to said one or more drugs is between about 4:1 and about 1 :1.
18. A method for improving the pharmacokinetics of a drug which is affected by cytochrome P450 monoxigenase, the method comprising administering to a human in need thereof an amount of the lysine-based compound of formula I of claim 1 effective to inhibit cytochrome P450 monoxigenase and the drug which is affected by cytochrome P450 monoxigenase, wherein the ratio (w/w) of the lysine- based compound to said drug is between about 4:1 and about 1:1.
PCT/IB2007/003593 2006-07-17 2007-07-16 Method for improving pharmacokinetics WO2008023273A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2009520074A JP2010502569A (en) 2006-07-17 2007-07-16 How to improve pharmacokinetics
AU2007287353A AU2007287353A1 (en) 2006-07-17 2007-07-16 Method for improving pharmacokinetics
CA002657316A CA2657316A1 (en) 2006-07-17 2007-07-16 Method for improving pharmacokinetics
EP07825714A EP2109461A2 (en) 2006-07-17 2007-07-16 Method for improving pharmacokinetics

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US83120906P 2006-07-17 2006-07-17
US60/831,209 2006-07-17

Publications (3)

Publication Number Publication Date
WO2008023273A2 true WO2008023273A2 (en) 2008-02-28
WO2008023273A8 WO2008023273A8 (en) 2008-06-05
WO2008023273A3 WO2008023273A3 (en) 2011-03-03

Family

ID=39107180

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2007/003593 WO2008023273A2 (en) 2006-07-17 2007-07-16 Method for improving pharmacokinetics

Country Status (5)

Country Link
EP (1) EP2109461A2 (en)
JP (1) JP2010502569A (en)
AU (1) AU2007287353A1 (en)
CA (1) CA2657316A1 (en)
WO (1) WO2008023273A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009148600A2 (en) * 2008-06-06 2009-12-10 Concert Pharmaceuticals, Inc. Deuterated lysine-based compounds
WO2010138338A1 (en) 2009-05-27 2010-12-02 Merck Sharp & Dohme Corp. Hiv protease inhibitors
WO2012055031A1 (en) 2010-10-28 2012-05-03 Merck Canada Inc. Hiv protease inhibitors
US8227450B2 (en) 2005-11-30 2012-07-24 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
WO2013059928A1 (en) 2011-10-26 2013-05-02 Merck Canada Inc. Hiv protease inhibitors
US9187415B2 (en) 2010-10-29 2015-11-17 Merck Canada Inc. Sulfonamides as HIV protease inhibitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2664118A1 (en) * 2006-09-21 2008-07-03 Ambrilia Biopharma Inc. Protease inhibitors
WO2017209732A1 (en) * 2016-05-31 2017-12-07 Taimed Biologics, Inc. A long acting pharmaceutical composition of protease inhibitor

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6632816B1 (en) * 2002-12-23 2003-10-14 Pharmacor Inc. Aromatic derivatives as HIV aspartyl protease inhibitors
US20060025592A1 (en) * 2004-08-02 2006-02-02 Stranix Brent R Lysine based compounds
WO2006012725A1 (en) * 2004-08-02 2006-02-09 Ambrilia Biopharma Inc. Lysine based compounds
WO2006114001A1 (en) * 2005-04-27 2006-11-02 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
WO2007062526A1 (en) * 2005-11-30 2007-06-07 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6632816B1 (en) * 2002-12-23 2003-10-14 Pharmacor Inc. Aromatic derivatives as HIV aspartyl protease inhibitors
US20060025592A1 (en) * 2004-08-02 2006-02-02 Stranix Brent R Lysine based compounds
WO2006012725A1 (en) * 2004-08-02 2006-02-09 Ambrilia Biopharma Inc. Lysine based compounds
WO2006114001A1 (en) * 2005-04-27 2006-11-02 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
US20060287316A1 (en) * 2005-04-27 2006-12-21 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
WO2007062526A1 (en) * 2005-11-30 2007-06-07 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
STRANIX ET AL: 'Antiviral activity and cross-resistance profile of P-1946, a novel human immunodeficiency virus type 1 protease inhibitor' ANTIVIRAL RES. vol. 70, no. 2, 2006, pages 17 - 20 *
STRANIX ET AL: 'In Viro Antiviral Activity and Cross-Resistance Profile of PL-100, a Novel Protease Inhibitor of Human Immunodeficiency Virus Type 1' ANTIMICROB. AGENTS CHEMOTHER. vol. 51, no. 11, 2007, pages 4036 - 4043 *
STRANIX ET AL: 'Lysine sulfonamides as novel HIV-protease inhibitors: Ne-Acyl aromatic a-amino acids' BIOORG. MED.CHEM. LETT. vol. 16, no. 13, 2006, pages 3459 - 3462 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8227450B2 (en) 2005-11-30 2012-07-24 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
US8580995B2 (en) 2005-11-30 2013-11-12 Taimed Biologics, Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
WO2009148600A2 (en) * 2008-06-06 2009-12-10 Concert Pharmaceuticals, Inc. Deuterated lysine-based compounds
WO2009148600A3 (en) * 2008-06-06 2010-04-15 Concert Pharmaceuticals, Inc. Deuterated lysine-based compounds
WO2010138338A1 (en) 2009-05-27 2010-12-02 Merck Sharp & Dohme Corp. Hiv protease inhibitors
WO2012055031A1 (en) 2010-10-28 2012-05-03 Merck Canada Inc. Hiv protease inhibitors
US9079834B2 (en) 2010-10-28 2015-07-14 Merck Canada Inc. HIV protease inhibitors
US9187415B2 (en) 2010-10-29 2015-11-17 Merck Canada Inc. Sulfonamides as HIV protease inhibitors
WO2013059928A1 (en) 2011-10-26 2013-05-02 Merck Canada Inc. Hiv protease inhibitors
US9133157B2 (en) 2011-10-26 2015-09-15 Merck Canada Inc. HIV protease inhibitors

Also Published As

Publication number Publication date
WO2008023273A3 (en) 2011-03-03
AU2007287353A1 (en) 2008-02-28
WO2008023273A8 (en) 2008-06-05
EP2109461A2 (en) 2009-10-21
JP2010502569A (en) 2010-01-28
CA2657316A1 (en) 2008-02-28
AU2007287353A8 (en) 2009-02-26

Similar Documents

Publication Publication Date Title
US8008297B2 (en) Lysine based compounds
WO2008023273A2 (en) Method for improving pharmacokinetics
EP1971615B1 (en) Lysme-based prodrugs of aspartyl protease inhibitors and processes for their preparation
WO2006114001A1 (en) Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
AU2009230800A1 (en) Lysine based compounds
US6632816B1 (en) Aromatic derivatives as HIV aspartyl protease inhibitors
RU2379312C2 (en) Lysine compounds, pharmaceutical composition containing these compounds, application of said compounds for treatment or prevention of hiv-infection
ZA200607352B (en) Lysine based compounds
CN102093411A (en) Lysine-based compounds
NZ552853A (en) Lysine based compounds
KR20070057815A (en) Lysine based compounds

Legal Events

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

Ref document number: 2009520074

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2007287353

Country of ref document: AU

Ref document number: 2657316

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007287353

Country of ref document: AU

Date of ref document: 20070716

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2007825714

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU

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

Ref document number: 07825714

Country of ref document: EP

Kind code of ref document: A2