OA16300A - Methods and compounds for treating paramyxoviridae virus infections. - Google Patents

Methods and compounds for treating paramyxoviridae virus infections. Download PDF

Info

Publication number
OA16300A
OA16300A OA1201300013 OA16300A OA 16300 A OA16300 A OA 16300A OA 1201300013 OA1201300013 OA 1201300013 OA 16300 A OA16300 A OA 16300A
Authority
OA
OAPI
Prior art keywords
alkyl
compound
independently
substituted
formula
Prior art date
Application number
OA1201300013
Inventor
Richard L. Mackman
Jay P. Parrish
Adrian S. Ray
Dorothy Agnes Theodore
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of OA16300A publication Critical patent/OA16300A/en

Links

Abstract

Provided are methods for treating Paramyxoviridae virus infections by administering ribosides, riboside phosphates and prodrugs thereof, of Formula (I) :

Description

METHODS AND COMPOUNDS FOR TREATING PARAMYXOVIRIDAE VIRUS INFECTIONS
FIELD OF THE INVENTION
The invention relates generally to methods and compounds for treating Paramyxoviridae virus infections, particularly methods and nucleosides for treating respiratory syncytial virus infections and paraînfluenza virus infections.
BACKGROUND OF THE INVENTION
Paramyxo viruses of the Paramyxoviridae family are negative-sense, singlestranded, RNA viruses that are responsible for many prévalent human and animal diseases. These viruses comprise at least two major subfamilies, Paramyxovirinae and Pneumovirinae. The subfamily Paramyxovirina includes the human paraînfluenza viruses (HPIV), measles virus and mumps virus. Although, vaccines are available to prevent measles and mumps infections, these infections caused 745, 00 deaths in 2001 so additional treatments would be desireable for susceptible populations. HPIV are the second most common cause of lower respiratory tract infection in younger children and coliectively cause about 75% of the cases of Croup (http://www.cdc.gov/ncidod/dvrd/revb/respiratory/hpivfeat.htm). HPIVs can cause repeated infections throughout life including upper respiratory tract illness and even serious lower respiratory tract disease (e.g., pneumonia, bronchitis, and bronchiolitis), the latter being especially of concem among the elderly, and among patients with compromised immune Systems (Sable, Infect. Dis. Clin. NorthAm. 1995, 9, 987-1003). Currently, no vaccines are available to prevent HPIV infections. Therefore there is a need for arAi-Paramyxovirina therapeutics.
The subfamily Pneumovirinae includes Human respiratory syncytial virus (HRSV). Almost ail children will hâve had an HRSV infection by their second birthday. HRSV is the major cause of lower respiratory tract infections in infancy and childhood with 0.5% to 2% of those infected requiring hospitalization. The elderly and adults with chrome heart, lung diease or those that are immunosuppressed also hâve a high risk for developing severe HRSV disease (http://www.cdc.gov/rsv/index.html). No vaccine to prevent HRSV infection is currently available. The monoclonal antibody palivîzumab is available for infants at high risk, e.g., prématuré infants or those with either cardiac or lung disease, but the cost for general use is often prohibitive. Ribavirin has also been used to treat HRSV infections but has limited efficacy. Therefore, there is a need for anti-Pneumovirinae therapeutics and antiParamyxoviridae therapeutics in general.
Ribosides of the nucleobases pyrrolo[l,2-f][l,2,4]triazine, imidazo[l,5-
f][l,2,4]triazine, îmidazo[l,2-f][l,2,4]triazine, and [l,2,4]triazolo[4,3-f][l,2,4]triazine hâve been disclosed in Carbohydrate Research 2001, 331 (l), 77-82; Nucleosides & Nucléotides (1996), 15(1-3), 793-807; Tetrahedron Letters (1994), 35(30), 5339-42; Heterocycles (1992), 34(3), 569-74; J. Chem. Soc. Perkin Trans. I 1985, 3, 621-30; J. Chem. Soc. Perkin Trans. 1 1984, 2, 229-38; WO 2000056734; Organic Letters (2001), 3(6), 839-842; J. Chem. Soc. Perkin Trans. 1 1999, 20, 2929-2936; and J. Med. Chem. 1986,29(11), 2231-5. Ribosides of pyrrolo [ 1,2-f] [ 1,2,4]triazine nucleobases with antiviral, antî-HCV, and anti-RdRp activity hâve been disclosed by Babu (W02008/089105 and W02008/141079) and Francom(W02010/002877).
Butler, et al., WO2009132135, disclose Γ substituted ribosides and prodrugs comprisingpyrrolo[l,2-f][l,2,4]triazine nucleobases whichhâve anti-HCV and antiRdRp activity. However, no methods of treating Paramyxoviridae infections with these compounds hâve been disclosed.
SUMMARY OF THE INVENTION
Provided are methods and compounds for the treatment of infections cased by the Paramyxoviridae virus family.
Provided, is a method for treating a Paramyxoviridae infection in a mammal in need thereof comprising admînistering a therapeutically effective amount of a compound of Formula I: xyA'
Formula I or a pharmaceutically acceptable sait or ester, thereof;
wherein:
each R1 is H or halogen;
each R2, R3 or R5 is independently H, ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Cj-C8)alkyl, (C4-C8)carbocyclylalkyl, (Cj-Cs)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl or (C2-C8)substituted alkynyl;
or any two R2, R3 or Rs on adjacent carbon atoms when taken together are O(CO)O- or when taken together with the ring carbon atoms to which they are attached form a double bond;
R6 îs ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, -C(=O)Rn, -C(=O)OR, C(=O)NRllR12, -C(=O)SR“, -SfOjR11, -S(O)2Rn, -S(O)(OR11), -SfOHOR1). -SO2NRHR12, halogen, (C]-C8)alkyl, (C4-C8)carbocyclylalkyl, (C1-C8)substituted alkyl, (C2-C8)alkenyl, (C>-Cs (substituted alkenyl, (C2- Csjalkynyl. (C2-C8)substituted alkynyl, or aryl(Ci-C8)alkyl;
each n is independently 0, 1, or 2;
each Ra is independently H, (Ci-C8)alkyl, (C2-C8 (alkenyl, (C2-C8)alkynyl, aryl(Ci-C8)aIkyl, (C4-C8)carbocyclylalkyl, -C(-O)R. -C(=O)OR11, -C(=O)NRhR12, C(=O)SRn, -S(O)Rn, -S(O)2R, -S(O)(ORh), -S(O)2(ORh), or-SO2NR1,R12;
R7 is H, -C(=O)R1!, -C(=O)OR, -C(=O)NRhR12, -C(=O)SRh, -S(O)Rh, S(O)2Rn, -S(O)(OR1!), -S(O)2(ORii(, -SO2NR1'r'2, or
Y
II
w2 each Y or Yf is, independently, O, S, NR, +N(0)(R), N(OR), +N(O)(OR), or N-NR2;
Wl and W2, when taken together, are -Y3(C(Ry)2)îY3-; or one of W1 or W2 together with either R3 or R4 is -Y3- and the other of W1 or W2 îs Formula la; or W1 and W2 are each, independently, a group of the Formula la:
M2
Formula la wherein:
each Y2 is independently a bond, O, CR2, NR, 'N(0)(R), N(0R), +N(0)(0R),
N-NR2, S, S-S, S(O), or S(O}>:
each Y3 is independently O, S, or NR;
M2 is 0, I or 2;
each R* is independently Ry or the formula:
each Ml a, Mlc, and Mld is independently 0 or 1;
M12c is 0, 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12;
each Ry is independently H, F, Cl, Br, I, OH, R, -C(-Y')R, -C(=Y*)OR, C(=Yl)N(Rh, -N(R)2, -+N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(OR), -S(0)2(OR), OC(=Y’)R, -OC(=Yl)OR, -OC(=Y')(N(R)2), -SC( y')R. -SC(=Y])OR, SC(=Yl)(N(R)2), -N(R)C(=Y1)R, -N(R)C(=Y1)OR, -N(R)C(=Y')N(R)2, -so2nr2, -CN, -N3, -NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
each R is independently H, (Ci-Cg) alkyl, (C]-C8) substituted alkyl, (C2C8)alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl, (C2-C8) substituted alkynyl, C6-C20 aryl, substituted aryl, C2-C2o heterocyclyl, C2-C2o substituted heterocyclyl, arylalkyl or substituted arylalkyl;
W3 is W4 or W5; W4 is R, -C(Y’)Ry, -C(Y')W5, -SO2Ry, or -SO2W5; and W5 is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups;
each R8 is halogen, NRr'2, Nîr'^OR11, NR1iNR,1R!2) N3, NO, NO2, CHO, CN, -CH(=NRn), -CH=NNHRh, -CH=N(OR11), -CH(OR)2, -C(=O)NR,lR12, -C(=S)NRllR12, -C(-O)ORH, (C,-Ce)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C4-Cs)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=O)(Cj-Cs)alkyl, -S(O)„(Ci-C8)alkyl, aryl(CrC8)alkyl, OR]1 or SR11;
each R9 or R10 is independently H, halogen, NR1!R12, NiR’^OR11, NRllNR11R12, Ν3, NO, NÛ2, CHO, CN, -CH(=NRn), -CH=NHNRn, -CH=N(ORn), -CH (OR1 ’)2, -C(=O)NR' lR12, -C(=S)NRR12, -C(=O)OR’ R11, OR11 or SR11 ;
each R11 or R12 is independently H, (C|-C8)alkyl, (C2-C8)alkenyl, (C2C8)alkynyl, (C4-C8)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=OXCi-C8)alkyl, -S(O)n(Cl-C8)alkyl or aryl(Ci-C8)alkyl; or R11 and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or —NRa-; and wherein each (C]-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(Ci-C8)alkyl of each R2, R3, R5, R6, R11 or R12 is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or ORa; and wherein one or more of the non terminal carbon atoms of each said (Ci-Cs)alkyl may be optionally replaced with -O-, S- or-NRa-.
In another embodiment, the method comprises administering a therapeutically effective amount of a racemate, enantiomer, diastereomer, tautomer, polymorph, pseudopolymorph, amorphous form, hydrate or solvaté of a compound of Formula I or a pharmaceutically acceptable sait or ester thereof to a mammal in need thereof.
In another embodiment, the method comprises treating a Paramyxovirina infection in a mammal in need thereof by administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, the method comprises treating a parainfluenza, measles or mumps virus infection în a mammal in need thereof by administering a therapeutically effective amount of a compound of Formula l or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, the method comprises treating a parainfluenza virus infection in a mammal in need thereof by administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, the method comprises treating a Pneumovirinae infection in a mammal in need thereof by administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, the method comprises treating a respiratory syncytial virus infection in a mammal in need thereof by administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, the method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising an effective amount of a Formula I compound, or a pharmaceutically acceptable sait or ester thereof, in combination with a pharmaceutically acceptable diluent or carrier. y/'
In another embodiment, the method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising an effective amount of a
Formula I compound, or a pharmaceutically acceptable sait or ester thereof, in combination with at least one additional therapeutic agent.
In another embodiment, the method comprises administering a therapeutically effective amount of a combination pharmaceutical agent comprising:
a) a first pharmaceutical composition comprising a compound of Formula I; or a pharmaceutically acceptable sait, solvaté, or ester thereof; and
b) a second pharmaceutical composition comprising at least one additional therapeutic agent active against infectious Paramyxoviridae viruses.
In another embodiment, the présent application provides for a method of inhibiting a Paramyxoviridae RNA-dependent RNA polymerase, comprising contacting a cell infected with a Paramyxoviridae virus with an effective amount of a compound of Formula I; or a pharmaceutically acceptable salts, solvaté, and/or ester thereof.
In another embodiment, provided is the use of a compound of Formula I or a pharmaceutically acceptable sait, solvaté, and/or ester thereof to treat a viral infection caused by a Paramyxoviridae virus.
In another aspect, the invention also provides processes and novel intermediates disclosed herein which are useful for preparing Formula I compounds of the invention.
In other aspects, novel methods for synthesis, analysis, séparation, isolation, purification, characterization, and testing of the compounds of this invention are provided.
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying description, structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover ail alternatives, modifications, and équivalents, which may be included within the scope of the présent invention.
In another embodiment, provided îs a method of treating a Paramyxoviridae infection in a mammal in need thereof comprising administering a therapeutically effective amount of a compound of Formula I represented by Formula II:
R3 Rz
Formula II or a pharmaceutically acceptable sait or ester, thereof;
wherein:
each R1 is H or halogen;
each R3 is ORa or halogen;
each R3 or R5 is independently H, ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Ci-Cg)alkyl, (C4-C8)carbocyclylalkyl, (C|-C8)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl or (C2-C8)substituted alkynyl;
or any two R2, R3 or R5 on adjacent carbon atoms when taken together are O(C0)0- or when taken together with the ring carbon atoms to which they are attached form a double bond;
R6 is ORa, N(Ra)2, N3, CN, S(O)nRa, -C(=O)Rn, -C(O)0Rh. -C(=O)NRHR12, C(=O)SRn, -S(O)R11, -S(O)2Rh, -S(O)(ORh), -S(O)?(ORfl). -SO2NR”R12, halogen, (Ci-Cs)alkyl, (C4- Cx)carbocyclylalkyl, (Ci-C8)substituted alkyl, (C2-C8)alkenyl, (C2- Cx(substituted alkenyl, (C2-C8)alkynyl, or (C2-C8)substituted alkynyl;
each n is independently 0, 1, or each Ra is independently H, (Ct-Cs)alkyl, (C2-Cs)alkenyl, (C2-Cs)alkynyl, aryliCi-Cslalkyl, (C4-C8)carbocyclylalkyl, -C(=O)R”, -C(=O)0R, -C(-O)NRllRl2, C(=O)SRJl, -S(O)R'‘, -S(O)2R, -S(O)(ORh), -S(O)2(ORu), or-SO2NR11R12;
R7 is H, -C(=O)R11, -C(=O)OR, -C(=O)NRl1R12, -C(=O)SRn, -S(O)Rn, 5 5(0)28. -StOXOR11), -S(O)2(OR11), -SO2NR' lR12, or
W‘ each Y or Y* is, independently, O, S, NR, +N(O)(R), N(0R), +N(O)(OR), or N-NR2;
W1 and W2, when taken together, are -Y3(C(Ry)2)3Y3-; or one of W1 or W2 together with either R3 or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of the Formula la:
R:
Formula la wherein:
each Y2 is independently a bond, O, CR2, NR, ’N(O)(R), N(OR), +N(O)(OR),
N-NR2, S, S-S, S(O), or S(O)2;
each Y3 is independently O, S, or N R;
M2 is 0, 1 or 2;
each R* is independently Ry or the formula:
each Mla, Mle, and Mld is independently 0 or 1;
M12c is 0, 1, 2, 3, 4, 5, 6, 7, 8,9, 10, 11 or 12;
each Ry is independently H, F, Cl, Br, I, OH, R, -C(=Y’)R, -C(=y')OR, C(=Y‘)N(R)2, -N(R)2, -+N(R)3î -SR, -S(O)R, -S(O)2R, -S(O)(OR), -S(0)2(OR), OC(=Y’)R, -OC(=Y!)OR, -OC(=Y')(N(R)2), -SC(=Y‘)R, -SC(=Y')OR, SC(=Y1)(N(R)2), -N(R)C(=Y')R, -N(R)C(=Y’)OR, -NtRjC^Y'jNCRE -SO2NR2, -CN, —N3, -NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
each R is independently H, (Ci-Cg) alkyl, (Ci-Q) substituted alkyl, (C2C8)alkenyl, (C2-C8) substituted alkenyl, (C2-Cg) alkynyl, (C2-C8) substituted alkynyl, C6-C2o aryl, C6-C2o substituted aryl, C2-C2o heterocyclyl, C2-C20 substituted heterocyclyl, arylalkyl or substituted arylalkyl;
W3 is W4 or W5; W4 is R, -C(Y')Ry, -C(Yl)W5, -SO2Ry, or -SO2W5; and W5 is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups;
each Rs is halogen, NR1^'2, N(RU)ORU, NRl1NRllR12, N3, NO, NO2, CHO, CN, -CH(=NR”), -CH=NNHR, -CH=N(ORu), -CH(ORh)2, -C(=O)NRhR12, -C(=S)NRllR12, -C(=O)ORn, (CrC8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C4-Cg)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=OXCi-C8)alkyI, -S(O)n(C]-C8)alkyl, aryl(CrC8)alkyl, OR11 or SR11;
each R9 is independently H, halogen, NR1 'R12, N(Rll)OR11, NR,1NR1,R12, N3,
NO, NO2, CHO, CN, -CH(=NR11), -CH=NHNR, -CH=N(ORh), -C(=O)NRliR12, -C(=S)NRR,?, -C(=O)ORh, R11, OR11 or SRn;
each R11 or R12 is independently H, (C|-C8)alkyl, (C2-C8)alkenyl, (C2C8)alkynyl, (C4--C8)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(CrC8)alkyl, -S(O)n(C]-C8)aIkyi or aryl(C[-C8)alkyl; or R11 and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or N R1'-; and wherein each (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(C]-C8)alkyl of each R3, R5, R6, R1[ or R12 is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or ORa; and wherein one or more of the non-terminal carbon atoms of each said (C[-C8)alkyl may be optionally replaced with -O-, -S- or NRa-.
In one embodiment of the method of treating a Paramyxoviridae infection by administering a compound of Formula II, R1 of Formula II is H. In another aspect of this embodiment R6 of Formula II is N3, CN, halogen, (Ci-Cg)alkyl, (Ci~C8)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl, or (C2-C8)substituted alkynyl. In another aspect of this embodiment, R6of Formula II îs CN, methyl, ethenyl, or ethynyl. In another aspect of this embodiment, R6 of Formula II is CN. In another aspect of this embodiment, R6of Formula II is methyl. In another aspect of this embodiment, R5 of Formula II is H. In another aspect of this embodiment, R2 of Formula II is ORa. In another aspect of this embodiment, R2 of Formula II is OH. In another aspect of this embodiment, R2of Formula II is F. In another aspect of this embodiment, R3 of Formula II is ORa. In another aspect of this embodiment, R3 of Formula II is OH, -OC(=O)R'l, or -OC(=O)OR'1. In another aspect of this embodiment, R3 of Formula II is OH. In another aspect of this embodiment, RK of Formula II is NR1 ‘R12. In another aspect of this embodiment, R8 of Formula II is u 11
NH2. In another aspect of this embodiment, R of Formula II is OR . In another aspect of this embodiment, RK of Formula II is OH. In another aspect of this embodiment, R9 of Formula II is H. In another aspect of this embodiment, R9 of Formula II is NR1 ‘R12. In another aspect of this embodiment, R9of Formula II is NH2. In another aspect of this embodiment, R7 of Formula 11 is H, -C(=O)R11, -C(=O)OR ' or
II
W2 , In another aspect of this embodiment, R7 of Formula II is H. In another aspect of this embodiment, R7 of Formula II is
O
II
w2
In another embodiment of the method of treating a Paramyxoviridae infection comprising administering a compound of Formula II, the Paramyxoviridae infection is caused by a Paramyxovirina virus. In another aspect of this embodiment, the Paramyxovirina virus is a parainfluenza, measles or mumps virus. In another aspect of this embodiment, the Paramyxovirina virus is a Respirovirus virus. In another aspect of this embodiment, the Paramyxovirina virus is a type l or 3 Human parainfluenza virus.
In another embodiment of the method of treating a Paramyxoviridae infection comprising administering a compound of Formula II, the Paramyxoviridae infection is caused by a Pneumovirinae virus. In another aspect of this embodiment, the Pneumovirinae virus is a respiratory syncytial virus. In another aspect of this embodiment, the Pneumovirinae virus is a Human respiratory syncytial virus.
In another embodiment, provided is a method of treating a Paramyxoviridae infection in a mammal in need thereof comprising administering a therapeutically effective amount of a compound of Formula I represented by Formula III:
Formula III or a pharmaceutically acceptable sait or ester, thereof; wherein:
each R2 is ORa or F;
each R3 îs ORa;
R6 is ORa, N(Ra)2, N3, CN, S(O)„Ra, -C( O)R. -C(=O)ORh, -C(-0)NRhR12, C(=O)SRh, -S(O)R11, -S(O)2Rh, -S(O)(OR11), -S(O)2(ORh), -SO2NRR12, halogen, (C]-Cg)alkyl, (C4-C8)carbocyclylalkyl, (Ci-Cg)substituted alkyl, (C2-Cy)alkenyl. (C2-Cs)substituted alkenyl, (C2-C8)alkynyl, or (C2-C8)substituted alkynyl;
each n is independently 0, 1, or 2;
each Ra is independently H, (C|-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, aryl(CrCs)alkyI, (C4-C8)carbocyclyl alkyl, -C(=O)R, -C(=O)ORh, -C(=O)NRllR12, C(=O)SR1], -S(O)R, -S(O)2Rh, -S(O)(ORu), -SfOhfOR11), or -SO2NRllR12;
R7 is H, -C(=O)R, -C(=O)OR, -C(=O)NRnR12,-C(=O)SR11, -S(O)Rh, S(O)2RU, -S(O)(ORn), -S(O)2(OR11), -SO2NR“r'2, or
Y
II
w2 each Y or Y1 is, independently, O, S, NR, +N(O)(R), N(OR), +N(O)(OR), or N-NR2;
W1 and W2, when taken together, are -Y3(C(Ry)2)3Y3-; or one of W1 or W2 together with either R3 or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of the Formula la:
M2
Formula la wherein:
each Y2 is independently a bond, O, CR?, NR, +N(O)(R), N(OR), +N(O)(OR), N-NR2, S, S-S, S(O), or S(O)2;
each Y3 is independently O, S, or NR;
M2 is 0, 1 or 2;
each R* is independently Ry or the formula:
each Ml a, Ml c, and Mld is independently 0 or 1;
M12c is 0, 1, 2, 3,4, 5, 6, 7, 8, 9, 10, 11 or 12;
each Ry is independently H, F, Cl, Br, I, OH, R, -C(=y')R, -C(=Y1)OR, C(=Y‘)N(R)2, -N(R)2, -+N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(OR), -S(O)2(OR), OC(=Y‘)R, -OC(=Y1)OR, -OC(=YlXN(Rh), -SC(=Y')R, -SC(=Y1)OR, SC(=Y*)(N(R)2), -N(R)C(=Y')R, -N(R)C(=Y‘)OR, -N(R)C(=Y')N(R)2, -so2nr2, -CN, —N3, -NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
each R is independently H, (Cj-Cg) alkyl, (Ci-Cg) substituted alkyl, (C2Cg)alkenyl, (C2-Cg) substituted alkenyl, (C2-Cg) alkynyl, (C2-Cg) substituted alkynyl, C6-C2o aryl, C6-C2o substituted aryl, C2-C2o heterocyclyl, C2-C2q substituted heterocyclyl, arylalkyl or substituted arylalkyl;
W3 is W4 or W5; W4 is R, -C(Yl)Ry, -C(Y’)W5, -SO2Ry, or -SO2W5; and W5 is a carbocycle or a heterocycle wherein Ws is independently substituted with 0 to 3 Ry groups;
each R8 is halogen, NRnR12, N^'W, NRuNRnR*2, N3, NO, NO2, CHO,
CN, -CH(=NRn), -CH=NNHRn, -CH=N(ORfl), -CH(ORn)2, -C(=O)NRR12,
-C(=S)NRr'2, -C(=O)OR, (Ci-Ci)alkyl, (C2-Cs)alkenyl, (C2-C8)alkynyl, (C4-Cs)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=O)(Ci-C8)alkyl, -StO^Cj-C^lkyl, aryl(Ci-Cg)alkyi, OR11 or SR11;
each R9 is independently H, halogen, NRllR12, N(R)ORn, NR'Wr12, N3, NO, NO2, CHO, CN, -CH(=NR]1), -CH=NHNRh, -CH =N(ORil), -CH(OR”)2, -C(=O)NRllR12, -C(=S)NRRI2,-C(=O)OR11, R11, OR11 or SR11; and each R1' or R12 is independently H, (Cj-C8)alkyl, (CL-Cx)alkenyl. (C2C8)alkynyl, (Q- C,s)carh<)cyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(C1-C8)alkyl, -S(O)n(CrC8)alkyl or aryl(CI-C8)alkyl; or Rn and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or -NRa-; and wherein each (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(Ci~C8)alkyl of each R6, R11 or R12 is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or ORa; and wherein one or more of the non-terminal carbon atoms of each said (Ci-Cg)alkyl may be optionally replaced with -O-, -S- or -NRa-.
In one embodiment of the method of treating a Paramyxovirîdae infection comprising administering a compound of Formula III, R6 of Formula III is N3, CN, halogen, (Ci-Cg)alkyl, (Cj-C8)substituted alkyl, (C2-C8)alkenyl, (C2~-C8)substituted alkenyl, (C2-C8)alkynyi, or (C2-C8)substituted alkynyl. In another aspect of this embodiment, R6 of Formula III is CN, methyl, ethenyl, or ethynyl. In another aspect of this embodiment, R6 of Formula III is CN. In another aspect of this embodiment, R6 of Formula III is methyl. In another aspect of this embodiment, R2 of Formula III îs ORL In another aspect of this embodiment, R2 of Formula III is OH. In another aspect of this embodiment, R2 of Formula III is F. In another aspect of this embodiment, R3 of Formula III is OH, -OC(=O)R’’, or -OC(=O)OR**. In another aspect of this embodiment, R of Formula III is OH. In another aspect of this embodiment, R of Formula III is NR1 lR12. In another aspect of this embodiment, R8 of Formula III is NH2. In another aspect of this embodiment, R of Formula III is OR . In another aspect of this embodiment, R8 of Formula III is OH. In another aspect of this embodiment, R9 of Formula III is H. In another aspect of this embodiment, R9 of
Formula III is NRl 'R12. In another aspect of this embodiment, R9 of Formula III is
NH2. In another aspect of this embodiment, R7 of Formula III is H, -C(=O)R11, C(=O)ORh or
O
II
w2 . In another aspect of this embodiment, R7 of Formula III is H. In another aspect of this embodiment, R7 of Formula III is
O
W2
In another embodiment of the method of treating a Paramyxoviridae infection comprising admînistering a compound of Formula III, R6 of Formula III is N3, CN, halogen, (Ci-Cg)alkyl, (C]-Cg)substituted alkyl, (C2-Cg)alkenyl, (C2-Cs)substituted alkenyl, (C2-Cg)alkynyl, or (C2-C8)substîtuted alkynyl and R8 is NH2. In another aspect of this embodiment, R6 of Formula III is CN, methyl, ethenyl, or ethynyl. In another aspect of this embodiment, R6 of Formula III is CN. In another aspect of this embodiment, R6 of Formula III is methyl. In another aspect of this embodiment, R2 of Formula III is ORa. In another aspect of this embodiment, R2 of Formula III is OH, OC(=O)R!’, or -OC(=O)ORl 1. In another aspect of this embodiment, R2of Formula III îs OH. In another aspect of this embodiment, R2 of Formula III is F. In another aspect ofthis embodiment, R3 of Formula III isOH, -OC^OJR11, or-OC(=O)ORU. In another aspect of this embodiment, R3 of Formula III is OH. In another aspect of this embodiment, R9 of Formula III is H. In another aspect of this embodiment, R9 of Formula III is NRllR12. In another aspect of this embodiment, R9 of Formula III is NH2. In another aspect of this embodiment, R7 of Formula III is H, -C(=O)R11, Cf-OjOR11 or yy' ο
II
W2 . In another aspect of this embodiment, R7 of Formula III is H. In another aspect of this embodiment, R7 of Formula III is
O
II
w2
In another embodiment of the method of treating a Paramyxoviridae infection comprising administering a compound of Formula III, R6 of Formula III îs CN, methyl, ethenyl, or ethynyl, R is NH2, and R is H. In another aspect of this embodiment, R of Formula III is CN. In another aspect of this embodiment, R6 of Formula III is methyl. In another aspect of this embodiment, R2 of Formula III is ORa. In another aspect of this embodiment, R2 of Formula III is OH, -OC(=O)Rn, or -OC(=O)OR' In another aspect of this embodiment, R2 of Formula III is OH. In another aspect of this embodiment, R2 of Formula III is F. In another aspect of this embodiment, R3 of Formula III is OH,-OC(=O)Rn, or-OC(=O)ORn. In another aspect of this embodiment, R3 of Formula III is OH. In another aspect of this embodiment, R7 of Formula III is H, -C(=O)Rn, -C(=O)ORI1 or
O
II
W2 . In another aspect of this embodiment, R7 of Formula III is H. In another aspect of this embodiment, R7 of Formula III is
O
II
w2
In another embodiment of the method of treating a Paramyxoviridae infection comprising administering a compound of Formula III, the Paramyxoviridae infection is caused by a Paramyxovirina virus. In another aspect of this embodiment, the
Paramyxovirina virus is a parainfluenza, measles or mumps virus. In another aspect of this embodiment, the Paramyxovirina virus is a Respirovirus virus. In another aspect of this embodiment, the Paramyxovirina virus is a type 1 or 3 Human parainfluenza virus.
In another embodiment of the method of treating a Paramyxoviridae infection comprising administering a compound of Formula III, the Paramyxoviridae infection is caused by a Pneumovirinae virus. In another aspect of this embodiment, the Pneumovirinae virus is a respiratory syncytial virus. In another aspect of this embodiment, the Pneumovirinae virus is a Human respiratory syncytial virus.
In one embodiment, provided is a compound of Formula IV:
R3 F
Formula IV or a pharmaceutically acceptable sait or ester, thereof;
wherein:
each R1 îs H or halogen;
each R3 or R5 is independently H, OR*, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Ci-Cg)alkyl, (Ct-CsJcarbocyclylalkyl, (C]-C8)substituted alkyl, (C2-C8)alkenyl, (C2-Cs)substituted alkenyl, (C2- Cx)alkynvl or (C2-Cg)substituted alkynyl;
R6 is ORa, N(Ra)2, N3, CN, S(O)nRa, -C(=O)RH, -C(=O)ORn, -C(=O)NRl,R12, C(=O)SR1], -S(O)R”, -S(O)2Rh, -S(O)(ORl,)> -SfOHOR11), --SO2NR,!Ri2, halogen, (Ci-Cs)alkyl, (C4--Cg)carbocyclylalkyl, (Ci-Cgjsubstituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-Cg)alkynyl, or (C2--Cg)substituted alkynyl;
each n is independently 0,1, or 2; yr' each Ra is independently H, (Ci-Cg)alkyl, (Cf-C^alkcnyl, (C;-(R)a!kynyl.
aryl(Cl-C8)alkyl, (C4-Q)carbocyclylalkyk -C(=O)Rn, -C(-O)OR. -C^NR1^12, C(=O)SR!l, -S(0)Rh, -S(O)2R11, -S(0)(0Rh), -S(O)2(OR11), or-SO2NRuR12;
R7 is H, -C(=O)R!1, -C(=0)0R, -C^NR1^'2, -C(=0)SRl], -S(O)R11, 5 S(0)2R, -S(O)(OR11), -S(O)2(OR11), -SOsNR1 'R12, or
Y
Il s
w2 each Y or Y1 is, independently, O, S, NR, +N(O)(R), N(OR), +N(O)(OR), or N-NR2;
W1 and W2, when taken together, are -Y3(C(Ry)2)3Y3-; or one of W1 or W2 together with either R3 or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of the Formula la:
M2
Formula la wherein:
each Y2 is independently a bond, O, CR2, NR, +N(O)(R), N(0R), +N(O)(OR),
N-NR2, s, S-S, S(O), or S(O)2;
each Y3 is independently O, S, or NR;
M2 is 0, 1 or 2;
each R* is independently Ry or the formula: yC'
each Mla, Mle, and Mld is independently 0 or l;
M12cis0, 1,2,3,4, 5, 6, 7, 8, 9, 10, 11 or 12;
each Ry is independently H, F, Cl, Br, I, OH, R, -C(=Y*)R, -C(=Y')OR, C^Y'Wh, -N(R)2, -*N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(OR), -S(O)2(OR), OC(=Y!)R, -OC(- Y')OR. -OC(=y’)(N(R)2), -SC(=Y’)R, -SC(=Y1)OR, SC(=Y‘)(N(R)2), -N(R)C(=y‘)R, -N(R)C(=Y‘)OR, -N(R)C(=V’)N(R)2, -SO2NR?. -CN, —N3, —NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
each R is independently H, (CpCg) alkyl, (Ci-C8) substituted alkyl, (C2Cg)alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl, (C2-C8) substituted alkynyl, C6-C?!! aryl, C6-C20 substituted aryl, C2-C2o heterocyclyl, C2-C2o substituted heterocyclyl, arylalkyl or substituted arylalkyl;
W3 is W4 or W5; W4 is R, -C(Y')Ry, -C(Y')WS, -SO2Ry, or -SO2W5; and Ws is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups;
each R8 is halogen, NRllR12, N(Rll)OR11, NRi,NR1IR12, N3, NO, NO2, CHO, CN,-CH(=NR), -CH=NNHR11,-CH=N(OR11), -CH(ORu)2, -C(=O)NRnR12, -C(=S)NRhR12, -C(=O)ORn, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C4-C8)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=O)(Ci-C8)alkyl, -S(O)n(Ci-C8)alkyl, aryl(C|-C8)alkyl, OR11 or SR11;
each R9 is independently H, halogen, NRnR12, N(R,l)ORu, NRllNR1]R12, N3, NO, NO2, CHO, CN, -CH(=NR' '), -CH=NHNR' ', -CH^NÎOR11), -CH(ORn)2, -C(=O)NRliR12,-C(=S)NRllRl2,-C(=O)ORll,R11,ORli or SR11;
each Rm or R!2 is independently H, (C]-C8)alkyl, (C2-Cg)alkenyl, (C2C8)alkynyl, (C4-Cg)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(C]-Cs)alkyl, -S(O)fl(C|-('s)alkyl or aryl(Cj-C8)alkyl; or Ru and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or -NRa-; and wherein each (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(Cj-C8)alkyl of each R3, R5, R6, R11 or R12 is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or OR'; and wherein one or more of the non-terminal carbon atoms of each said (Ci-Cg)alkyl may be optionally replaced with -O-, -S- or NRa-,
In one embodiment of the compound of Formula IV, R6 is N3, CN, halogen, (Ci-Cg)alkyl, (C|-C8)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl, or (C2-C8)substituted alkynyl. In another aspect of this embodiment, R6 is CN, methyl, ethenyl, or ethynyl. In another aspect of this embodiment, R6 is CN. In another aspect of this embodiment, R6 is methyl. In another aspect of this embodiment, R1 is H. In another aspect of this embodiment, R3 is OH, -OC(=O)R11, or OC(=O)OR . In another aspect of this embodiment, R is OH. In another aspect of this embodiment, R8 is NR1 'R12. In another aspect of this embodiment, R8 is NH2. In another aspect of this embodiment, R8 is OR1 In another aspect of this embodiment, R8 is OH. In another aspect of this embodiment, R9 is H. In another aspect of this embodiment, R9is NR]1R12. In another aspect of this embodiment, R9 is NH2. In another aspect of this embodiment, R7 is H, -C(=O)R11, -C(=O)OR1] or
. In another aspect of this embodiment, R is H. In another aspect ot this embodiment, R7 is ο
II
w2
In another embodiment of a compound of Formula IV, R6 is N3, CN, halogen, (C|-- Cx)alkyl, (CrCslsubstitutcd alkyl, (C2-Cg)alkenyl, (C?-Cg)substituted alkenyl, (C2-Cg)alkynyl, or (C? Cÿ)substitiited alkynyl and R8 is NH2. In another aspect of this embodiment, R6 is CN, methyl, ethenyl, or ethynyl. In another aspect of this embodiment, R6 is CN. In another aspect of this embodiment, R6 is methyl. In another aspect of this embodiment, R1 is H. In another aspect of this embodiment, R3 is OH, OC(=O)R11, or -OC(=O)OR11. ln another aspect of this embodiment, R3 is OH. In another aspect of this embodiment, R9 is H. In another aspect of this embodiment, R9 is NR1 'R12. In another aspect of this embodiment, R9is NH2. In another aspect of this embodiment, R7 is H, -C(=O)Rn, -C(=O)OR or
O
W2 . In another aspect of this embodiment, R7 is H. In another aspect of
T this embodiment, R is
O
II
w2
In another embodiment of the compound of Formula IV, R6 is CN, methyl, ethenyl, or ethynyl, R8 is NH2, and R9 is H. In another aspect of this embodiment, R1 is H. In another aspect of this embodiment, R6 is CN. In another aspect of this embodiment, R is methyl. In another aspect of this embodiment, R is OH, OC(=O)RU, or-OC(=O)ORn. In another aspect of this embodiment, R3is OH. In another aspect of this embodiment, R7 is H, -C(=O)RI 1, -C(=O)OR11 or
Ο
W2 . In another aspect of this embodiment, R7 îs H. In another aspect of this embodiment, R7 is
O
II
W2
In another embodiment, provided is a method of treating a Paramyxoviridae infection in a mammal in need thereof comprising administering a therapeutically effective amount of a compound of Formulas I-IV, wherein Ru or R12 is independently H, (Cj-Cs)alkyl, (CB-Qialkenyl, (C2-('8)alkynyL (C4~C8)carbocyclylalkyI, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(Ci-C8)alkyi, -S(O)n(CiC8)alkyl or aryl(Ci-C8)aIkyl. In another embodiment, R and R taken together with a nitrogen to which they are both attached, form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with O-, -S- or -NRa-. Therefore, by way of example and not limitation, the moiety NR1 lR13 can be represented by the heterocycles:
and the like.
In another embodiment, provided is a method of treating a Paramyxoviridae infection in a mammal în need thereof comprising administering a therapeutically effective amount of a compound of Formula I-IV, wherein each R3, R4, R5, R6, R11 or R12 îs, independently, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(CrCg)alkyl, wherein said (C[-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(C]-C8)alkyl are, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or ORa. Therefore, by way of example and not limitation, R3, R4, R5, R6, R11 or R12 could represent moieties such as -CH(NH2)CH3, -CH(OH)CH2CH3, CH(NH2)CH(CH3)2, -CH2CF3, -(CH2)2CH(N3)CH3, -(CH2)6NH2 and the like.
In another embodiment, provided is a method of treating a Paramyxoviridae infection in a mammal in need thereof comprising administering a therapeutically effective amount of a compound of Formula I-IV, wherein R3, R4, R5, R6, Rl ‘ or R12 is (Ci-Cjj)alkyl wherein one or more of the non-terminal carbon atoms of each said (Ct5 Cs)alkyl may be optionally replaced with -O-, -S- or -NRa-, Therefore, by way of example and not limitation, R , R , R , R , R or R could represent moîeties such as CH2OCH3, -CH2OCH2CH3, -CH2OCH(CH3)2, -CH2SCH3, -(CH2)6OCH3, (CH2)éN(CH3)2 and the like.
In another embodiment, provided is a method of treating a Paramyxoviridae infection in a sample comprising administering an effective amount of a compound of Formula I selected from the group consisting of:
or a pharmaceutically acceptable sait or ester thereof.
In another embodiment, provided is a compound of Formula IV that îs
pharmaceutically acceptable sait or ester thereof.
In another embodiment, provided is a compound of Formula l that is
DEFINITIONS
Unless stated otherwise, the following terms and phrases as used herein are intended to hâve the following meanings:
When trade names are used herein, applicants intend to independently include the tradename product and the active pharmaceutical ingredîent(s) of the tradename product.
As used herein, a compound of the invention or a compound of Formula I means a compound of Formula I or a pharmaceutically acceptable sait, thereof.
Simiiarly, with respect to isolatable intermediates, the phrase a compound of Formula (number) means a compound of that formula and pharmaceutically acceptable salts, thereof.
“Alkyl” is hydrocarbon containing normal, secondary, tertiary or cyclic carbon xa/'’- atoms. For example, an alkyl group can hâve l to 20 carbon atoms (i.e, Ci-C20 alkyl), l to 8 carbon atoms (i.e., Cj-Cg alkyl), or l to 6 carbon atoms (i.e., Cj-Cô alkyl). Examples of suitable alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, -CH2CHj), 1-propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1-butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-1 -propyl (i-Bu, ibutyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CI ΓΟΟ, 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CHî)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl-2-butyl (-C(CH3hCH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 3-methyl-1-butyl (-CH2CH2CH(CH3)2), 2-methyl-1-butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-CH(CH2CH3)CH(CH3)2),
2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3-dimethyI-2-butyl (-CH(CH3)C(CH3)3, and octyl (-(CH2)7CH3).
“Alkoxy” means a group having the formula O-alkyl. in which an alkyl group, as defined above, is attached to the parent molécule via an oxygen atom. The alkyl portion of an alkoxy group can hâve 1 to 20 carbon atoms (i.e., Ci-C2o alkoxy), 1 to 12 carbon atoms(i.e., Cj-Ci2 alkoxy), or 1 to 6 carbon atoms(r.e., Ci-Ce alkoxy). Examples of suitable alkoxy groups include, but are not limited to, methoxy (-O-CH3 or -OMe), ethoxy (-OCH2CH3 or -OEt), t-butoxy (-O-C(CH3)3 or -OtBu) and the like.
“Haloalkyl” is an alkyl group, as defined above, in which one or more hydrogen atoms of the alkyi group is replaced with a halogen atom. The alkyl portion of a haloalkyl group can hâve 1 to 20 carbon atoms (i.e., Cj-C2o haloalkyl), 1 to 12 carbon atoms)/.λ, C)-C|2 haloalkyl), or 1 to 6 carbon atoms((.e., Cj-Cô alkyl). Examples of suitable haloalkyl groups include, but are not limited to, -CF3, -CHF2, -CFH2, -CH2CF3, and the like.
“Alkenyl” is a hydrocarbon containing normal, secondary, tertîary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond. For example, an alkenyl group can hâve 2 to 20 carbon atoms (i.e., C2-C2o alkenyl), 2 to 8 carbon atoms (i.e., C2-Cg alkenyl), or 2 to 6 carbon atoms (i.e., C2-C6 alkenyl). Examples of suîtable alkenyl groups include, but are not limited to, ethylene or vinyl (-CH-CI f), allyl (-CH2CH =CH2), cyclopentenyl (-C5H7), and 5-hexenyl (-CH2CH2CH2CH2CH-CH2).
“Alkynyl” is a hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. For example, an alkynyl group can hâve 2 to 20 carbon atoms (i.e., C2-C2o alkynyl), 2 to 8 carbon atoms (i.e., C2-Ck alkyne,), or 2 to 6 carbon atoms (i.e., C^-Ct, alkynyl). Examples of suîtable alkynyl groups include, but are not limited to, acetylenic (-C=CH), propargyl (-CH?C--CH), and the like.
“Alkylene” refers to a saturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. For exampie, an alkylene group can hâve 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkylene radicals include, but are not limited to, methylene (-CH2-),
1,1-ethyl (-CH(CH3)-), 1,2-ethyl (-CH2CH2-), 1,1-propyl (-CH(CH2CH3)-), 1,2-propyl (-CH2CH(CH3)-), 1,3-propyl (-CH2CH2CH2-), 1,4-butyl (-CH2CH2CH2CH2-), and the like.
“Alkenylene” refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. For example, and alkenylene group can hâve 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkenylene radicals include, but are not limited to, 1,2ethylene (-CIHCH-).
“Alkynylene” refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. For example, an alkynylene group can hâve 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkynylene radicals include, but are not limited to, acetylene (-CX2-), propargyl l-CIbC-C-). and 4-pentynyl (-ClfCIfCli?C-C-).
“Amîno” refers generally to a nitrogen radical which can be considered a dérivative of ammonia, having the formula -N(X)2, where each “X” is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, etc. The hybridization of the nitrogen is approximately sp . Nonlimiting types of amino include -NH2, -N(alkyl)2, -NH(alkyl), -N(carbocyclyl)2, NH(carbocyclyl), -N(heterocyclyl)2, -NH(heterocyclyl), -N(aryl)2, -NH(aryl), N(alkyl)faryl), -N(alkyl)(heterocyclyl), -N(carbocyclyl)(heterocyclyl), N(aryl)(heteroaryl), -N(alkyl)(heteroaryl), etc. The terrn “alkylamino” refers to an amino group substituted with at least one alkyl group. Nonlimiting examples of amino groups include -NH2, -NH(CH3), -N(CH0?. -NH(CH2CH3), - N(CH2CH3)2, -NH(phenyl), N(phenyl)2, -NH(benzyl), -N(benzyl)2, etc. Substituted alkylamino refers generally to alkylamino groups, as defined above, in which at least one substituted alkyl, as defined herein, is attached to the amino nitrogen atom. Non-limiting examples of substituted alkylamino includes -NH(alkylene-C(O)-OH), -NH(alkylene-C(O)-O-alkyl), N(alkylene-C(O)-OH)2, -N(alkylene-C(O)-O-alkyl)2, etc.
“Aryl” means an aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring System. For example, an aryl group can hâve 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 10 carbon atoms. Typîcal aryl groups include, but are not limited to, radicals derived from benzene (e.g., phenyl), substituted benzene, naphthalene, anthracene, bîphenyl, and the like.
“Arylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2phenylethan-l-yl, naphthylmethyl, 2-naphthylethan-l-yl, naphthobenzyl,
2-naphthophenylethan-l-yI and the like. The arylalkyl group can comprise 7 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
“Arylalkenyl” refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, but also an sp carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkenyl can include, for example, any of the aryl groups disclosed herein, and the alkenyl portion of γ/ the arylalkenyl can include, for example, any of the alkenyl groups disclosed herein.
The arylalkenyl group can comprise 8 to 20 carbon atoms, e.g., the alkenyl moiety is 2 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
“Arylalkynyl” refers to an acyclic alkynyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, but also an sp carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkynyl can include, for example, any of the aryl groups disclosed herein, and the alkynyl portion of the arylalkynyl can include, for example, any of the alkynyl groups disclosed herein. The arylalkynyl group can comprise 8 to 20 carbon atoms, e.g., the alkynyl moiety is 2 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms.
The term “substituted” in reference to alkyl, alkylene, aryl, arylalkyl, alkoxy, heterocyclyl, heteroaryl, carbocyclyl, etc., for example, “substituted alkyl”, “substituted alkylene”, “substituted aryl”, “substituted arylalkyl”, “substituted heterocyclyl”, and “substituted carbocyclyl” means alkyl, alkylene, aryl, arylalkyl, heterocyclyl, carbocyclyl respectîvely, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent. Typical substituents include, but are not limited to, -X, -Rb, -O, =O, -ORb, -SRb, -S , -NRb2, -N R\. =NRh, -CX3, -CN, -OCN, -SCN, -N=C=O, -NCS, -NO, -NO2, =N2, -N3, -NHC(=O)Rb, -OC(=O)Rb, -NHC(=O)NRb2, -S(=O)2-, -S(=O)2OH, -S(=O)2Rb, -OS(=O)2ORb, -8(Ο)2ΝκΛ. -S(=O)Rb, -OP(=O)(ORb)2,-P(=OXORb)2, -P(=O)(O )2, -P(=O)(OH)2, -P(O)(ORb)(O ), -C(=O)Rb, -C(=O)X, -C(S)Rb, -C(O)ORb, -C(O)O , -C(S)ORb, -C(O)SRb, -C(S)SRb, -C(O)NRb2, -C(S)NRb2, -C(=NRb)NRb2, where each X is independently a halogen: F, Cl, Br, or I; and each Rb is independently H, alkyl, aryl, arylalkyl, a heterocycle, or a protecting group or prodrug moiety. Alkylene, alkenylene, and alkynylene groups may also be similarly substituted. Unless otherwîse indicated, when the term substituted is used in conjunction with groups such as arylalkyl, which have two or more moieties capable of substitution, the substituents can be attached to the aryl moiety, the alkyl moiety, or both.
The term “prodrug” as used herein refers to any cornpound that when administered to a biological system generates the drug substance, i.e., active ingrédient, as a resuit of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, < / and/or metabolic chemical reaction(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically active compound.
One skilled in the art will recognize that substituents and other moieties of the compounds of Formula l-IV should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated into an acceptably stable pharmaceutical composition. Compounds of Formula I-IV which hâve such stability are contemplated as falling within the scope of the présent invention.
“Heteroalkyl” refers to an alkyl group where one or more carbon atoms hâve been replaced with a heteroatom, such as, O, N, or S. For example, if the carbon atom of the alkyl group which is attached to the parent molécule is replaced with a heteroatom (e.g., O, N, or S) the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., -OCH3, etc ), an amine (e.g., -NHCH3, -N(CH3)2, etc.), or a thioalkyl group (e.g., -SCH3). If a non-terminal carbon atom of the alkyl group which is not attached to the parent molécule is replaced with a heteroatom (e.g., O, N, or S) the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g., -CH2CH2-O-CH3, etc.), an alkyl amine (e.g., -CH2NHCH3, -CH2N(CH3)2, etc.), or a thioalkyl ether (e.g.,-CH2-S-CH3). If a terminal carbon atom of the alkyl group is replaced with a heteroatom (e.g., O, N, or S), the resulting heteroalkyl groups are, respectively, a hydroxyalkyl group (e.g., -CH2CH2-OH), an aminoalkyl group (e.g., -CH2NH2), or an alkyl thiol group (e.g., -CH2CH2-SH). A heteroalkyl group can hâve, for example, l to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. A Ci-Cg heteroalkyl group means a heteroalkyl group having 1 to 6 carbon atoms.
“Heterocycle” or “heterocyclyl” as used herein includes by way of example and not limitation those heterocycles described in Paquette, Léo A.; Principles of Modem Heterocyclic Chemistry (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Sériés of Monographs” (John Wiley & Sons, New York, 1950 to présent), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. In one spécifie embodiment of the invention “heterocycle” includes a “carbocycle” as defined herein, wherein one or more (e.g. 1, 2, 3, or 4) carbon atoms hâve been replaced with a heteroatom (e.g. O, N, or S). u/
The terms “heterocycle” or “heterocyclyl” includes saturated rings, partially unsaturated rings, and aromatic rings (i.e., heteroaromatic rings). Substituted heterocyclyls include, for example, heterocyclic rings substituted with any of the substituents disclosed herein including carbonyl groups. A non-limiting example of a carbonyl substituted heterocyclyl is:
O
Examples of heterocycles include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolînyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-l,2,5-thiadiazinyl, 2H,6H-l,5,2-dithiazinyI, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H-pyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoîndolyl, 3H-indolyl, lH-indazoly, purinyl, 4H-quinolizinyl, phthalazînyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, β-carbolînyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolînyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, isatinoyl, and bis-tetrahydrofuranyl:
By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3,4, 5, or 6 of a pyridine, position 3,4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3,4, or 5 of an isoxazole, pyrazole, or îsothîazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3,4, 5,6, 7, or 8 of a quinoline or position 1,3,4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl,
2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimîdinyl, 2-pyrazinyl, 3-pyrazinyl,
5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrrolîne, 3-pyrroline, imidazole, imîdazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl, and 1-piperidinyl.
“Heterocyclylalkyl” refers to an acyclic alkyl radical în which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl-alkylene- moiety). Typical heterocyclyl alkyl groups include, but are not limited to heterocyclyl-CH2-, 2(heterocyclyl)ethan-I-yl, and the like, wherein the “heterocyclyl” portion includes any of the heterocyclyl groups described above, including those described in Principles of Modem Heterocyclic Chemistry. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkyl portion of the heterocyclyl alkyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkyl group comprises 3 to 20 carbon atoms, e.g., the alkyl portion of the arylalkyl group is 1 to 6 carbon atoms and the heterocyclyl moiety is 2 to 14 carbon atoms. Examples of heterocyclylalkyls include by way of example and not limitation 5-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as thiazolylmethyl, 2-thiazolylethan-l-yl, imidazolylmethyl, oxazolylmethyl, thiadiazolylmethyl, etc., 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridînylmethyl, pyridizylmethyl, pyrimidylmethyl, pyrazinylmethyl, etc.
“Heterocyclylalkenyl” refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp carbon atom, but also a sp2 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclylalkenyl ene- moiety). The heterocyclyl portion of the heterocyclyl alkenyl group includes any of the heterocyclyl groups described herein, including those described in Principles of Modem Heterocyclic Chemistry, and the alkenyl portion of the heterocyclyl alkenyl group includes any of the alkenyl groups disclosed herein. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkenyl portion of the heterocyclyl alkenyl by means of a carbon-carbon bond or a carbon-hetero atom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkenyl group comprises 4 to 20 carbon atoms, e.g., the alkenyl portion of the heterocyclyl alkenyl group is 2 to 6 carbon atoms and the heterocyclyl moiety is 2 to 14 carbon atoms.
“Heterocyclylalkynyl” refers to an acyclic alkynyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclylalkynylene- moiety). The heterocyclyl portion of the heterocyclyl alkynyl group includes any of the heterocyclyl groups described herein, including those described in Principles of Modem Heterocyclic Chemistry, and the alkynyl portion of the heterocyclyl alkynyl group includes any of the alkynyl groups disclosed herein. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkynyl portion of the heterocyclyl alkynyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkynyl group comprises 4 to 20 carbon atoms, e.g., the alkynyl portion of the heterocyclyl alkynyl group is 2 to 6 carbon atoms and the heterocyclyl moiety is 2 to 14 carbon atoms.
“Heteroaryl” refers to an aromatic heterocyclyl having at least one heteroatom in the ring. Non-limiting examples of suitable heteroatoms which can be included in the aromatic ring include oxygen, sulfur, and nitrogen. Non-limiting examples of vv' heteroaryl rings include ail of those aromatic rings listed in the définition of “heterocyclyl”, including pyridinyl, pyrrolyl, oxazolyl, indolyl, isoindolyl, purinyl, furanyl, thienyl, benzofuranyl, benzothiophenyl, carbazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, quinolyl, isoquinolyl, pyridazyl, pyrimidyl, pyrazyl, etc.
“Carbocycle” or “carbocyclyl” refers to a saturated (i.e., cycloalkyl), partially unsaturated (e.g., cycloakenyl, cycloalkadienyl, etc.) or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle. Monocyclic carbocycles hâve 3 to 7 ring atoms, still more typîcally 5 or 6 ring atoms. Bicyclic carbocycles hâve 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] System, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] System, or spîro-fused rings. Non-limiting examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1 -cyclopent-1 enyl, 1 -cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-l-enyl, 1cyclohex-2-enyl, l-cyclohex-3-enyl, and phenyl. Non-limiting examples of bicyclo carbocycles includes naphthyl, tetrahydronapthalene, and decaline.
“Carbocyclylalkyl” refers to to an acyclic akyl radical in which one of the hydrogen atoms bonded to a carbon atom is replaced with a carbocyclyl radical as described herein. Typical, but non-limiting, examples of carbocyclylalkyl groups include cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl and cyclohexylmethyl.
“Arylheteroalkyl” refers to a heteroalkyl as defined herein, in which a hydrogen atom (which may be attached either to a carbon atom or a heteroatom) has been replaced with an aryl group as defined herein. The aryl groups may be bonded to a carbon atom of the heteroalkyl group, or to a heteroatom of the heteroalkyl group, provided that the resulting arylheteroalkyl group provides a chemically stable moiety. For example, an arylheteroalkyl group can hâve the general formulae -alkylene-O-aryl, -alkylene-O-alkylene-aryl, -alkylene-NH-aryl, -alkylene-NH-alkylene-aryl, -alkyleneS-aryl, -alkylene-S-alkylene-aryl, etc. In addition, any of the alkylene moieties in the general formulae above can be further substituted with any of the substituents defined or exemplified herein.
“Heteroarylalkyl” refers to an alkyl group, as defined herein, in which a hydrogen atom has been replaced with a heteroaryl group as defined herein. Nonlimîting examples of heteroaryl alkyl include -CH2-pyridinyl, -CH2-pyrrolyI, -CH2-oxazolyl, -CH2-indolyl, -CH2-isoindolyl, -ClC-purinyi. -CH2-furanyl, -CH2-thienyl, -CH2-benzoiïiranyl, -CH2-benzothiophenyl, -CH2-carbazolyl, -CH2-îmidazolyl, -CH2-thiazolyl, -CH2-isoxazolyl, -CH2-pyrazolyl, -CH2-isothiazolyl, -CH2-quînolyl, -CH2-isoquinolyl, -CH2-pyridazyl, -CH2-pyrimidyl, -CH2-pyrazyl, -CH(CH3)-pyridinyl, -CH(CH3)-pyrrolyl, -CH(CH3)-oxazolyl, -CH(CH3)-indolyl, -CH(CH3)-isoindolyl, -CH(CH3)-purinyl, -CH(CH3)-furanyl, -CH(CH3)-thienyl, -CH(CH3)-benzofuranyl, -CH(CH3)-benzothiophenyl, -CH(CH3)-carbazolyl, -CH(CH3)-imidazolyl, -CH(CH3)-thiazolyl, -CH(CH3)-isoxazolyl, -CH(CH3)-pyrazolyl, -CH(CH3)-îsothîazolyl, -CH(CH3)-quinolyl, -CH(CH3)-isoquinolyl, -CH(CH3)-pyridazyl, -CH(CH3)-pyrimidyl, -CH(CH3)-pyrazyl, etc.
The term “optionally substituted” in reference to a particular moiety of the compound of Formula I-III (e.g., an optionally substituted aryl group) refers to a moiety wherein ail substiutents are hydrogen or wherein one or more of the hydrogens of the moiety may be replaced by substituents such as those listed under the définition of “substituted”.
The term “optionally replaced” in reference to a particular moiety of the compound of Formula I-III (e.g., the carbon atoms of said (Ci-Cg)alkyl may be optionally replaced by -O-, -S-, or -NRa-) means that one or more of the methylene groups of the (C]-C8)alkyl may be replaced by 0, 1, 2, or more of the groups specified (e.g.,-O-, -S-, or-NRa-).
The term “non-terminal carbon atom(s)” in reference to an alkyl, alkenyl, alkynyl, alkylene, alkenylene, or alkynylene moiety refers to the carbon atoms in the moiety that intervene between the first carbon atom of the moiety and the last carbon atom in the moiety. Therefore, by way of example and not limitation, in the alkyl moiety -CH2(C*)H2(C*)H2CH3 or alkylene moiety -CH2(C*)H2(C*)H2CH2- the C’ atoms would be considered to be the non-terminal carbon atoms.
Certain Y and Y1 alternatives are nitrogen oxides such as *N(O)(R) or +N(O)(OR). These nitrogen oxides, as shown here attached to a carbon atom, can also
be represented by charge separated groups such as R or
respectively, and are intended to be équivalent to the aforementioned représentations for the purposes of describing this invention.
Linker or “link” means a chemical moiety comprising a covalent bond or a chain of atoms. Linkers include repeating units of alkyloxy (e.g. polyethyleneoxy, PEG, polymethyleneoxy) and alkylamino (e.g. polyethyleneamino, Jeffamine™); and diacîd ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
The terms such as “oxygen-linked”, “nitrogen-linked”, “carbon-linked”, “sulfurlinked”, or “phosphorous-linked” mean that if a bond between two moieties can be formed by using more than one type of atom in a moiety, then the bond formed between the moieties is through the atom specified. For example, a nitrogen-linked amino acid would be bonded through a nitrogen atom of the amino acid rather than through an oxygen or carbon atom of the amino acid.
In some embodiments of the compounds of Formula I-IV, one or more of W1 or W2 are independently a radical of a nitrogen-linked naturally occurring α-amino acid ester. Examples of naturally occurring amino acids include isoleucine, leucîne, lysine, méthionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, arginine, histidine, omithine and taurine. The esters of these amino acids comprise any of those described for the substitutent R, particularly those in which R is optionally substituted (Ci-Cg)alkyl.
The term “purine” or “pyrimidine” base comprises, but is not limited to, adenine, N6-alkylpurines, N6-acylpurines (wherein acyl is C(O)(alkyl, aryl, alkylaryl, or arylalkyl), N6-benzylpurine, N6-halopurine, N6-vinylpurine, N6-acetylenic purine, N6acyl purine, N6-hydroxyalkyl purine, N6-allylaminopurine, N6-thioallyl purine, N2alkylpurines, N2-alkyl-6-thîopurines, thymine, cytosine, 5-fluorocytosine, 5methylcytosine, 6-azapyrimidine, including 6-azacytosine, 2- and/or 4-
mercaptopyrmidine, uracil, 5-halouracil, including 5-fluorouracil, Cs-alkylpyrimidines,
C5-benzylpyrimidines, C5-halopyrimidines, C5-vinylpyrimîdine, C5-acetylenîc pyrimidine, C5-acyI pyrimidine, C5-hydroxyalkyl purine, Cs-amidopyrimidine, C5cyanopyrimidine, C5-5-iodopyrimidîne, C6-iodo-pyrimidine, C5-Br-vînyl pyrimidine,
C’-Br-vmyl pyriniîdine, C5-nitropyrimidine, C5-ammo-pyrimidine, N2-alkylpurines,
N2-alkyl-6-thîopurines, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl. Purine bases include, but are not limited to, guanine, adenine, hypoxanthine, 2,6-diaminopurme, and 6chloropurine. The purine and pyrimidine bases of Formula I-III are linked to the ribose 10 sugar, or analog thereof, through a nitrogen atom of the base. Functional oxygen and nitrogen groups on the base can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl, dimethylhexylsilyl, t-butyldimethylsilyl, and t-butyldiphenylsîlyl, trityl, alkyl groups, and acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
Unless otherwise specified, the carbon atoms of the compounds of Formula I-IV are intended to hâve a valence of four. In some chemical structure représentations where carbon atoms do not hâve a sufficient number of variables attached to produce a valence of four, the remaining carbon substitutents needed to provide a valence of four should be assumed to be hydrogen. For example,
“Protectîng group” refers to a moiety of a compound that masks or alters the properties of a functional group or the properties of the compound as a whole. The chemical substructure of a protectîng group varies widely. One function of a protectîng group is to serve as an intermediate în the synthesis of the parental drug substance. Chemical protectîng groups and strategies for protection/deprotectîon are well known in the art. See: Protective Groups in Organic Chemistry, Theodora W. Greene (John Wiley & Sons, Inc., New York, 1991. Protectîng groups are often utilized to mask the reactivîty of certain functional groups, to assist in the efficîency of desired chemical reactions, e.g. makîng and breaking chemical bonds in an ordered and planned fashion. Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties which can be measured by common analytical tools. Chemically protected intermediates may themselves be bîologically active or inactive, •of''
Protected compounds may also cxhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and résistance to enzymatic dégradation or séquestration. In this rôle, protected compounds with intended therapeutic effects may be referred to as prodrugs. Another function of a protecting group is to couvert the parental drug into a prodrug, whereby the parental drug is released upon conversion of the prodrug in vivo. Because active prodrugs may be absorbed more effectîvely than the parental drug, prodrugs may possess greater potency in vivo than the parental drug. Protecting groups are removed either in vitro, in the instance of chemical intermediates, or in vivo, in the case of prodrugs. With chemical intermediates, it is not partîcularly important that the resulting products after deprotection, e.g. alcohols, be physiologically acceptable, although in general it is more désirable if the products are pharmacologically innocuous.
“Prodrug moiety” means a labile functional group which séparâtes from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, “Design and Application of Prodrugs” in Textbook of Drug Design and Development ( 1991 ), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academie Publishers, ρρ. 113191 ). Enzymes which are capable of an enzymatic activation mechanism with the phosphonate prodrug compounds of the invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases. Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimize drug delivery, bioavailability and efficacy.
A prodrug moiety may include an active métabolite or drug itself.
Exemplary prodrug moieties include the hydrolytically sensitive or labile acyloxymethyl esters CH2OC( ())R and acyloxymethyl carbonates -CH2OC(=O)OR30 where R30 îs C|-C() alkyl, C|-C7, substituted alkyl, Cfc -C2o aryl or C6-C20 substituted aryl. The acyloxyalkyl ester was used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al (1983) J. Pharm. Soi. 72: 324; also US Patent Nos. 4816570,4968788, 5663159 and 5792756. In certain compounds of the invention, a prodrug moiety is part of a phosphate group. The acyloxyalkyl ester may be used to deliver phosphoric acids \V across cell membranes and to enhance oral bioavailability. A close variant of the acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bioavailability as a prodrug moiety in the compounds of the combinations of the invention. An exemplary acyloxymethyl ester is pivaloyloxymethoxy, (POM) -CH2OC(=O)C(CH3)3. An exemplary acyloxymethyl carbonate prodrug moiety is pi valoyloxym ethyl carbonate (POC) -CH2OC(=O)OC(CH3)3.
The phosphate group may be a phosphate prodrug moiety. The prodrug moiety may be sensitive to hydrolysis, such as, but not limited to those comprising a pivaloyloxymethyl carbonate (POC) or POM group. Altemativcly, the prodrug moiety may be sensitive to enzymatic potentiated cleavage, such as a lactate ester or a phosphonamidate-ester group.
Aryl esters of phosphorus groups, especially phenyl esters, are reported to enhance oral bioavailability (DeLambert et al (1994) J. Med. Chem. 37: 498). Phenyl esters containing a carboxylic ester ortho to the phosphate hâve also been described (Khamnei and Torrence, (1996) J. Med. Chem. 39:4109-4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ortho-or pnra-position may accelerate the hydrolysis. Benzyl analogs with an acylated phénol or an alkylated phénol may generate the phenolic compound through the action of enzymes, e.g. esterases, oxidases, etc., which in tum undergoes cleavage at the benzylic C-0 bond to generate the phosphoric acid and the quinone methide intermediate. Examples of this class of prodrugs are described by Mitchell et ai (1992) J. Chem. Soc. Perkin Trans. 72345; Brook et al WO 91/19721. Still other benzylic prodrugs hâve been described containing a carboxylic ester-containing group attached to the benzylic methylene (Glazier et al WO 91/19721). Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphonate drugs. These proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide. Deesterification or réduction of the disulfide generates the free thio intermediate which subsequently breaks down to the phosphoric acid and cpisulfide (Puech et al (1993) Antiviral Res., 22: 155-174; Benzaria et al (1996) J. Med. Chem. 39: 4958). Cyclic phosphonate esters hâve also been described as prodrugs of phosphorus-containing compounds (Erion et al, --42
US Patent No. 6312662).
It is to be noted that ail enantiomers, dîastereomers, and racemic mixtures, tautomers, polymorphs, pseudopolymorphs of compounds within the scope of Formula
I-IV and pharmaceutically acceptable salts thereof are embraced by the présent invention. Ail mixtures of such enantiomers and dîastereomers are within the scope of the présent invention.
A compound of Formula I-IV and its pharmaceutically acceptable salts may exist as different polymorphs or pseudopolymorphs. As used herein, crystalline polymorphism means the ability of a crystalline compound to exist in different crystal structures. The crystalline polymorphism may resuit from différences in crystal packing (packing polymorphism) or différences in packing between different conformera of the same molécule (conformational polymorphism). As used herein, crystalline pseudopolymorphism means the ability of a hydrate or solvaté of a compound to exist in different crystal structures. The pseudopolymorphs of the instant invention may exist due to différences in crystal packing (packing pseudopolymorphism) or due to différences in packing between different conformera of the same molécule (conformational pseudopolymorphism). The instant invention comprises ail polymorphs and pseudopolymorphs of the compounds of Formula I-III and their pharmaceutically acceptable salts.
A compound of Formula I-IV and its pharmaceutically acceptable salts may also exist as an amorphous solid. As used herein, an amorphous solid is a solid in which there is no long-range order of the positions of the atoms in the solid. This définition applîes as well when the crystal size is two nanometers or less. Additives, including solvents, may be used to create the amorphous forms of the instant invention. The instant invention comprises ail amorphous forms of the compounds of Formula IIV and their pharmaceutically acceptable salts.
Selected substituents comprising the compounds of Formula I-IV are présent to a recursive degree. In this context, “recursive substituent” means that a substituent may recite another instance of itself. Because of the recursive nature of such substituents, theoretically, a large number of compounds may be présent in any given embodiment. For example, R* comprises a Ry substituent. Ry can be R. R can be W3. W3 can be W4 and W4 can be R or comprise substituents comprising Ry. One of ordinary skill in the art of médicinal chemistry understands that the total number of such substituents is reasonably limited by the desired properties of the compound intended. Such properties include, by way of example and not limitation, physical properties such as molecular weight, solubility or log P, application properties such as activity against the intended target, and practical properties such as ease of synthesis.
By way of example and not limitation, W3 and Ry are recursive substituents in certain embodiments. Typically, each recursive substituent can independently occur 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7,6, 5,4, 3, 2, 1, or 0, times in a given embodiment. More typically, each recursive substituent can independently occur 12 or fewer times in a given embodiment. Even more typically, each recursive substituent can independently occur 3 or fewer times in a given embodiment. For example, W3 will occur 0 to 8 times, Ry will occur 0 to 6 times in a given embodiment. Even more typically, W3 will occur 0 to 6 times and Ry will occur 0 to 4 times in a given embodiment.
Recursive substituents are an intended aspect of the invention. One of ordinary skill in the art of médicinal chemistry understands the versatility of such substituents. To the degree that recursive substituents are présent in an embodiment of the invention, the total number will be determined as set forth above.
The modifier about used in connection with a quantity is inclusive of the stated value and has the meaning dictated by the context (e.g., includes the degree of enor associated with measurement of the particular quantity).
The term “treating”, as used herein, unless otherwise indicated, means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term “treatment”, as used herein, refers to the act of treating, as “treating” is defined immediately above.
The term “therapeutically effective amount”, as used herein, is the amount of compound of Formula I-IV présent in a composition described herein that is needed to provide a desired level of drug în the sécrétions and tissues of the airways and lungs, or alternatively, în the bloodstream of a subject to be treated to give an anticipated physiological response or desired biological effect when such a composition is administered by the chosen route of administration. The précisé amount will dépend upon numerous factors, for example the particular compound of Formula I-IV, the spécifie activity of the composition, the delivery device employed, the physical characteristics of the composition, its intended use, as well as patient considérations such as severity of the disease state, patient coopération, etc., and can readily be determined by one skilled in the art based upon the information provided herein.
The term “normal saline” means a water solution containing 0.9% (w/v) NaCl. The term “hypertonie saline” means a water solution containing greater than 0.9% (w/v) NaCl. For example, 3% hypertonie saline would contain 3% (w/v) NaCl.
The compounds of the Formula I-IV may comprise a phosphate group as R7,
Y
Il s
which may be a prodrug moiety w wherein each Y or Yl is, independently, O, S, NR, 'N(O)(R). N(OR), +N(O)(OR), or N-NR2; W1 and W2, when taken together, are -Y3(C(Ry)2)rY3-; or one of W1 or W2 together with either R3or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of Formula la:
M2 wherein:
each Y2 is independently a bond, O, CR2, NR, +N(O)(R), N(OR), 'N(O)(OR), N-NR2, S, S-S, S(O), or S(O)2;
Ί each Y is independently O, S, or NR;
M2 îs 0, 1 or 2;
each Ry is independently H, F, Cl, Br, I, OH, R, -C(=Yl)R, -C(=Y‘)OR, C(=Yl)N(R)2, -N(R)2, -+N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(0R), -S(O)2(OR), OC(=Y*)R, -OC(=Y')OR, -OC(=Y')(N(R)2), -SC(=Y')R, -SC(=Y')OR, SC(=y')(N(R)2), -N(R)C(=Yl)R, -N(R)C(=y‘)OR, or-N(R)C(=Y')N(R)2, -SO2NR2, -CN, —N3, -NO2, -OR, a protecting group or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
each R* is independently Ry, a protecting group, or the formula:
wherein:
Mla, Mlc, and Mld are independently 0 or l;
M12cis0, 1,2,3,4, 5,6, 7, 8, 9, 10, 11 or 12;
each R is H, halogen, (Ci-C8) alkyl, (C|-C8) substituted alkyl, (C2-C8) alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl, (C2-C8) substituted alkynyl, C6-C2o aryl, C(,-C2o substituted aryl, C2-C20 heterocycle, C2-C20 substituted heterocyclyl, arylalkyl, substituted arylalkyl or a protecting group;
W3 is W4 or W5; W4 is R, -C(Y')Ry, -C(Y')W5, -SO2Ry, or -SO2W5; and W5 is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups.
Ws carbocycles and W5 heterocycles may be independently substituted with 0 to 3 Ry groups. W5 may be a saturated, unsaturated or aromatic ring comprising a monoor bicyclic carbocycle or heterocycle. W5 may hâve 3 to 10 ring atoms, e.g., 3 to 7 ring atoms. The W5 rings are saturated when containing 3 ring atoms, saturated or monounsaturated when containing 4 ring atoms, saturated, or mono- or di-unsaturated when containing 5 ring atoms, and saturated, mono- or di-unsaturated, or aromatic when containing 6 ring atoms.
A W5 heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and l to 3 heteroatoms selected from N, O, P, and S) or a bicycle having to 10 ring members (4 to 9 carbon atoms and l to 3 heteroatoms selected from N, O,
P, and S). W5 heterocyclic monocycles may hâve 3 to 6 ring atoms (2 to 5 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S); or 5 or 6 ring atoms (3 to 5 carbon atoms and 1 to 2 heteroatoms selected from N and S). W5 heterocyclic bicycles hâve 7 to 10 ring atoms (6 to 9 carbon atoms and 1 to 2 heteroatoms selected from N, O, and S) arranged as a bicyclo [4,5], [5,5], [5,6], or [6,6] System; or 9 to 10 ring atoms (8 to 9 carbon atoms and 1 to 2 hetero atoms selected from N and S) arranged as a bicyclo [5,6] or [6,6] System. The W5 heterocycle may be bonded to Y2 through a carbon, nitrogen, sulfiir or other atom by a stable covalent bond.
W5 heterocycles include for example, pyridyl, dihydropyridyl isomers, piperidine, pyrîdazinyl, pyrimidinyl, pyrazinyl, s-triazinyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, furanyl, thiofuranyl, thienyl, and pyrrolyl. W5 also includes, but is not limited to, examples such as:
W5 carbocycles and heterocycles may be independently substituted with 0 to 3 R groups, as defined above. For example, substituted W5 carbocycles include:
OH
5 Exa^csofsubsù.u.edphenyicarbocydes
HN—\ r p0
X /O'V
X
NH2 nh2
O —nh2 oz
NH
-NH2 oz _
Y
II
Embodiments of of Formula I-IV compounds include substructures such as:
2b wherein each Y is, independently, O or N(R). In another aspect of this embodiment, each Y2b is O and each Rx is independently:
•J wherein M12c is l, 2 or 3 and each Y is independently a bond, O, CR2, or S. In another aspect of this embodiment, one Y2b-R* is NH(R) and the other Y2b-Rx is O-Rx wherein Rx is:
wherein Ml2c is 2. In another aspect of this embodiment, each Y2b is O and each R* is independently:
M12c wherein Ml2c is 2. In another aspect of this embodiment, each Y2b is O and each Rx is independently:
wherein Μ12c is l and Y is a bond, O, or CR2.
Other embodiments of
Y
II
w2 of Formulas I-IV compounds include substructures such as:
wherein each Y3 is, independently, O or N(R). In another aspect of this embodiment, each Y3 is O. In another aspect ofthis embodiment, the substructure is:
wherein Ry is W5 as defîned herein.
Another embodiment of
Y
II
w2 of Formula I-IV includes the substructures:
wherein each Y2c is, independently, O, N(Ry) or S.
Another embodiment of
Y
II
of Formula I-IV compounds includes the substructures wherein one of Wl or W2 together with either R3 or R4 is -Y3- and the
2 » other ofW orW is Formula la. Such an embodiment is represented by a compound of Formula Ib selected from:
Formula Ib
In another aspect ofthe embodiment of Formula Ib, each Y and Y3 is O. In another aspect of the embodiment of Formula Ib, W1 or W2 is Y2b-R*; each Y, Y3 and Y2b is O and R* is: >vs/
wherein Μ12c is l, 2 or 3 and each Y is independently a bond, O, CR2, or S. In another aspect of the embodiment of Formula Ib, W1 or W2 is Y2b-Rx; each Y, Y3 and Y2b is O and R* is:
wherein Μ12c is 2. In another aspect of the embodiment of Formula Ib, W1 or W2 is Y21’-R?; each Y, Y3 and Y2b is O and R* is:
wherein Μ12c is 1 and Y2 is a bond, O, or CR2.
Y
II
Another embodiment of a substructure:
of Formula I-IV compounds includes
wherein W5 is a carbocycle such as phenyl or substituted phenyl. In another aspect of this embodiment, the substructure is:
2b wherein Y is O or N(R) and the phenyl carbocycle is substituted with 0 to 3 R groups.
In another aspect of this embodiment of the substructure, R* is:
wherein Ml2c is 1, 2 or 3 and each Y is independently a bond, O, CR2, or S.
Another embodiment of
of Formula I-IV includes substructures:
The chiral carbon of the amino acid and lactate moieties may be either the R or S configuration or the racemic mixture.
Y
Hk p1 w1-/’
Another embodiment of W2 of Formula I-IV îs substructure
wherein each Y2 is, independently, -O- or -NH-. In another aspect of this embodiment, Ry is (C]-Cg) alkyl, (C]-C8) substituted alkyl, (C2-C8) alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl or (C2-C8) substituted alkynyl. In another aspect of this embodiment, Ry is (C|-C8) alkyl, (C]-C8) substituted alkyl, (C2-C8) alkenyl, (C2-C8) 10 substituted alkenyl, (C2-C8) alkynyl or (C2-C8) substituted alkynyl; and R is CH3. In another aspect of this embodiment, Ry is (Ci-C8) alkyl, (C|-C8) substituted alkyl, (C2C8) alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl or (C2-C8) substituted alkynyl;
R is CH3; and each Y is -NH-. In another aspect of this embodiment, W and W are, independently, nitrogen-linked, naturally occurring amino acids or naturally occurring amino acid esters. In another aspect of this embodiment, W and W are, independently, naturally-occurring 2-hydroxy carboxylic acids or naturally-occurring 2hydroxy carboxylic acid esters wherein the acid or ester is linked to P through the 2hydroxy group. iaX
Another embodiment of
Y
II
of Formula I-IV is substructure:
O
In one aspect of this embodiment, each Rx is, independently, (Ci-C8) alkyl. In another aspect of this embodiment, each R* is, independently, C6-C20 aryl or C6-C20 substituted 5 aryl.
In a preferred embodiment,
Another embodiment of
Y
of Formulas I-IV is substructure
O
W2
7 wherein W and W are independently selected from one of the formulas in Tables
20.1-20.37 and Table 30.1 below. The variables used in Tables 20.1-20.37 (e.g., W23, R21, etc.) pertaîn only to Tables 20.1-20.37, unless otherwise indicated.
The variables used in Tables 20.1 to 20.37 hâve the following définitions:
each R21 is independently H or (C]-C8)alkyl;
each R is independently H, R , R or R wherein each R is independently substituted with 0 to 3 R23;
each R23 is independently R23a, R23b, R23c or R23d, provided that when R23 is bound to a heteroatom, then R33 is R23c or R23d;
each R23a is independently F, Cl, Br, I, -CN, N3 or -NO2;
each R23b is independently Y21;
each R23c îs independently -R2x, -N(R2x)(R2x). -SR2*, -S(O)R2*, -S(O)2R2*, S(O)(OR2*), -S(O)2(OR2*), -OC(=Y2I)R2x, -OC(=Y21)OR2*, -OC(=Y21)(N(R2*)(R2x)), SC(=Y2I)R2x, -SC(=Y2I)OR2x, -SC(=Y2,)(N(R2*)(R2*)), -N(R2x)C(=Y21)R2*, N(R2x)C(=Y21)OR2x, or -N(R2*)C(=Y21)(N(R2x)(R2*)) ;
each R23d is independently -C(=Y2,)R2x, -C(=Y21)OR2* or C(=Y2!)(N(R2x)(R2*));
each R2x is independently H, (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, aryl, heteroaryl; or two R2* taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or -NR21-; and wherein one or more of the non-terminal carbon atoms of each said (Ci-Cg)alkyl may be optionally replaced with -O-, -S- or -NR21-;
each R24 is independently (C[-C8)alkyl, (C2-C8)alkenyl, or (C2-C8)alkynyl;
each R25 is independently R24 wherein each R24 is substituted with 0 to 3 R23 groups;
each R25a is independently (Ci-C8)alkylene, (C2-C8)alkenylene, or (C210
C8)alkynylene any one of which said (Ci-Cs)alkylene, (C2-C8)alkenylene, or (C?C8)alkynylene is substituted with 0-3 R23 groups;
each W23 is independently W24 or W25;
each W24 is independently R25, -C(=Y2I)R2S, -C(=Y2I)W25, -SO2R25, or SO2W25;
each W25 is independently carbocycle or heterocycle wherein W25 is independently substituted with 0 to 3 R groups; and
1 each Y is independently O or S.
Table 20.1
O 0 0
2 3
Table 20.2
Ο
Table 20.4
Table 20.6
Table 20.7
Table 20.8
Table 20.9
Table 20.10
59
Table 20.11
62 63
68
Table 20,12
Table 20.13
Table 20.15
W23
W23
Table 20.16
W23
Table 20.17
102
104
106
Table 20.18
Table 20.20
Table 20.21
121
122
123
124
125
126
127
128
Table 20.22
Table 20.23
Table 20.24
R24
144
146
145
Table 20.25
148 149 150 151 152 153
< .W23 0 /R25 0 /R24 0 /R21 0 0 0 ^R23
154 155 156 157 158 159
Table 20.26
A. ,W23 N I ^R25 N /R24 N I ^R21 N I N <x .R23 N I
I H I H I H I H I H I H
160 161 162 163 164 165
/W23 N ^R25 N ''x xR24 N 'x ^R21 N | N ^R23 N |
I R23 R23 R23 R23 R23 R23
166 167 168 169 170 171
Table 20.27
179
178
Table 20.28
192 193 χΛ/
200
202
204
206
201
203
207
Table 20.32
208
209
210
223
228°
Ο
Ο
231
W23
233°
235°
Table 20.36
236
I
R23
O
237
I
R23
R24
R21
Table 30.1
257 'jZ
Embodiments of R* include esters, carbamates, carbonates, thioesters, amides, thioamides, and urea groups:
Any reference to the compounds of the invention described heereîn also includes a reference to a physiologically acceptable sait thereof. Examples of physiologically acceptable salts of the compounds of the invention include salts derived from an appropriate base, such as an alkalî métal or an alkaline earth (for example, Na+, Li+, K+’ Ca+- and Mg’ ^). ammonium and N R/ (wherein R is defined herein). Physiologically acceptable salts of a nitrogen atom or an amino group include (a) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acids, phosphoric acid, nîtric acid and the like; (b) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, isethionic acid, lactobîonic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, malonic acid, sulfosalicylic acid, glycolic acid, 2-hydroxy-3naphthoate, pamoate, salicylic acid, stearic acid, phthalic acid, mandelic acid, lactic acid, ethanesulfonic acid, lysine, arginine, glutamic acid, glycine, serine, threonîne, alanine, isoleucine, leucine and the like; and (c) salts formed from elemental anions for example, chlorine, bromine, and iodine. Physiologically acceptable salts of a compound of a hydroxy group include the anion of said compound in combination with a suîtable cation such as Na+ and NR4+.
For therapeutic use, salts of active ingrédients of the compounds of the invention will be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not physiologically acceptable may also find use, for example, in the préparation or purification of a physiologically acceptable compound. Ail salts, whether or not derived form a physiologically acceptable acid or base, are within the scope of the présent invention.
Finally, it is to be understood that the compositions herein comprise compounds of the invention in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
The compounds of the invention, ex empli fied by Formula I-IV may hâve chiral centers, e.g. chiral carbon or phosphorus atoms. The compounds of the invention thus include racemic mixtures of ali stereoisomers, including enantiomers, diastereomers, and atropisomers. In addition, the compounds of the invention include enriched or resolved optical isomers at any or ail asymmetric, chiral atoms. In other words, the chiral centers apparent from the depictions are provided as the chiral isomers or racemic mixtures. Both racemic and diastereomeric mixtures, as well as the individual optical isomers isolated or synthesized, substantially free of their enantiomeric or diastereomeric partners, are ail within the scope of the invention. The racemic mixtures are separated into their individual, substantially optically pure isomers through wellknown techniques such as, for example, the séparation of diastereomeric salts formed with optically active adjuncts, e.g., acids or bases followed by conversion back to the optically active substances. In most instances, the desired optical isomer is synthesized by means of stereospecîfic reactions, beginning with the appropriate stereoisomer of the desired starting material.
The term chiral refers to molécules which hâve the property of nonsuperimposability of the mirror image partner, while the term achîral refers to molécules which are superimposable on their mirror image partner.
The term stereoisomers refers to compounds which hâve identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molécules are not mirror images of one another. Diastereomers hâve different physical properties, e.g. melting points, boiling points, spectral properties, reactivities and biological properties. For example, the compounds of Formula I-IV
Y
II
may hâve a chiral phosphorus atom when R7 is W2 and Wl and W2 are different. When at least one of either W1 or W2 also has a chiral center, for example with W or W is a nitrogen-linked, chiral, naturally occurring α-amino acid ester, then the compound of Formula I-IV will exists as diastereomers because there are two centers of chirality in the molécule. Ail such diastereomers and their uses described herein are encompassed by the instant invention. Mixtures of diastereomers may be separate under high resolution analytical procedures such as electrophoresis, crystallization and/or chromatography. Diastereomeres may hâve different physical attributes such as, but not limited to, solubility, chemical stabilîties and crystallinity and may also hâve different biological properties such as, but not limited to, enzymatic stability, absorption and metabolic stability.
Enantiomers refer to two stereoisomers of a compound which are nonsuperimposable mirror images of one another.
Stereochemical définitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms ( 1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wîlen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. Many organic compounds exist in optîcally active forms, i.e., they hâve the ability to rotate the plane of planepolarized light. In describing an optîcally active compound, the préfixés D and L or R and S are used to dénoté the absolute configuration of the molécule about its chiral center(s). The préfixés d and 1, D and L, or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with S, (-), or 1 meaning that the compound is levorotatory while a compound prefixed with R, (+), or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A spécifie stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers îs often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic u/ mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms racemic mixture and racemate refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
Whenever a compound described herein is substituted with more than one of the same designated group, e.g., R or Rl, then it will be understood that the groups may be the same or different, i.e., each group is independently selected. Wavy fines, , indicate the site of covalent bond attachments to the adjoining substructures, groups, moietîes, or atoms.
The compounds of the invention can also exist as tautomeric isomers in certain cases. Although only one delocalized résonance structure may be depicted, ail such forms are contemplated within the scope of the invention. For example, ene-amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazole Systems and ail their possible tautomeric forms are within the scope of the invention.
Methods of Inhibition of a Paramyxoviridae polymerase
Another aspect of the invention relates to methods of inhibiting the activity of Paramyxoviridae polymerase comprising the step of treating a sample suspected of containing Paramyxoviridae with a composition of the invention.
Compositions of the invention may act as inhibitors of Paramyxoviridae polymerase, as intermediates for such inhibitors or hâve other utilities as described below. The inhibitors will bind to locations on the surface or in a cavity of Paramyxoviridae polymerase having a geometry unique to Paramyxoviridae polymerase . Compositions binding Paramyxoviridae polymerase may bind with varying degrees of reversibilîty. Those compounds binding substantially irreversibly are idéal candidates for use in this method of the invention. Once labeled, the substantially irreversibly binding compositions are useful as probes for the détection of Paramyxoviridae polymerase. Accordingly, the invention relates to methods of detecting Paramyxoviridae polymerase in a sample suspected of containing Paramyxoviridae polymerase comprising the steps of: treating a sample suspected of containing Paramyxoviridae polymerase with a composition comprising a compound f of the invention bound to a label; and observing the effect of the sample on the activity of the label. Suitable labels are well known in the diagnostics field and include stable free radicals, fluorophores, radioisotopes, enzymes, chemiluminescent groups and chromogens. The compounds herein are labeled in conventional fashion using functional groups such as hydroxyl, carboxyl, sulfhydryl or amino.
Within the context of the invention, samples suspected of containing Paramyxoviridae polymerase include natural or man-made materials such as living organisme; tissue or cell cultures; biological samples such as biological material samples (blood, sérum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like. Typically the sample will be suspected of containing an organism which produces Paramyxoviridae polymerase, frequently a pathogenîc organism such as a Paramyxoviridae virus. Samples can be contained in any medium including water and organic solvent\water mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures.
The treating step of the invention comprises adding the composition of the invention to the sample or it comprises adding a precursor of the composition to the sample. The addition step comprises any method of administration as described above.
If desired, the activity of Paramyxoviridae polymerase after application of the composition can be observed by any method including direct and indirect methods of detectîng Paramyxoviridae polymerase activity. Quantitative, qualitative, and semiquantitative methods of determining Paramyxoviridae polymerase activity are ail contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation of the physiological properties of a living organism are also applicable.
Organisms that contain Paramyxoviridae polymerase include the Paramyxoviridae virus. The compounds of this invention are useful in the treatment or prophylaxis of Paramyxoviridae infections in animais or in man.
However, in screening compounds capable of inhibiting human Paramyxoviridae viruses, it should be kept in mind that the results of enzyme assays may not correlate with cell culture assays. Thus, a cell based assay should be the primary screening tool.
Screens for Paramyxoviridae polymerase Inhibitors.
Compositions of the invention are screened for inhibitory activity against Paramyxoviridae polymerase by any of the conventional techniques for evaluating enzyme activity. Within the context of the invention, typîcally compositions are first screened for inhibition of Paramyxoviridae polymerase in vitro and compositions showing inhibitory activity are then screened for activity in vivo. Compositions having in vitro Ki (inhibitory constants) of less then about 5 X 10'6 M and preferably less than about 1 X 10? M are preferred for in vivo use.
Useful in vitro screens hâve been described in detail and will not be elaborated here. However, the examples describe suitable in vitro assays.
Pharmaceutical Formulations
The compounds of this invention are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glîdants, fillers, binders and the like. Aqueous formulations are prepared in stérile form, and when intended for delivery by other than oral administration generally will be isotonie. Ail formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986). Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextran, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10.
While it is possible for the active ingrédients to be administered alone it may be préférable to présent them as pharmaceutical formulations. The formulations, both for veterinary and for human use, of the invention comprise at least one active ingrédient, as above defined, together with one or more acceptable carriers therefor and optionally other therapeutic ingrédients, particularly those additional therapeutic ingrédients as discussed herein. The carrier(s) must be acceptable in the sense of beîng compatible with the other ingrédients of the formulation and physiologically innocuous to the récipient thereof.
The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington’s Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingrédient with the carrier which constitutes one or more accessory ingrédients. In general the formulations are prepared by uniformly and intîmately bringing into association the active ingrédient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Formulations of the présent invention suitable for oral administration may be presented as discrète units such as capsules, cachets or tablets each containing a predetermined amount of the active ingrédient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid émulsion or a water-in-oil liquid émulsion. The active ingrédient may also be administered as a bolus, electuary or paste.
A tablet is made by compression or molding, optionally with one or more accessory ingrédients. Compressed tablets may be prepared by compressing în a suitable machine the active ingrédient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingrédient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingrédient therefrom.
For infections of the eye or other external tissues e.g. mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1 % and 20% in incréments of 0.1 % w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. When formulated in an ointment, the active ingrédients may be employed with either a paraffinic or a water-miscible ointment base. Altematively, the active ingrédients may be formulated in a cream with an oil-in-water cream base.
If desired, the aqueous phase of the cream base may include, for example, at least 30% w/w of a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or pénétration of the active ingrédient through the skin or other affected areas. Examples of such dermal pénétration enhancers include dimethyl sulphoxide and related analogs.
The oîly phase of the émulsions of this invention may be constituted from known ingrédients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilie emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
Emulgents and émulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties. The cream should preferably be a non-greasy, nonstaining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as diisoadipate, isocetyl stéarate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stéarate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Altematively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other minerai oils are used.
Pharmaceutical formulations according to the présent invention comprise a combination according to the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingrédient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, émulsions, hard or soft capsules, syrups or élixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable préparation. Tablets containing the active ingrédient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnésium stéarate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
Formulations for oral use may be also presented as hard gelatin capsules where the active ingrédient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingrédient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally-occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty — acid (e.g., polyoxyethylene stéarate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingrédient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a minerai oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral préparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dîspersible powders and granules of the invention suitable for préparation of an aqueous suspension by the addition of water provide the active ingrédient in admixture with a dispersîng or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersîng or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be présent.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water émulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a minerai oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally-occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The émulsion may also contain sweetening and flavoring agents. Syrups and élixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the invention may be in the form of a stérile injectable préparation, such as a stérile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which hâve been mentioned above. The stérile injectable préparation may also be a stérile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in l ,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Rînger's solution and isotonie sodium chloride solution. In addition, stérile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleîc acid may lîkewise be used in the préparation of injectables.
The amount of active ingrédient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ug of the active ingrédient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations suitable for topical administration to the eye also include eye drops wherein the active ingrédient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingrédient. The active ingrédient is preferably présent in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10%, and particularly about 1.5% w/w.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingrédient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingrédient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingrédient in a suitable liquid carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for intrapulmonary or nasal administration hâve a particle size for example in the range of 0.1 to 500 microns, such as 0.5, 1, 30, 35 etc., which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oîly solutions of the active ingrédient. Formulations suitable for aérosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of Paramyxoviridae infections as described below.
In another aspect, the invention is a novel, efïicacious, safe, nonirritating and physiologically compatible inhalable composition comprising a compound of Formula
I-IV, or a pharmaceutically acceptable sait thereof, suitable for treating Paramyxoviridae infections and potentially associated bronchiolitis. Preferred pharmaceutically acceptable salts are inorganic acid salts including hydrochloride, hydrobromide, sulfate or phosphate salts as they may cause less pulmonary irritation. Preferably, the inhalable formulation is delivered to the endobronchial space in an aérosol comprising particies with a mass médian aerodynamîc diameter (MMAD) between about 1 and about 5 pm. Preferably, the compound of Formula I-IV is formulated for aérosol delivery using a nebulizer, pressurized metered dose inhaler (pMDI), or dry powder inhaler (DPI).
Non-limiting examples of nebulizers include atomizing, jet, ultrasonîc, pressurized, vibrating porous plate, or équivalent nebulizers including those nebulizers utilizing adaptive aérosol delivery technology (Denyer, J. Aérosol medicine Pulmonary Drug Delivery 2010, 23 Supp 1, Sl-S 10). A jet nebulizer utilizes air pressure to break a liquid solution into aérosol droplets. An ultrasonic nebulizer works by a piezoelectric crystal that shears a liquid into small aérosol droplets. A pressurized nebulization system forces solution under pressure through small pores to generate aérosol droplets. A vibrating porous plate device utilizes rapid vibration to shear a stream of liquid into appropriate droplet sîzes.
In a preferred embodiment, the formulation for nebulization is delivered to the endobronchial space in an aérosol comprising particles with a MMAD predominantly between about 1 pm and about 5 pm using a nebulizer able to aerosolize the formulation of the compound of Formula I-IV into particles of the required MMAD. To be optimally therapeutically effective and to avoid upper respiratory and systemic side effects, the majority of aerosolized particles should not have a MMAD greater than about 5 pm. If an aérosol contains a large number of particles with a MMAD larger than 5 pm, the particles are deposited in the upper airways decreasing the amount of drug delivered to the site of inflammation and bronchoconstriction in the lower respiratory tract. If the MMAD of the aérosol is smaller than about 1 pm, then the particles have a tendency to remain suspended in the inhaled air and are subsequently exhaled during expiration.
When formulated and delivered according to the method of the invention, the aérosol formulation for nebulization delivers a therapeutically efficacious dose of the compound of Formula I-IV to the site of Paramyxoviridae infection sufficient to treat the Paramyxoviridae infection. The amount of drug administered must be adjusted to reflect the efficiency of the delivery of a therapeutically efficacious dose of the compound of Formula I-IV. In a preferred embodiment, a combination of the aqueous aérosol formulation with the atomizing, jet, pressurized, vibrating porous plate, or ultrasonic nebulizer permits, depending on the nebulizer, about, at Ieast, 20, to about 90%, typically about 70% delivery of the administered dose of the compound of Formula I-IV into the airways. In a preferred embodiment, at Ieast about 30 to about 50% of the active compound is delivered. More preferably, about 70 to about 90% of the active compound is delivered.
In another embodiment of the instant invention, a compound of Formula I-IV or a pharmaceutically acceptable sait thereof, is delivered as a dry inhalable powder. The compounds of the invention are administered endobronchially as a dry powder formulation to efficacious deliver fine particles of compound into the endobronchial space using dry powder or metered dose inhalers. For delivery by DPI, the compound of Formula I-IV is processed into particles with, predominantly, MMAD between about 1 pm and about 5 pm by milling spray drying, critical fluid processing, or précipitation from solution. Media milling, jet milling and spray-drying devices and procedures capable of producing the particle sizes with a MMAD between about l pm and about 5 pm are well know in the art. In one embodiment, excipients are added to the compound of Formula I-IV before processing into particles of the required sizes. In another embodiment, excipients are blended with the particles of the required size to aid in dispersion of the drug particles, for example by using lactose as an excipient.
Particle size déterminations are made using devices well known in the art. For example a multi-stage Anderson cascade impactor or other suitable method such as those specifically cited within the US Pharmacopoeia Chapter 601 as characterizing devices for aérosols within metered-dose and dry powder inhalers.
In another preferred embodiment, a compound of Formula I-IV is delivered as a dry powder using a device such as a dry powder inhaler or other dry powder dispersion devices. Non-limiting examples of dry powder inhalers and devices include those disclosed in US5,458,135; US5,740,794; US5775320; US5,785,049; US3,906,950; US4,013,075; US4,069,819; US4,995,385; US5,522,385; US4,668,218; US4,667,668; US4,805,811 and US5,388,572. There are two major designs of dry powder inhalers. One design is a metering device in which a réservoir for the drug is place within the device and the patient adds a dose of the drug into the inhalation chamber. The second design îs a factory-metered device in which each individual dose has been manufactured in a separate container. Both Systems dépend on the formulation of the drug into small particles of MMAD from 1 pm and about 5 pm, and often involve coformulation with larger excipient particles such as, but not limited to, lactose. Drug powder is placed in the inhalation chamber (either by device metering or by breakage of a factory-metered dosage) and the inspiratory flow of the patient accelerates the powder out of the device and into the oral cavity. Non-laminar flow characteristics of the powder path cause the excipient-drug aggregates to décomposé, and the mass of the large excipient particles causes their impaction at the back of the throat, while the smaller drug particles are deposited deep in the lungs. In preferred embodiments, a compound of Formula I-IV, or a pharmaceutically acceptable sait thereof, is delivered as a dry powder using either type of dry powder inhaler as described herein, wherein the MMAD of the dry powder, exclusive of any excipients, is predominantly in the range of l pm to about 5 pm.
In another preferred embodiment, a compound of Formula I-IV is delivered as a dry powder using a metered dose inhaler. Non-limiting examples of metered dose inhalers and devices include those disclosed in US5,261,538; US5,544,647;
US5,622,163; US4,955,371; US3,565,070; US3,361306 and US6.116,234. In preferred embodiments, a compound of Formula I-IV, or a pharmaceutically acceptable sait thereof, is delivered as a dry powder using a metered dose inhaler wherein the MMAD of the dry powder, exclusive of any excipients, is predominantly in the range of about 1 -5 pm.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingrédient such carriers as are known in the art to be appropriate.
Formulations suitable for parentéral administration include aqueous and nonaqueous stérile injection solutions which may contain anti-oxidants, buffets, bacteriostats and solutés which render the formulation isotonie with the blood of the intended récipient; and aqueous and non-aqueous stérile suspensions which may include suspending agents and thickening agents.
The formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the stérile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from stérile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingrédient.
It should be understood that in addition to the ingrédients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
The invention further provides veterinary compositions comprising at least one active ingrédient as above defined together with a veterinary carrier therefor.
Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingrédient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
Compounds of the invention are used to provide controlled release pharmaceutical formulations containing as active ingrédient one or more compounds of the invention (controlled release formulations) in which the release of the active ingrédient are controlled and regulated to allow less frequency dosing or to improve the pharmacokinetic or toxicity profile of a given active ingrédient.
Effective dose of active ingrédient dépends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active viral infection, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day; typically, from about 0.01 to about 10 mg/kg body weight per day; more typically, from about .01 to about 5 mg/kg body weight per day; most typically, from about .05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses.
Routes of Administration
One or more compounds of the invention (herein referred to as the active ingrédients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, pulmonary, topical (including buccal and sublingual), vaginal and parentéral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and épidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the récipient. An advantage ofthe compounds of this invention is that they are orally bioavailable and can be dosed orally.
Combination Therapy
Compositions of the invention are also used in combination with other active ingrédients. For the treatment of Paramyxoviridae virus infections, preferably, the 5 other active therapeutic agent is active against Paramyxoviridae virus infections, particularly respiratory syncytial virus infections and/or parainfluenza virus infections. Non-limiting examples of these other active therapeutic agents are ribavirin, palivizumab, motavizumab, RSV-IGIV (RespiGam®), MEDI-557, A-60444, MDT-637, BMS-433771, and mixtures thereof.
Many of the infections of the Paramyxoviridae viruses are respiratory infections. Therefore, additional active therapeutics used to treat respiratory symptoms and sequelae of infection may be used in combination with the compounds of Formula
I-IV. The additional agents are preferrably administered orally or by direct inhalation. For example, other preferred additional therapeutic agents in combination with the compounds of Formula I-IV for the treatment of viral respiratory infections include, but are not limited to, bronchodîlators and corticosteroids.
Glucocorticoids, which were first introduced as an asthma therapy in 1950 (Carryer, Journal of Allergy, 21, 282-287, 1950), remain the most potent and consistently effective therapy for this disease, although their mechanîsm of action is not 20 yet fully understood (Morris, J. Allergy Clin. Immunol., 75 (1 Pt) 1-13, 1985).
Unfortunately, oral glucocorticoid thérapies are associated with profound undesirable side effects such as truncal obesity, hypertension, glaucoma, glucose intolérance, accélération of cataract formation, bone minerai loss, and psychological effects, ail of which limit their use as long-term therapeutic agents (Goodman and Gilman, 1 Oth 25 édition, 2001). A solution to systemic side effects is to delîver steroid drugs directly to the site of inflammation. Inhaled corticosteroids (ICS) hâve been developed to mitigate the severe adverse effects of oral steroids. Non-limiting examples of corticosteroids that may be used in combinations with the compounds of Formula I-IV are dexamethasone, dexamethasone sodium phosphate, fluorometholone, fluorometholone acetate, loteprednol, loteprednol etabonate, hydrocortisone, prednisolone.
fludrocortisones, triamcinolone, triamcînolone acetonide, betamethasone, beclomethasone diproprionate, méthylprednisolone, fluocinolone, fluocînolone acetonide, flunisolide, fluocortin-21-butylate, flumethasone, flumetasone pivalate, budesonide, halobetasol propionate, mometasone furoate, fluticasone propionate, ciclesonide; or a pharmaceutically acceptable salts thereof.
Other anti-inflamatory agents workîng through anti-inflamatory cascade mechanisms are also useful as additional therapeutic agents in combination with the compounds of Formula I-IV for the treatment of viral respiratory infections. Applying “anti-inflammatory signal transduction modulators” (referred to in this text as AISTM), like phosphodiesterase inhibitors (e.g. PDE-4, PDE-5, or PDE-7 spécifie), transcription factor inhibitors (e.g. blocking NFkB through IKK inhibition), or kinase inhibitors (e.g. blocking P38 MAP, JNK, PI3K, EGFR or Syk) is a logical approach to switching off inflammation as these small molécules target a limited number of common intracellular pathways - those signal transduction pathways that are critical points for the antiinflammatory therapeutic intervention (see review by P.J. Bames, 2006). These nonlimiting additional therapeutic agents include: 5-(2,4-Dîfluoro-phenoxy)-l-isobutyl-lHindazole-6-carboxylic acid (2-dimethylamino-ethyl)-amide (P38 Map kinase inhibitor ARRY-797); 3-Cyclopropylmethoxy-N-(3,5-dichioro-pyridin-4-yl)-4difluorormethoxy-benzamide (PDE-4 inhibitor Roflumilast); 4-[2-(3-cyclopentyloxy-4methoxyphenyl)-2-phenyI-ethyl]-pyridine (PDE-4 inhibitor CDP-840); N-(3,5dichloro-4-pyridinyI)-4-(difluoromethoxy)-8-[(methylsulfonyl)amino]-ldibenzofurancarboxamide (PDE-4 inhibitor Oglemilast); N-(3,5-Dichloro-pyridin-4yl)-2-[l-(4-fluorobenzyl)-5-hydroxy-lH-indol-3-yl]-2-oxo-acetamide (PDE-4 inhibitor AWD 12-281); 8-Methoxy-2-trifluoromethyl-quinoline-5-carboxylic acid (3,5dichloro-l-oxy-pyridin-4-yl)-amide (PDE-4 inhibitor Sch 351591); 4-[5-(4Fluorophenyl)-2-(4-methanesuIfinyl-phenyl)-l H-imidazol-4-yl]-pyridine (P38 inhibitor SB-203850); 4-[4-(4-Fluoro-phenyl)-l-(3-phenyl-propyl)-5-pyridin-4-yl-lH-imidazol-
2-yl]-but-3-yn-l-ol (P38 inhibitor RWJ-67657); 4-Cyano-4-(3-cyclopentyloxy-4methoxy-phenyl)-cyclohexanecarboxylic acid 2-diethylamino-ethyl ester (2-diethylethyl ester prodrug of Cilomilast, PDE-4 inhibitor); (3-Chloro-4-fluorophenyl)-[7methoxy-6-(3-morpholin-4-yl-propoxy)-quinazolin-4-yl]-amine (Gefitinib, EGFR -gf inhibitor); and 4-(4-Methyl-piperazin-l -ylmethyl)-N-[4-methyl-3-(4-pyridin-3-ylpyrimidin-2-ylamino)-phenyl]-benzamide (Imatinib, EGFR inhibitor).
Combinations comprising inhaled p2-adrenoreceptor agonist bronchodilators such as formoterol, albuterol or salmeterol with the compounds of Formula I-IV are also suitable, but non-Iîmiting, combinations useful for the treatment of respiratory viral infections.
Combinations of inhaled p2-adrenoreceptor agonist bronchodilators such as formoterol or salmeterol with ICS’s are also used to treat both the bronchoconstriction and the inflammation (Symbicort® and Advair®, respectively). The combinations comprising these ICS and p2-adrenoreceptor agonist combinations along with the compounds of Formula I-IV are also suitable, but non-limiting, combinations useful for the treatment of respiratory viral infections.
For the treatment or prophylaxis of pulmonary broncho-constriction, anticholinergics are of potential use and, therefore, useful as an additional therapeutic agents in combination with the compounds of Formula I-IV for the treatment of viral respiratory infections. These anticholinergics include, but are not limited to, antagonists of the muscarinic receptor (particularly of the M3 subtype) which hâve shown therapeutic efficacy in man for the control of cholinergic tone in COPD (Witek, 1999); l -{4-Hydroxy-1 -[3,3,3-tris-(4-fluoro-phenyl)-propionyl]-pyrrolidine-2carbonyl}-pyrrolîdine-2-carboxylic acid (l-rnethyl-piperidm-4-ylmethyl)-arnide; 3-[3(2-Dieth ylamino-acetoxy)-2-phenyl -propi on yloxy]-8-isopropyl-8-methyl- 8-azoniabicyclo[3.2.1]octane (Ipratropium-N,N-diethylglycinate); l-Cyclohexyl-3,4-dihydro1 H-isoquînoline-2-carboxylic acid l-aza-bicyclo[2.2.2]oct-3-yl ester (Solifenacin); 2Hydroxymethyl-4-methanesulfmyl-2-phenyl-butyric acid l-aza-bicyclo[2.2.2]oct-3-yl ester (Revatropate); 2-{l-[2-(2,3-Dihydro-benzofuran-5-yl)-ethyl]-pyrrolidin-3-yl}-2,2dîphenyl-acetamide (Darifenacin); 4-Azepan-l-yl-2,2-diphenyl-butyramide (Buzepide); 7-[3-(2-Diethylamîno-acetoxy)-2-phenyl-propionyloxy]-9-ethyl-9-methyl-3-oxa-9azonia-tricyclo[3.3.1.02,4]nonane (Oxitropiurn-N,N-diethylglycinate); 7-(2-(2DÎethylamino-acetoxy)-2,2-di-thiophen-2-yl-acetoxy]-9,9-dimcthyl-3-oxa-9-azoniatricyclo[3.3.1.02,4]nonane (Tiotropium-N.N-diethylglycinate); Dimethylamino-acetîc acid 2-(3-diisopropylamino-l-phenyl-propyl)-4-methyl-phenyl ester (Tolterodine-Ν,Ν dimethylglycinate); 3-[4,4-Bis-(4-fIuoro-phenyl)-2-oxo-imidazolidin-l-yl]-l-methyl-1(2-oxo-2-pyridin-2-yl-ethyl)-pyrrolidinium; l-[l-(3-Fluoro-benzyl)-piperidin-4-yl]-4,4bis-(4-fluoro-phenyl)-imidazolidin-2-one; l-Cyclooctyl-3-(3-methoxy-l-azabicyclo[2.2.2]oct-3-yl)-l -phenyl-prop-2-yn-l-ol; 3-[2-(2-Diethylamino-acetoxy)-2,2di-thiophen-2-yl-acetoxy]-l-(3-phenoxy-propyl)-l-azonia-bicyclo[2.2.2]octane ( Aclidinium-N,N-diethylgiycinate); or (2-Diethylamino-acetoxy)-di-thiophen-2-ylacetic acid l-methyl-l-(2-phenoxy-ethyl)-piperidin-4-yl ester.
The compounds of Formula I-IV may also be combined with mucolytîc agents to treat both the infection and symptoms of respiratory infections. A non-limiting example of a mucolytîc agent is ambroxol. Similarly, the compounds of Formula I-IV may be combined with expectorants to treat both the infection and symptoms of respiratory infections. A non-limiting example of an expectorant is guaifenesin.
Nebulized hypertonie saline is used to improve immédiate and lon-term clearance of small airways in patients with lung diseases (Kuzik, J. Pediatrics 2007, 266). The compounds of Formula I-IV may also be combined with nebulized hypertonie saline particularly when the Paramyxoviridae virus infection is complicated with bronchiolitis. The combination of the compounds of Formula I-IV with hypertonie saline may also comprise any of the additional agents discussed above. In a preferred aspect, nebulized about 3% hypertonie saline is used.
It is also possible to combine any compound of the invention with one or more additional active therapeutic agents in a unitary dosage form for simultaneous or sequentîal administration to a patient. The combination therapy may be administered as a simultaneous or sequentîal regimen. When administered sequentially, the combination may be administered in two or more administrations.
Co-administration of a compound of the invention with one or more other active therapeutic agents generally refers to simultaneous or sequentîal administration of a compound of the invention and one or more other active therapeutic agents, such that therapeutically effective amounts of the compound of the invention and one or more other active therapeutic agents are both présent in the body of the patient.
Co-administration includes administration of unit dosages of the compounds of the invention before or after administration of unit dosages of one or more other active v·/ therapeutic agents, for example, administration of the compounds of the invention within seconds, minutes, or hours of the administration of one or more other active therapeutic agents. For example, a unit dose of a compound of the invention can be administered first, followed within seconds or minutes by administration of a unit dose of one or more other active therapeutic agents. Alternative! y, a unit dose of one or more other therapeutic agents can be administered first, followed by administration of a unit dose of a compound of the invention within seconds or minutes. In some cases, it may be désirable to administer a unit dose of a compound of the invention first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more other active therapeutic agents. In other cases, it may be désirable to administer a unit dose of one or more other active therapeutic agents first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound of the invention.
The combination therapy may provide “synergy” and “synergistic”, i.e. the effect achieved when the active ingrédients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingrédients are: (1) co-formulated and administered or delîvered simultaneously in a combined formulation; (2) delîvered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delîvered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delîvered sequentially, e.g. in separate tablets, piils or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingrédient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingrédients are administered together. A synergistic anti-viral effect dénotés an antiviral effect which is greater than the predicted purely additîve effects of the individual compounds of the combination.
In still yet another embodiment, the présent application provides for methods of inhibiting Pciramyxoviridae polymerase in a cell, comprising: contacting a cell infected with HCV with an effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof, whereby Paramyxoviridae polymerase is inhibited.
In still yet another embodiment, the présent application provides for methods of inhibiting Paramyxoviridae polymerase in a cell, comprising: contacting a cell infected with HCV with an effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof, and at least one additionaî active therapeutic agent, whereby Paramyxoviridae polymerase îs inhibited.
In stîll yet another embodiment, the présent application provides for methods of inhibiting Paramyxoviridae polymerase in a cell, comprising: contacting a cell infected with Paramyxoviridae virus with an effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof, and at least one additionaî active therapeutic agent selected
In still yet another embodiment, the présent application provides for methods of treating Paramyxoviridae virus infection in a patient, comprising: administering to the patient a therapeutically effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof.
In still yet another embodiment, the présent application provides for methods of treating Paramyxoviridae virus infection in a patient, comprising: administering to the patient a therapeutically effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof, and at least one additionaî active therapeutic agent, whereby Paramyxoviridae polymerase îs inhibited.
In still yet another embodiment, the présent application provides for methods of treating Paramyxoviridae virus infection in a patient, comprising: administering to the patient a therapeutically effective amount of a compound of Formula I-IV, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof, and at least one additionaî active therapeutic agent.
Métabolites of the Compounds of the Invention
Also falling within the scope of this invention are the in vivo metabolic products of the compounds described herein, to the extent such products are novel and unobvious over the prior art. Such products may resuit for example fforn the oxidation, yf' réduction, hydrolysis, amidation, estérification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes novel and unobvious compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabelled (e.g. or ^H) compound ofthe invention, administering it parenterally in a détectable dose (e.g. greater than about 0.5 mg/kg) to an animal such as rat, mouse, guînea pîg, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the métabolite). The métabolite structures are determined in conventional fashion, e.g. by MS or NMR analysis. In general, analysis of métabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no HCV polymerase inhibitory activity of their own.
Recipes and methods for determining stability of compounds in surrogate gastrointestinal sécrétions are known. Compounds are defined herein as stable in the gastrointestinal tract where less than about 50 mole percent of the protected groups are deprotected în surrogate intestinal or gastric juice upon incubation for l hour at 37°C. Simply because the compounds are stable to the gastrointestinal tract does not mean that they cannot be hydrolyzed in vivo. The prodrugs of the invention typically will be stable in the digestive system but may be substantially hydrolyzed to the parental drug in the digestive lumen, liver or other metabolic organ, or within cells in general.
Examples
Certain abbreviations and acronyms are used in describing the experimental details. Although most of these would be understood by one skilled in the art, Table 1 contains a list of many of these abbreviations and acronyms.
Table l. List of abbreviations and acronyms.
Abbreviation Meanîng
Ac2O acetic anhydride
AIBN 2,2’-azobis(2-methylpropionitrile)
Bn benzyl
BnBr benzylbromide
BSA bis(trimethylsilyl)acetamide
BzCl benzoyl chloride
CDI carbonyl diimidazole
DABCO l,4-diazabicyclo[2.2.2]octane
DBN I,5-diazabicyclo[4.3.0]non-5-ene
DDQ 2,3-dichloro-5,6-dîcyano-l,4-benzoquinone
DBU I,5-diazabicyclo[5.4.0]undec-5-ene
DCA dichloroacetamide
DCC dicyclohexylcarbodiimide
DCM dichloromethane
DMAP 4-dimethylaminopyridine
DME 1,2-dimethoxyethane
DMTCl dimethoxytrityl chloride
DMSO dimethylsulfoxide
DMTr 4, 4’-dimethoxytrityl
DMF dîmethylformamide
EtOAc ethyl acetate
ESI electrospray ionizatîon
HMDS hexamethyldisilazane
HPLC High pressure liquid chromatography
LDA lithium diisopropylamide
LRMS low resolution mass spectrum
MCPBA meta-chloroperbenzoic acid
MeCN acetonitrile
MeOH méthanol
MMTC mono methoxytrityl chloride
m/z or m/e mass to charge ratio
MH+ mass plus 1
MH' mass minus 1
MsOH methanesulfonic acid
MS or ms mass spectrum
NBS N-bromosuccinimide
Ph phenyl
rt or r.t. room température
TBAF tetrabutylammonium fluoride
TMSCl chlorotrimethylsilane
TMSBr bromotrimethylsilane
TMSI iodotrimethylsilane
TMSOTf (trimethylsilyl)trifluoromethylsulfonate
TEA triethylamine
TBA tributyl amine
TBAP tributylammonium pyrophosphate
TBSCl t-butyldimethylsilyl chloride
TEAB triethylammonium bicarbonate
TFA trifluoroacetic acid
TLC or tic thin layer chromatography
Tr triphenylmethyl
Toi 4-methylbenzoyl
Turbo Grignard 1:1 mixture of isopropylmagnesium chloride and lithium chloride
δ parts per million down field from tetramethylsilane
Préparation of Compounds (2S)-ethvl 2-(chIorofphenoxv)phosphorvlamino)propanoate (Chloridate A)
100
A
Ethyl alanine ester hydrochloride sait (1.69 g, 11 mmol) was dissolved in anhydrous CH2CI2 (10 mL) and the mixture stirred with cooling to 0 °C under N2(g). Phenyl dichlorophosphate (1.49 mL, 10 mmol) was added followed by dropwise addition of EbN over 10 min. The reaction mixture was then slowly warmed to RT and stirred for 12 h. Anhydrous Et2O (50 mL) was added and the mixture stirred for 30 min. The solid that formed was removed by filtration, and the filtrate concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-50% EtOAc in hexanes to provide intermediate A (1.13 g, 39%).
'H NMR (300 MHz, CDC13) δ 7.39-7.27 (m, 5H), 4.27 (m, 3H), 1.52 (m, 3H), 1.32 (m, 3 H).
31P NMR (121.4 MHz, CDC13) δ 8.2, 7.8.
(2S)-2-cthvlbutyl 2-(chÎoro(phenoxv)phosphorvÎamino)propanoate (Chloridate B)
The 2-ethylbutyl alanine chlorophosphoramidate ester B was prepared using the same procedure as chloridate A except substituting 2-ethylbutyl alanine ester for ethyl alanine ester. The material is used crude in the next reaction. Treatment with methanol or éthanol forms the displaced product with the requisite LCMS signal. vZ
101 (2S)-isopropyl 2-(chloro(phenoxv)phosphorylamino)propanoate (Chloridate C)
O-P-CI
Cl
The isopropyl alanine chlorophosphoramidate ester C was prepared using the same procedure as chloridate A except substituting isopropyl alanine ester for the ethyl alanine ester. The material is used crude in the next reaction. Treatment with methanol or éthanol forms the displaced product with the requisite LCMS signal. 'W'
102 (2R, 3R, 4S, 5RÎ-2-(4-aniinopvrroloH,2-f|[1.2,4]trÎazin-7-vl)-3.4-dihvdroxv-5(hvdroxvmethvl)tetrahydrofuran-2-carbonitrile (Compound l)
BnÔ ÔBn
DMSO
BnÔ ÔBn
AC2O
The commercially available lactol (10 g, 23.8 mmol) was dissolved in anhydrous DMSO (30 mL) under N2(g). Ac2O (20 mL) was added and the résultant reaction mixture stirred at RT for 48 h. The reaction mixture was poured onto ice H2O (500 mL) and the mixture stirred for 20 min. The mixture was extracted with EtOAc (3 x 200 mL) and the combined organic extracts were then washed with H2O (3 x 200 mL). The organic extract was dried over anhydrous MgSO4. filtered and concentrated under reduced pressure. The residue was dissolved in CH2C12 and subjected to silica gel chromatography elutîng with 25% EtOAc in hexanes to provide the lactone (9.55 g, 96%).
lH NMR (400 MHz, DMSO) δΠ □ 7.30-7.34 (m, 13H), 7.19-7.21 (m, 2H), 4.55-4.72 (m, 6H), 4.47 (s, 2H), 4.28 (d.,/ - 3.9 Ηζ,ΙΗ), 3.66 (m, 2H).
LCMS m/z 436.1 [M+H2O], 435.2 [M+OH]- Tr = 2.82 min HPLC Tr = 4.59 [2-98% ACN in H2) over 5 min @ 2ml / min flow.
BnÔ ÔBn
The bromopyrazole (prepared according to WO2009/132135) (0.5 g, 2.4 mmol) was suspended in anhydrous THF (10 mL) under N2(g). The suspension was stirred and
103
TMSCl (0.67 mL, 5.28 mmol) was added. The mixture was stirred for 20 min. at RT and then cooled to -78 °C after which time a solution of n-BuLi (6 mL, l .6 N in hexanes, 9.6 mmol) was added slowly. The reaction mixture was stirred for 10 min. at 78 °C and then the lactone (I g, 2.4 mmol) was added via syringe. When the reaction was complété as measured by LCMS, AcOH was added to quench the reaction. The mixture was concentrated under reduced pressure and the residue dissolved in a mixture of CH2CI2 and H2O (100 mL, 1:1). The organic layer was separated and washed with H2O (50 mL). The organic layer was then dried over anhydrous MgSO,·., filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-50% EtOAc in hexanes to provide the product as a 1 : i mixture of anomers (345 mg, 26% yield).
LCMS m/z 553 [M+HJ.
The hydroxy nucleoside (1.1 g, 2.0 mmol) was dissolved in anhydrous CH2CI2 (40 mL) and the solution cooled with stirring to 0 °C under N2(g). TMSCN (0.931 mL, 7 mmol) was added and the mixture stirred for a further 10 min. TMSOTf (1.63 mL, 9.0 mmol) was slowly added to the reaction and the mixture stirred for 1 h. The reaction mixture was then diluted with CH2C12 (120 mL) and aqueous NaHCO3 (120 mL) was added to quench the reaction. The reaction mixture was stirred for a further 10 min and the organic layer separated. The aqueous layer was extracted with CH2C12 (150 mL) and the combined organic extracts dried over anhydrous MgSCU, filtered and concentrated under reduced pressure. The residue was dissolved in a minimal amount of CH2C12 and subjected to silica gel chromatography eluting with a gradient of 0-75% EtOAc and hexanes to provide the tribenzyl cyano nucleoside as a mixture of anomers. (0.9 g, 80%). NkA
104 ‘h NMR (300 MHz, CD3CN) δ 7.94 (s, 0.5H), 7.88 (s, 0.5H), 7.29-7.43 (m, 13H),
7.11-7.19 (m, IH), 6.82-6.88 (m,lH), 6.70-6.76 (m, IH), 6.41 (bs, 2H), 5.10 (d, J = 3.9
Hz, 0.5H), 4.96 (d, J = 5.1 Hz, 0.5H), 4.31-4.85 (m, 7H), 4.09-4.18 (m, 2H), 3.61-3.90 (m, 2H).
LCMS m/z 562 [M+H].
beta 1
The tribenzyl cyano nucleoside (70 mg, 0.124 mmol) was dissolved in anhydrous CILCL (2 mL) and cooled to -78 °C under N2(g). A solution of BC13 (IN in CH2C12, 0.506 mL, 0.506 mmol) was added and the reaction mixture stirred for 1 h. at 78 °C. When the reaction was complété by LC/MS, MeOH was added to quench the reaction. The reaction mixture was allowed to warm to room RT and the solvent removed under reduced pressure. The residue was subjected to Cl8 reverse phase HPLC, eluting for 5 min with H2O (0.1 % TFA), followed by a gradient of 0-70% MeCN în H2O (0.1 % TFA) over 35 min, to elute the a-anomer (20 mg, 37%), and βanomer 1 (20 mg, 37%).
(a-anomer) ‘H NMR (300 MHz, D2O) δ 7.96 (s, IH), 7.20 (d, J = 4.8 Hz, IH), 6.91 (d, J =4.8 Hz, IH), 4.97 (d, J =4.4 Hz, IH), 4.56-4.62 (m, IH), 4.08-4.14 (m, IH), 3.90 (dd, J = 12.9,
2.4 Hz, IH), 3.70 (dd, J = 13.2,4.5 Hz, IH).
(β-anomer) lH NMR (400 MHz, DMSO) δ 7.91 (s, IH), 7.80-8.00 (br s, 2H), 6.85-6.89 (m, 2H), 6.07 (d, J =6.0 Hz, IH), 5.17 (br s, IH), 4.90 (br s, IH), 4.63 (t, J =3.9 Hz, 1 H), 4.024.06 (m, IH), 3.94 (br s, IH), 3.48-3.64 (m, 2H).
105
LCMS m/z 292.2 [M+H], 290.0 [M-H], Tr= 0.35 min.
13CNMR (400 MHZ, DMSO), 156.0, 148.3, 124.3, 117.8, 117.0, 111.2, 101.3, 85.8,
79.0, 74.7, 70.5,61.4
HPLC Tr = 1.32 min (2R,3R.4R«5R)-2-(4-aminopyrToloï 1,2-f| 11,2,4| (riaz.in-7-yl)-3-fluoro-4-h ydroxv-5(hvdroxvmcthvl)tetrahvdrofuran-2-carbonitrile (Cornpound 2)
BnO' F
TFA, H2O (9:1)
r.t., 18 h
2-Deoxy-2-fluoro-4,5-O,O-dibenzyl-D-arabinose. l’-Methoxy-2-deoxy-2-fluoro-4,5O,O-dibenzyl-D-arabinose (1.0 g, 2.88 mmol) in TFA (13.5 mL) was treated with H2O (1.5 mL) and the résultant mixture stirred for 5 h. The mixture was then diluted with EtOAc (100 mL) and treated with saturated NaHCO? (50 mL). The organic layer was separated and washed with NaCl (50 mL), dried over anhydrous MgSO.i, filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography (80 g SiO2 Combiflash HP Gold Column) eluting with 0-100% EtOAc in hexanes to afford 2-deoxy-2-fluoro-4,5-O,O-dibenzyl-D-arabînose (695 mg, 72%) as a white solid: Rf- 0.52 (25% EtOAc in hexanes);
lH NMR (300 MHz, CDCl3) δ 7.30 (m, 10H), 5.35 (m, 1H), 4.68 4.29 (m, 7H), 3.70 (d, J = 10.5 Hz, 1H), 3.50 (d, J= 10.5 Hz, 2H).
19F NMR (282.2 MHz, CDC13) δ -207 (m), -211 (m).
LCMS m/z 350 [M+H2O].
PDC, 4 A MS, DCM
r.t., 18 h
BnÛ F χλ/'
106 (3Λ, 4R, 5/I)-4-(benzyloxy)-5-(benzyloxymethyl)-3-fluorodihydrofuran-2(3H)-one.
2-Deoxy-2-fIuoro-4, 5-O,O-dibenzyl-D-arabinose (4.3 g, 12.8 mmol) was dissolved in CH2C12 (85 mL) was treated with 4 Â MS (10 g) and pyridinium dichromate (14.4 g, 38.3 mmol). The résultant mixture was stirred for 24 h and then filtered through a pad of Celite. The éluant was concentrated under reduced pressure and the residue subjected to silica gel chromatography (120 g SiO? HP Gold Combiflash Column) eluting with 0-100% EtOAc in hexanes to afford (3R, 4R, 52f)-4-(benzyloxy)-5(benzyloxymethyI)-3-fIuorodihydrofuran-2(3H)-one as a clear oil (3.5 g, 83%): R/ = 0.25 (25% EtOAc in hexanes).
'H NMR (300 MHz, CDC13) δ 7.37 (m, 10H), 5.45 (dd, J= 49, 5.7, Hz, IH), 4.85 (d, J = 11.7 Hz, IH), 4.52 (m, 4 H), 4.29 (d, 7=5.4 Hz, 1 H), 2.08 (dd, 7= 15.3, 10.2 Hz, 2H).
l9F NMR (282.2 MHz, CDC13) δ-216.
LCMS m/z 348 [M+H2O].
HPLC (6-98% MeCN-H2O gradient, 0.05% TFA modifier) tR = 5.29 min. Phenomenex Synergî 4 m Hydro-RP 80 A, 50 x 4.60 mm, 4 micron; 2 mL/min flow rate
(3Λ, 4Æ, 5A)-2-(4-aminopyrrolo[l,2-f]|l,2,4|triazin-7-yl)-4-(benzyloxy)-5(benzyloxymethyl)-3-fluorotetrahydrofuran-2-ol. 7-Bromopyrrolo[l,2-f][l,2,4]triazin-4-amine (68 mg, 0.319 mmol) in THF (1.4 mL) was treated with TMSC1 (89 pL,0 .703 mmol) and the mixture stirred for 2 h. The mixture was then cooled to -78 t
°C and treated wîth«BuLi (1.0 M in hexanes, 1.09 mL, 1.09 mmol). The solution was stirred for 30 min and then treated with (3Λ, 4Λ, 5/?)-4-(benzyloxy)-5107 (benzyloxymethyl)-3-fluorodihydrofuran-2(3H)-one (106 mg, 0.319 mmol) dropwise in THF (1.4 mL). The résultant mixture was stirred for 30 min and then AcOH (83 pL,
1.44 mmol) in THF (1.0 mL) was added to quench the reaction. The mixture was warmed to RT and then concentrated under reduced pressure. The residue was diluted with EtOAc (100 mL) and washed with saturated NaCl solution (50 mL). The organic layer was dried over anhydrous MgSO4) filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography (40 g SiO? HP Gold Combiflash Column) eluting with 0-100% EtOAc in hexanes followed by a 0-100% gradient of (20% MeOH in EtOAc) in EtOAc to afford (3R, 4R, 5R)-2-(4aminopyrrolo[ 1,2-fJ [ 1,2,4]triazin-7-yl)-4-(benzyloxy)-5-(benzyloxymethyl)-3fluorotetrahydrofuran-2-ol as a white solid (68 mg, 44%, 60/40 mixture of α/β isomers). Ry= 0.32 (EtOAc).
‘H NMR (300 MHz, CDC13) δ 8.05 (s, IH), 7.86 (s, IH), 7.81 (s, IH), 7.64 (s, IH),
7.26 (m, 10H), 6.95 (m, IH), 6.71 (m, 1H),6.O8 (m, IH), 5.34 (m, 1 H), 4.65 (m, 6H), 4.71 (m, 2H).
,9F NMR (282.2 MHz, CDC13) S -211 (m).
LCMS m/z 465 [M+H].
HPLC (6-98% MeCN-H2Û gradient, 0.05% TFA modifier) tR = 4.37 min. (a-isomer), 4.54 min. (β-isomer).
TMSCN, ln(OTf)3, MeCN °C,18 h
(3Λ, 4R, 5/?)-2-(4-aminopyrrolo|l,2-f]|L2,4|triazin-7-yl)-4-(benzyloxy)-5(benzyloxymethyl)-3-fluorotetrahydrofuran-2-carbonitrile; (3 R, 4R, 5//)-2-(4aminopyrrolo[ 1,2-f] [ 1,2,4]triazin-7-yl)-4-(benzyloxy)-5-(benzyloxymethyl)-3fluorotetrahydrofuran-2-ol (195 mg, 0.42 mmol) was dissolved in MeCN (1.4 mL) was treated with TMSCN (336 pL, 2.52 mmol) and In(OTf)3 (708 mg, 1.26 mmol). The
108 solution was stirred at 70 °C for 18 h and then cooled to 0 °C. The mixture was treated with saturated N ail CO 3 solution (20 drops) then warmed to RT and diluted with EtOAc ( 100 mL) and H2O (50 mL). The organic layer was separated and washed with saturated NaCl solution (50 mL), dried over MgSCh, filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography (40 g SiO2 HP Gold Combiflash Column) eluting with 0-100% EtOAc in hexanes to afford (3R, 4R, 57?)-2-(4-aminopyrrolo[l,2-f][l,2,4]triazin-7-yl)-4-(benzyloxy)-5(benzyloxymethyl)-3-fluorotetrahydrofuran-2-carbonitrile as a white solid (110 mg, 55%, 60/40 mixture of α/β isomers). Data for both isomers: Rf= 0.53 (EtOAc).
lH NMR (300 MHz, CDC13) δ 8.01 (s, IH), 7.94 (s, lH),7.30(m, 10H), 7.00 (d, 7 =
4.5 Hz, IH), 6.93 (d,.7=4.8 Hz, IH), 6.87 (d,7 = 5.4 Hz, IH), 6.70 (d,.7=4.8 Hz, 1 H), 5.85 (dd, 7=52,3.3 Hz, 1 H), 5.55 (dd, 7= 53,4.5 Hz, IH), 4.71 (m, 7H), 3.87 (m, 2H), 3.72 (m, 2H).
,9F NMR (282.2 MHz, CDCf) δ -196 (m), -203 (m).
LCMS m/z 474 [M+H].
HPLC (6-98% MeCN-H2O gradient, 0.05% TFA modifier) tg = 4.98 min.
(27?, 3/?, 47?, 5/?)-2-(4-aminopyrrolo| l,2-f|[l,2,4]triazin-7-yl)-3-fluoro-4-hydroxy-5(hydroxymethyl)tetrahydrofuran-2-carbonitrile (2) (3R, 47?, 57?)-2-(4aminopyrrolo[ 1,2-f][ 1,2,4]triazin-7-yl)-4-(benzyloxy)-5-(benzyloxymethyl)-3fluorotetrahydrofüran-2-carbonitrile (110 mg, 0.23 mmol) was dissolved in CH2C12 (1.5 mL) and cooled to 0 °C. The reaction mixture was treated with BCI3 (1.0 M in CH2C12,
766 pL, 0.77 mmol) and stirred for 2 h. The mixture was then cooled to -78 °C and treated with Et3N (340 pL, 2.44 mmol) followed by MeOH (2 mL) before allowîng toM/-'
109 warm to RT. The reaction was concentrated under reduced pressure and then coevaporated with MeOH (3x5 mL). The residue was then suspended in H?O (5 mL) and treated with NaHCO3 (1 g). The solution was stirred for 10 min and then concentrated under reduced pressure. The residue was filtered and washed with MeOH (3x10 mL) on a fritted glass funnel (coarse) and the éluant concentrated under reduced pressure. The residue was subjected to reverse phase HPLC (6-98% MeCN in H2O gradient with 0.05% TFA modifier) to afford (2R, 3Λ, 4R, 57f)-2-(4-aminopyrrolo[ 1,2-
f][l,2,4]triazin-7-yl)-3-fluoro-4-hydroxy-5-(hydroxymethyl)tetrahydrofuran-2carbonitrile 2 as a white solid (16.8 mg, 25%) and the a-isomer.
Data for the β-isomer: R/= 0.13 (10% MeOH in EtOAc).
‘H NMR (300 MHz, CD3OD) δ 8.09 (s, 1 H), 7.28 (d, J = 5.1 Hz, 1 H), 7.17 (d, J = 5.1 Hz, IH), 5.42 (dd, J= 53, 3.3 Hz, 1H),4.2O (m, 2H), 3.99 (d, J= 3.6 Hz, IH), 3.77 (d, .7=3.6 Hz, IH).
I9F NMR (282.2 MHz, CDC13) δ -197 (m).
LCMS m/z 294 [M+HJ.
HPLC (2-98% MeCN-H2O gradient, 0.05% TFA modifier) tu = 1.49 min.
(2R, 3 R, 4R, 5S)-5-(4-aminopyrrolo[l,2-fH 1,2,41 triazin-7-vl)-4-fluoro-2(hvdroxvmethvD-5-methvltetrahvdrofuran-3-ol (Compound 3)
The starting nucleoside (prepared as described in the sysnthesis of compound 2) (0.355 g, 0.765 mmol) was dissolved in anhydrous THF (35 mL) and cooled to 0°C with stirring under N2(g). A solution of methyl magnésium chloride (2 mL, 6 mmol) (3 N in THF) was added and the résultant mixture stirred overnight. Acetic acid (7 mmol) was added to quench the reaction and then the solvents were removed by rotory
110 under reduced pressure. The residue was re-dissolved in CH2CI2 and the solution subjected to a plug of silica gel to isolate the product (0.355 g) as a crude mixture. LC/MS (m/z : 480, M+1). The crude material was dissolved in anhydrous CH2CI2 (20 mL) and placed under N2(g). The solution was stirred and treated with methanesulfonic acid (0.2 mL, 2.74 mmol). The reaction mixture was stirred for 12 h at RT and then quenched by the addition of Et3N (3.5 mmol). The mixture was concentrated under reduced pressure and the residue subjected to silica gel chromatography to provide the methyl substituted nucleoside (0.174 g, 0.377 mmol, 44% yield) as a 4:1 mixture of beta- and alpha-anomers respectively.
’H NMR (300 MHz, CD3CN) major anomer δ 7.87 (s, IH), 7.27-7.40 (m, 10 H), 6.77 (d, J = 4.5 HZ, IH), 6.70 (d, J =4.5 Hz, IH), 6.23 (brs, 2H), 5.53 (dd, J = 55, 3.3 Hz, IH), 4.42-4.75 (m, 4H), 4.19-4.26 (m, IH), 3.65-4.00 (m, 3H), 1.74 (d, J = 3.9 Hz, 3H). l9F NMR (282.2 MHz, CD3CN) major anomer δ -207 (m, 1F)
LCMS m/z 463 [M+H].
beta alpha
The benzylated nucleoside material (0.134 g, 0.290 mmol), Degussa catalyst (0.268 g) and AcOH (30 mL) were mixed together. The reaction atmosphère was charged with H2 (g) and the reaction stirred for 2 h. The catalyst was removed by filtration and the mixture concentrated under reduced pressure. The residue was dissolved in a minimal amount of H2O and subjected to reverse phase HPLC (C18 hydro RP column) to isolate the β-anomer 3 (0.086 g, 0.217 mmol, 57% yield).
111 ‘H NMR (300 MHz, D2O) ÔD7.87 (s, 1H), 7.22 (d, J = 4.8 Hz, 1H), 6.87 (d, J = 4.8
Hz, 1H), 5.35 (dd, J = 54, 3.6 Hz, IH), 3.97-4.10 (m, 2H), 3.81 (dd, J = 12.6, 2.1 Hz,
H), 3.64 (dd, J = 12.6, 4.8 Hz, 1H), 1.65 (d, J =4.2 Hz, 3H).
,9F NMR (282.2 MHz, CD3CN) ÔD -207 (m, 1F).
A small amount of alpha anomer was characterized as follows.
lH NMR (300 MHz, D2O) δΠ7.86 (s, 1H), 7.26 (d, J = 4.8 Hz, 1H), 6.85 (d, J = 4.8 Hz, 1H), 5.31 (dd, J = 54, 3.9 Hz, 1H), 4.39 (ddd, J = 26.1, 9.9, 3.6 Hz, 2H), 4.00 4.05 (m, 1 H), 3.90 (dd, J = 12.3, 2.1 Hz, 1H), 3.66 (dd, J = 12.6, 4.8, 1H), 1.56 (s, 3H). 19F NMR (282.2 MHz, CD3CN) δΠ -198 (dd, J = 54, 26 Hz, 1 F).
(2R)-isopropyl 2-((((2R.3R.4R,5S)-5-(4-aminopvrrolo|l,2-f||l,2,4|triazin-7-yl)-4fluoro-3-hvdroxv-5-methvltetrahydrofuran-2-vl)methoxv)(phenoxv)phosphorylamîno)propanoate (Compound 4)
beta
The nucleoside 3 (0.011 g, 0.04 mmol) was dissolved in trimethylphosphate (2 mL) and cooled to 0°C. The mixture was stirred under an atmosphère of N2(g) andlMethylimidazole(0.320 mL, 5 mmol) followed by the alaninylmonoisopropyl, monophenol phosphorchloridate C (0.240 mL, 4.4 mmol) was added. The reaction mixture was stirred for 2 h. at 0°C and then allowed to warm slowly to RT. while monitoring by LC/MS. When complété by LCMS, the reaction mixture was treated with H2O (5 mL) and then concentrated under reduced pressure. The residue was dissolved in CH2C12 and subjected to silica gel chromatography eluting with 0-100% EtOAc in hexanes. The product fractions were collected and concentrated. The residue
112 was subjected to prep HPLC to yield the alanine isopropyl monoamidate prodrug 4 as a mixture of isomers (4.7 mg, 0.003 mmol, 6%).
lH NMR (300 MHz, CD3CN) δ 7.87 (s, l H), 7.17-7.44 (m, 5 H), 6.71-6.83 (m, 2H),
6.14 (br, s, 2H), 5.38 (dd, J = 56, 3.3 Hz, IH), 4.92-5.01 (m, IH), 3.86-4.46 (m, 6H),
3.58 (m, IH), 1.73 (m, 3H), 1.18-1.34 (m, 9H)
LCMS m/z 552 [M+H], (2R)-ethyl 2-((((2 R,3R.4R.5S)-5-(4-aminopvrrolo| 1,2-f| 11,2,41 triazin-7-yl)-4fluoro-3-hydroxv-5-methvltetrahvdrofiiran-2vl)methoxv)(phenoxv)phosphorvlamino)propanoatfc (Compound 5)
beta
The nucleoside 3 (0.026 g, 0.092 mmol) was dissolved in trimethylphosphate (2 mL) and cooled to 0°C. The mixture was stirred under N2(g) and 1-methylimidazole (0.062 mL, 0.763 mmol) followed by the chloridate A (0.160 g, 0.552 mmol) were added. The reaction mixture was stirred for 2 h. at 0°C and then allowed to warm slowly to RT. H2O (5 mL) was added to quench the reaction and then the mixture concentrated under reduced pressure. The residue was dissolved in CH2C12 and subjected to silica gel chromatography eluting with 0-100% EtOAc in hexanes. The product fractions were collected and concentrated. . Crude product was eluted using 0 to 100 percent EtOAc in hexanes. The crude product was collected and concentrated under reduced pressure. The residue was subjected to prep HPLC to yield 5 (2.0 mg, 4% yield).
113
LCMS m/z 538 [M+H].
«2 R, 3 R, 4 R, 5S)-5-(4-aminopvrrolo[1.2-f| [1.2.41 triazin-7-vl)-4-fluoro-3-hydroxv5-methvltetrahvdrofuran-2-vDmethvl tetrahydrogen triphosphate (Compound 6)
beta
The nucleoside 3 (0.022 g, 0.056 mmol) was dissolved in trimethylphosphate (1 mL) and stirred under N2(g). Phosphorous oxychloride (0.067 mL, 0.73 mmol) was added and the mixture stirred for 2 h. Monitoring by analytîcal ion-exchange column determined the time at which > 80 percent of monophosphate was formed. A solution of tributylamine (0.44 mL, 1.85 mmol) and triethylammonium pyrophosphate (0.327 g, 0.72 mmol) dissolved in anhydrous DMF (1 mL) was added. The reaction mixture was stirred for 20 min and then quenched by the addition of IN triethylammonium bicarbonate solution in H2O (5 mL). The mixture was concentrated under reduced pressure and the residue re-dissolved in H2O. The solution was subjected to ion ex change chromatography to yield the title product 6 (1.7 mg, 6% yield).
LCMS m/z 521 [M-H]. Tr = 0.41 HPLC ion exchange TR = 9.40 min (2R.3R.5S)-2-(4-aminopvrrolo[l,2-fl11.2.41 triazin-7-vl)-3-hydroxy-5(hydroxvmethvl)-tetrahvdrofuran-2-carbonitrile (Compound 7)
OAc
NaOH(aq)
THF/MeOH
OH •O 'Ό
114 ((3aR,5S,6aR)-2,2-dimethyl-tetrahydrofuro[2,3-d][l,3]dioxol-5-yI)methanol
The acetate material (1.2 g, 5.5 mmol) (J. Org. Chem. 1985, 50, 3547, De Bemardo et al) was dissolved in a 1:1 mixture MeOH and THF (10 mL). A IN solution of NaOH(aq) (lOmL) was added until the pH was 13. The reaction mixture was stirred for 2h and then neutralized to pH 8-9 by the addition of AcOH. The mixture was extracted with EtOAc (10 x 30mL) and the combined organic extracts dried over anhydrous NajSO.j, filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-70% EtOAc in hexanes to give the desired product (866 mg, 90%).
lH NMR (300 MHz, CDC13) δ 5.84 (d, J= 3.6 Hz, IH), 4.78 (t, 4.5 Hz, 1 H), 4.38 (m, IH), 3.93-3.54 (m, 2H), 2.04-1.84 (m, 2H), 1.52 (s, 3H), 1.33 (s, 3H).
NaH, BnBr
THF
OBn
(3aR,5S,6aR)-5-(henzyloxymethvl)-2,2-diinethyl-tetrahydrofuro|2,3d|[l,3]dioxole. Sodium hydride (188 mg, 7.46 mmol) was dissolved în anhydrous THF (5 mL) and stirred under N2(g) at RT. The alcohol (866 mg, 4.97 mmol) was dissolved in anhydrous THF (3 mL) and then added in portions over 5 min. to the sodium hydride mixture. The résultant mixture was stirred for 20 min. and then benzyl bromide (892 pL, 7.46 mmol) was added. The reaction was stirred for 2 h and then poured onto a mixture of ice cold aqueous NaHCO3 and EtOAc (30mL). The organic layer was separated and then the aqueous layer re-extracted with EtOAc (30 mL). The combined organic extracts were dried over anhydrous Na>SO.|. filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-40% EtOAc in hexanes to give the benzyl ether product (912 mg, 69%).
‘H NMR (300 MHz, CDC13) δ 7.35-7.27 (m, 5H), 5.86 (d, J= 3.6 Hz, IH), 4.74 (t, J =
4.2 Hz, IH), 4.60 (s, 2H), 4.42 (m, IH), 3.69-3.53 (m, 2H), 2.10-2.04 (m, IH), 1.831.77 (m, IH), 1.52 (s, 3H), 1.33 (s, 3H).
115
OBn
60°C
HOAc / H2O
(3R,5S)-5-(benzyloxymethyl)-tetrahydrofuran-2,3-diol. The benzyl ether (910 mg,
3.44 mmol) was dissolved in a 1:1 AcOH and H?O (20 mL) mixture and stirred at 60°C for 7h. The mixture was concentrated under reduced pressure and the residue subjected to silica gel chromatography eluting with 0-70% EtOAc in hexanes to give the diol product (705 mg, 91%).
‘H NMR (300 MHz, CDC13) δ 7.36-7.27 (m, 5H), 5.40 (d, J= 3.9 Hz, 0.5H), 5.17 (s, 0.5H), 4.67-4.56 (m, 3H), 4.33 (m, 0.5H), 4.24 (d, J= 4.8 Hz, 0.5H), 3.71-3.67 (m, IH), 3.56-3.42 (m, 2H), 2.31-2.22 (m, IH), 2.08-1.89 (m, 2H).
BnO BnO <γ·°·γ^ΟΗ Ag2CO3 / Celite l^/K^O 'OH Benzene, 80°C 'OH (3R,5S)-5-(benzyloxymethyl)-3-hydroxy-dihydrofuran-2(3H)-one. The diol (705 mg, 3.14 mmol) was dissolved in benzene (30 mL) and treated with a silver carbonate celite mixture (3.46 g, 6.28 mmol). The résultant mixture was stirred at 80°C under N2(g) for 2h. The mixture was then cooled to RT, filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-70% EtOAc in hexanes to give the lactone product (600 mg, 86%).
lH NMR (300 MHz, CDC13) δ 7.39-7.27 (m, 5H), 4.75-4.68 (m, IH), 4.60-4.49 (m, 2H), 3.74-3.54 (m, 2H), 2.61-2.35 (m, 2H), 2.38-2.28 (m, IH).
BnO
'OH
Ag2O
EtOAc
116 (3R, 5S)-3-(benzyloxy)-5-(benzyloxymethyl)-dihydrofuran-2(3H)-one. The lactone (600 mg, 2.7 mmol) was dissolved in EtOAc (30mL) and treated with sîlver oxide (626 mg, 2.7 mmol) followed by benzyl bromide (387 μL, 3.24 mmol). The reaction mixture was then stirred at 50°C under N2(g) for 8h. Additional silver oxide (300 mg) was then added and the résultant mixture stirred at 50°C for 16h. Additional benzyl bromide (50 uL) and silver oxide (150 mg) were added and the mixture stirred for an additional 8h. The réaction mixture was allowed to cool, filtered and then concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-20% EtOAc in hexanes to give the title product (742 mg, 88%).
lH NMR (300 MHz, CDC13) δ 7.39-7.27 (m, 10H), 4.99 (d, J= 11.4 Hz, 1 H), 4.72 (m, 2H), 4.56 (m, 2H),4.39 (t,-/ 8.1 Hz, IH), 3.72-3.51 (m, 2H), 2.42-2.25 (m, 2H).
(3lL5S)-2-(4-aininopyrrolo|l,2-f||l,2,4]triazin-7-yl)-3-(benzyloxy)-5(benzyloxymethyl)-tetrahydrofuran-2-ol. The 7-bromopyrrolo[l ,2-f][ 1,2,4]triazin-4amine (607 mg, 2.85 mmol) was dissolved in anhydrous THF (10 mL) and stirred under Ar(g) at RT. TMSC1 (1.1 mL, 8.55 mmol) was added dropwise and the mixture stirred for 2h. The reaction was concentrated under reduced pressure and then dried under high vacuum. The residue was suspended in THF (20 mL) and stirred under Ar(g) at -78°C. A 2.5M n-BuLi solution in hexane (2.28 mL, 5.7 mmol) was added dropwise over 10 min. and the résultant mixture stirred for 60 min. The lactone (742 mg, 2.37 mmol) dissolved in anhydrous THF (7 mL) was added to the above mixture over 20 min. The reaction mixture was stirred for 2 h. and then quenched with AcOH until pH was 5-6. The mixture was allowed to warm to RT and then diluted with EtOAc. The solution was washed with saturated NaHCO3 solution, saturated NaCl, dried over anhydrous Na2SÛ4 and concentrated under reduced pressure. The residue was subjected to silica
117 gel chromatography eluting with 0-80% EtOAc in hexanes to give the title product (250 mg, 24%).
LCMS m/z 447.2 [M+H], 445.1 [M-H],
nh2 nh2
/^1 N TMS-CN
Bn9 „ V-N. Bn9 Vn-
l__SL·/ N TMSOTf L Λ / N
/ /OH DCM __/CN
ÔBn -15°C ÔBn
(3R,5S)-2-(4-aminopy rroloj 1,2-f| [ 1,2,4] triazin-7-yl)-3-(benzyloxy)-5(benzyloxymethyl)-tetrahydrofuran-2-carbonitriie. The alcohol (250 mg, 0.56 mmol) was dissolved in anhydrous CH2Cl2(10 mL) and stirred under Ar(g) at -15°C.
TMSCN (448 liL, 3.36 mmol) was added dropwise and the mixture stirred for 10 min. TMSOTf (466 pL, 2.58 mmol) was added dropwise over 10 min and the résultant mixture stirred for 90 min. at -15°C. Additional TMSCN (224 pL, 3 eq.) and TMSOTf (202 pL, 2 eq.) was added and stirring continued for 5 h. Saturated aqueous NaHCO3 solution was added to quench the reaction and the mixture stirred for 10 min. The organic layer was separated and washed with saturated aqueous NaHCO3 solution, saturated NaCl solution, dried over anhydrous Na2S().| filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-70% EtOAc in hexanes to give the title product (150 mg, 59%).
LCMS m/z 456.3 [M+H], 454.1 [M-H].
118 (2R,3R.5S)-2-(4-aniinopyrrolo[l,2-f||l,2,4|tiiazin-7-yl)-3-hydroxy-5(hydroxy niethyl)-tetrahydrofuran-2-carbonitrile (7). The benzyl ether (150 mg,
0.329 mmol) was dissolved in anhydrous CH2CI2 (2 mL) and the mixture stirred under Ar(g) at -20°C. A IM BC13solution in CH2CI2 (724 pL, 0.724 mmol) was added dropwise and the résultant mixture stirred for 2h. Additional IM BC13 in CH2CI2 (724 μΙ„ 0.724 mmol) was added and stirring continued for 2h. The mixture was then cooled to -78°C and slowly treated with a 2:1 mixture of Et3N and MeOH (3 mL). The mixture was stirred for 10 min and then treated with MeOH (10 mL). The reaction was allowed to warm to RT and then concentrated under reduced pressure. The residue was dissolved in MeOH and concentrated under reduced pressure. The residue was dissolved in MeOH again and treated with solid NaHCO3. The mixture was stirred for 5 min and then the solid removed by filtration. The solution was concentrated under reduced pressure and subjected to préparative HPLC to provide the desired product 7 (10 mg, 11%).
'H NMR (300 MHz, D2O) δ 7.71 (s, IH), 6.75 (d, J= 4.5 Hz, IH), 6.65 (d, J= 4.8 Hz, IH), 4.91 (t, 7=6.3 Hz, IH), 4.57 (m, IH), 3.67-3.47 (m, 2H), 2.18 (m, 2H).
LCMS m/z 276.1 [M+H], 274.0 [M-H], (2S)-isopropyl 2-((((2 R.3S.4R,5R)-5-(4-aminopvrrolo 11.2-11 [1.2.4] triazin-7-vl)-5cvano-3.4-dihvdroxvtetrahvdrofuran-2-vl)methoxv)(phenoxvÎphosphorylaminolpropanoate (Compound 8)
NH
NH
The nucleoside 1 (45mg, 0.15mmol) was dissolved în anhydrous trimethyl phosphate (0.5 mL) and the solution stirred under N?(g) at 0°C. Methyl imidazole (36 liL. 0.45 mmol) was added to the solution. Chlorophosphoramidate C (69 mg, 0.225
119 mmol) was dissolved in anhydrous THF (0.25 mL) and added dropwise to the nucleoside mixture. When the reaction was complété by LCMS, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NaHCO3 solution, saturated NaCI, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0-5% MeOH in CH2Cl2 followed by préparative HPLC to give the product (20.9 mg, 25%). lH NMR (300 MHz, CD3OD) δ 7.95 (m, IH), 7.31-6.97 (m, 7H), 4.94 (m, IH), 4.78 (m, IH), 4.43 (m, 3H), 4.20 (m, IH), 3.80 (d, IH), 1.30-1.18 (m, 9H);
3iP NMR (121.4 MHz, CD3OD) δ 3.8.
LCMS m/z 561.0 [M+H], 559.0 [M-H], (2S)-2-eth ylbutyl 2-((((2 R.3S.4R.5R)-5-(4-aniinopvrrolo 11,2-f| [ 1,2,41 triazin-7-yl)-5cyano-3,4-dihvdroxvtetrahvdrofuran-2vl)methoxvXphenoxv)phosphorvlamino)propanoate (Compound 9)
Prepared from Compound 1 and chloridate B according to the same method as for the préparation of compound 8.
'H NMR (300 MHz, CD3OD) δ 7.87 (m, IH), 7.31-7.16 (m, 5H), 6.92-6.89 (m, 2H), 4.78 (m, IH), 4.50-3.80 (m, 7H), 1.45-1.24 (m, 8H), 0.95-0.84 (m, 6H).
31P NMR (121.4 MHz, CD3OD) δ 3.7.
LCMS m/z 603.1 [M+H], 601.0 [M-H].
120 (2S)-ethyl 2-((((2 R,3 S,4 R,5R)-5-(4-aminopyrrolo[ l,2-f| 11,2.41 triazin-7-yl)-5-cyano3,4-dihydroxytetrahvdrofuran-2vl)methoxv)(phenoxy)phosphorvlamino)propanoate (Compound 10)
Prepared from Compound 1 and chloridate A using same method as for the préparation of compound 8.
lH NMR (300 MHz, CD3OD) δ 7.95 (m, IH), 7.32-6.97 (m, 7H), 4.78 (m, IH), 4.434.08 (m, 6H), 3.83 (m, 1 H), 1.31 -1.18 (m, 6H).
3IP NMR (121.4 MHz, CD3OD) δ 3.7.
LCMS m/z 547.0 [M+H], 545.0 [M-H].
(2Sl-ethvl 2-((((2R,3R,4R,5R)-5-(4-aininopvrrolo[1.2-fHl,2,4Îtriazin-7-vl)-5-cvano4-fluoro-3-hvdroxvtetrahvdrofuran-2vl)methoxv)(phenoxv)phosphorylamino)propanoate (Compound 11)
Compound 11 was prepared from Compound 2 and chloridate A using same method as for the préparation of compound 8.
lH NMR (300 MHz, CD3OD) δ 7.91 (m, IH), 7.33-7.16 (m, 5H), 6.98-6.90 (m, 2H), 5.59 (m, IH), 4.50-4.15 (m, 4H), 4.12-3.90 (m, 3H), 1.33-1.18 (m, 6H).
3IP NMR (121.4 MHz, CD3OD) δ 3.8.
121
LCMS m/z 549.0 [M+H], 547.1 [M-H].
(ZS^'Sl-dicthvI 2,2t-((((2R.3S,4R,5R)-5-(4-aminopvrrolo|1.2-flll.2.41triazin-7-yl)5-cyano-3,4-dihvdroxvtetrahydrofuran-2vl)methoxv)phosphorvl)bis(azanediyl)dipropanoate (Compound 12)
The nucleoside 1 (14.6 mg, 0.05 mmol) was dissolved in anhydrous trimethyl phosphate (0.5 mL) and stirred under N2(g) at RT. POCI3 (9.2 pL, 0.1 mmol) was added and the mixture stirred for 60 min. Alanine ethyl ester hydrochloride (61 mg, 0.4 mmol) and then EÎ3N (70 pL, 0.5 mmol) was added. The résultant mixture was stirred for 15 min. and then additional Et3N (70 pl, 0.5 mmol) was added to give a solution pH of 9-10. The mixture was stirred for 2 h. and then diluted with EtOAc, washed with saturated aqueous NaHCO3 solution followed by saturated aqueous NaCl solution. The organic layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was subjected to préparative HPLC (Cig column) to yield the product 12 (5.5 mg, 16%).
‘H NMR (400 MHz, CD3OD) δ 8.13 (s, IH), 7.41 (d, J= 4.8 Hz, IH), 7.18 (d, J= 4.8 Hz, IH), 4.78 (d, J= 5.6 Hz, IH), 4.36 (m, IH), 4.25-4.08 (m, 7H), 3.83 (m, 2H), 1.331.23 (m, 12H).
3 lP NMR ( 121.4 MHz, CD3OD) δ 13.8. LCMS m/z 570.0 [M+H], 568.0 [M-H].
(2S,3R,4S,5R)-2-(4-aniinopvrrolo[l,2-f| [ 1.2.4|triazin-7-vl)-2-ethvnyl-5(hvdroxvmethvl)tetrahvdrofuran-3.4-diol (Compound 13)
122
The nucleoside alcohol (0.6 g, 1.08 mmol) (prepared as described in Compound 1 synthesis) was dissolved in anhydrous THF (8mL) and placed under N?(g). The reaction mixture was stirred and cooled to 0°C and then treated with a 0.5N solution of ethynyl magnésium bromide in THF (17.2 mL, 17.2 mmol). The reaction mixture was stirred overnight at RT. AcOH (1.5 mL) was added to quench the reaction. The mixture was concentrated under reduced pressure and the residue redissolved in CH2CI2. The solution subjected to a plug of silca gel eluting with 0 to 80% EtOAc in Hexanes to provide the title product as a crude mixture.
LCMS m/z 579 [M+H].
The crude ethynyl alcohol (0.624 g, 1.08 mmol) was dissolved in anhydrous CH2CI2 (10 mL) and placed under N2(g). The mixture was stirred and sulfonic acid (0.2 mL, 2.74 mmol) was added. The reaction mixture was stirred for 12 h. at RT. When complété by LCMS, Et3N (0.56 mL) was added to quench the reaction. The reaction was concentrated under reduced pressure and the residue subjected to silica gel chromatography eluting with 0 to 75% EtOAc in Hexanes to yield the ethynyl nucleoside as a mixture of anomers (0.200 g, 33% over 2 steps).
LCMS m/z 561 [M+H].
123
The tribenzyl nucleoside (0.650 g, 1.16 mmol) was dissolved in anhydrous CH2C12 (30 mL) and cooled to -78°C under N2(g). A solution of boron tribromîde (1 N in CH2C12, 5.5 mL) was added and the reaction mixture stirred for 1 h. at -78°C. A solution of MeOH (10 mL) and pyridine (2 mL) was added to quench the reaction and the mixture was allowed to rise to RT. The mixture was concentrated under reduced pressure and subjected to préparative HPLC to provide the a-anomer (20 mg) and βanomer 13(110 mg) (β -anomer) ’H NMR (300 MHz, DMSO) δ 7.81 (s, 1H), 7.76 (brs, 2H), 6.80-6.85 (m, 2H), 5.11 (d, J = 7.2 Hz, 1H), 4.90 (d, J = 6.0 Hz, 1H), 4.82 (dd, J = 7.2, 4.8 Hz, 1H), 4.62 (t, J = 6.3 Hz, 1H), 3.95-3.99 (m, 1H), 3.85-3.91 (dd, J = 11.4, 5.7 Hz, 1H), 3.61-
3.67 (m, 1H), 3.47-3.55 (m, 1H), 3.52 (d, J = 0.9 Hz, 1H).
(a -anomer) ’H NMR (300 MHz, DMSO) δ 7.80 (s, 1H), 7.59 (bs, 2H), 6.80 (d, J = 4.5 Hz, 1H), 6.54 (d, J = 4.2 Hz, 1H), 5.00 (d, J = 7.2 Hz, 1H), 4.89 (d, J = 4.8 Hz, 1 H),
4.74 (t, J = 5.7 Hz, 1H), 4.58 (t, J = 4.5 Hz, 1H), 4.27 (m, 1H), 3.88 (m, 1H), 3.64-3.72 (m, 1H), 3.51-3.59 (m, 1 H), 3.48 (d,J = 0.6 Hz, 1H)
LCMS m/z 291 [M+H].
(2R,3R,4R)-5-(4-aminopyrrolo[1.2-f| I L2,4|triazin-7-vl)-l ,3.4tris(benzvloxv)hexane-2,5-diol (Compound 14)
124
The tribenzyl alcohol from Compound 1 synthesis (0.250 g, 0.453 mmol) was dissolved in anhydrous THF (25 mL) and stirred under N2(g). The reaction mixture was cooled to 0'C and then a 3.0 N solution of methyl magnésium chloride in THF(1.2 mL, 3.62 mmol) was added. The reaction mixture was stirred overnight at RT. Acetic acid (1.5 mL) was added to quench the reaction and then the mixture was concentrated under reduced pressure. The residue was redissoved in CH2CI2 and subjected to a plug of silca gel eluting with 0 to 80% EtOAc in hexanes. The crude product (0.452 g) was then used in the next reaction without further purification.
LCMS m/z 569 [M+H].
The crude methyl nucleoside (0.452 g, 0.796 mmol) was dissolved in anhydrous CH2CI2 (20 mL) and stirred under N2(g). Methanesulfonic acid (0.2 mL, 2.78 mmol) was added and the reaction stirred for 12 hr at RT. Et3N (0.56 mL) was added to quench the reaction and then the mixture concentrated under reduced pressure. The residue was subjected to silica gel chromatography eluting with 0 to 75% EtOAc in Hexanes to yield the product as a mixture of anomers (0.20 g, 46% over 2 steps). LCMS m/z 551 [M+H].
125
The tribenzyl nucleoside (0.20 g, 0.364 mmol) was dissolved in AcOH (30 mL). and charged with Pd/C (Degussa) (400 mg). The stirred mixture was flushed with N2(g) three times and then H2 (g) was introduced, The reaction was stirred under H2 (g) for 2 h. and then the catalyst removed by filtration. The solution was concentrated under reduced pressure and under the residue was re-dissolved in H2O. The solution was subjected to préparative HPLC under neutral conditions to provide the a-anomer and β-anomer 14 in 81% yield.
(α-anomer) ]H NMR (300 MHz, D2O) ôüü 7.81 (s, IH), 7.22 (d, IH), 6.75 (d, IH), 4.47 (d, IH), 4.25-4.31 (m, 1 H), 3.88-4.95 (m, IH), 3.58-3.86 (dd, 2H), 1.50 (s, 3H). (β-anomer) ‘H NMR (300 MHz, D2O) 0007.91 (s, IH), 7.26 (d, IH), 6.90 (d, IH), 4.61 (d, IH), 4.00-4.09 (m, 2H), 3.63-3.82 (dd, 2H), 1.67 (s, 3H).
LCMS m/z 281 [M+H],
S,Sl-2<2t-((((2R,3S,4R,5R)-5-(4-aminopvrroloil,2-fin,2,41triazin-7-vl)-5-cvano-3,4dihvdroxvtetrahvdrofnran-2-yl)methoxv)phosphorvl)bis(oxv)bis(ethane-2.1-divl) bis(2,2-dimethylpropanethioate) (Compound 15)
The nucleoside 1 (0.028 g, 0.096 mmol) was dissolved in trimethylphosphate (1 mL). The reaction was stirred under N2(g) and then treated with lH-tetrazole (0.021 g, 0.29 mmol). The reaction mixture was cooled to 0°C and the phosphane (Nucleoside Nucléotides, Nucieic acids; 14; 3-5; 1995; 763 - 766. Lefebvre, Isabelle; Pompon, Alain; Perigaud, Christian; Girardet, Jean-Luc; Gosselin, Gilles; et al.) (87 mg, 0.192 mmol) was added. The reaction was stirred for 2 h. and then quenched with 30% hydrogen peroxide (0.120 mL). The mixture was stirred for 30 min at RT and then treated with saturated aqueous sodium thiosulfate (1 mL). The mixture was stirred for
126 min. and then concentrated under reduced pressure. The residue was subjected to préparative HPLC to isolate the title product 15.
lH NMR (300 MHz, CD3CN) ÔDU 7.98 (s, 1H), 6.92 (d, 1H), 6.81 (d, 1H), 6.44 (bs,
2H), 4.82 (m, 2H), 4.47 (m, 1 H), 4.24 (m, 2H), 4.00 (m, 4H), 3.80 (bs, 1 H), 3.11 (m,
4H), 1.24 (s, 9H).
3IP NMR (121.4 MHz, CD3CN) δ -1.85 (s).
LCMS m/z 661 [M+H].
S,S,-2,2’-((((2R, 3S, 4R. 5S)-5-(4-aminopyrrololl,2-f| 11,2,4 ]triazin-7-vl)-5-ethynyl-
3,4-dihvdroxvtetrahvdrofïiran-2-vl)methoxv)phosphorvl)bis(oxv)bis(ethane-2.1divD bis(2,2-dimethylpropanethioate) (Cornpound 16)
Cornpound 16 was prepared using the same method as cornpound 15 except substituting cornpound 13 as the starting nucleosîde.
’HNMR (300 MHz, CD3CN) δ 0 07.91 (s, 1H), 6.86 (d, J = 4,8 Hz, 1H), 6.76 (d, J =
4.5 Hz, 1H), 6.29 (bs, 2H), 4.69 (t, J = 2.7 Hz, 1H), 4.58 (d, J = 5.7 Hz, 1H), 4.14-4.33 (m, 5H), 3.99-4.07 (m, 4H), 3.53 (d, J = 5.4 Hz, 1H), 3.11 (q, J = 5.7 Hz, 4H), 1.22 (s, 18H).
LCMS m/z 658.9 [M+]. Tr=2.31 ((2R, 3S, 4R, 5R)-5-(4-aminopyrrolo|l,2-f]| 1,2,41 triazin-7-vl)-5-cyano-3,4dihvdroxvtetrahydrofuran-2-vDmethyl tetrahydrogen triphosphate (Cornpound , 17) kX'
127
Compound 17 was prepared from compound 1 using a similar procedure to the préparation of compound 6. The product was isolated as the sodium sait.
‘H NMR (400 MHz, D2O) δ 7.76 (s, IH), 6.88 (d, J = 4.8 Hz, IH), 6.73 (d, J = 4.4 Hz, 1 H), 4.86 (d, J = 5.2 Hz, 1H),4.43 (m, IH), 4.39 (m, IH), 4.05 (m, IH), 3.94 (m, IH) 31P NMR (121.4 MHz, EhO) δ D-5.4(d, lP),-10.8(d, IP), -21.1 (t, IP).
LCMS m/z 530 [M-H], 531.9 [M+H] Tr = 0.22 min
HPLC ion exchange Tr=9.95 min t(2R, 3S, 4R, 5S)-5-(4-aimnopvrrolo[L2-fHL2<41triazin-7-vD-5-ethvnvl-3.4dihvdroxvtetrahvdrofuran-2-vDmethyl tetrahydrogen triphosphate (Compound 181
Compound 18 was prepared from compound 13 using a similar procedure to the préparation of compound 6. The product was isolated as the TEA sait.
‘H NMR (300 MHz, D2O) δ 7.85 (s, IH), 7.09 (d, J = 4.6 Hz, IH), 6.95 (d, J = 4.7 Hz, IH), 4.23 (m, 2H), 4.08 (m, 2H), 3.06 (q, J = 7.4 Hz, 20H), 1.14 (t, J = 7.3 Hz, 30H) 31P NMR (121.4 MHz, D2O) δΠ -10.8 (d, 1 P), -11.2 (d, IP), -23.2 (t, IP).
128
LCMS m/z 530.8 [M+H], Tr = 0.46
HPLC ion exchange Tr = 9.40 min ((2R, 3S, 4R, 5S)-5-(4-aminoBvrrololL2-t1IL2.4|triazin-7-vl)-3.4-dihvdroxv-5methyltetrahvdrofuran-2-yl)methyl tetrahydrogen triphosphate (Compound 19)
Compound 19 was prepared from compound 14 using a similar procedure to the préparation of compound 6.
H NMR (400 MHz, D2O) δ 7.78 (s, IH), 6.98 (m, IH), 6.84 (m, IH), 4.45 (m, IH), 4.04 (m, 4H), 1.54 (s, 3H).
31P NMR (161 MHz, D2O) δ -10.6 (m), -23.0 (m).
LCMS m/z 521.0 [M+H].
((2R,3R,4R,5R)-5-(4-aminopyrrolo[l,2-f|(lî2,41triazin-7-yl)-5-cyano-4-fluoro-3hydroxytetrahydrofuran-2-yl)methyl tetrahydrogen triphosphate (Compound 20)
Compound 20 was prepared from compound 2 using a similar procedure to the préparation of compound 6. aZ
129 'H NMR (400 MHz, D2O) δ 7.78 (s, IH), 6.93 (d, J = 4.4 Hz, IH), 6.78 (d, J = 4.8 Hz,
IH), 5.45 (dd, J = 53,4.4 Hz, lH), 4.38-4.50(m, 2H), 4.13-4.20 (m, 2H).
3,PNMR(161 MHz, D2O) δ-5.7 (d, lP),-11.0(d, IP), -21.5 (t, IP).
LCMS m/z 533.9.0 [M+H], 532.0 [M-H] Tr = 1.25 min.
HPLC ion exchange Tr=l 1.0 min
Antiviral Activity
Another aspect of the invention relates to methods of inhibiting viral infections, comprising the step of treating a sample or subject suspected of needing such inhibition with a composition of the invention.
Within the context of the invention samples suspected of containing a virus include natural or man-made materials such as living organisms; tissue or cell cultures; biological samples such as biological material samples (blood, sérum, urine, cerebraspinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like. Typically the sample will be suspected of containing an organism which induces a viral infection, frequently a pathogenic organism such as a tumor virus. Samples can be contained in any medium including water and organic solventVwater mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures.
If desired, the anti-virus activity of a compound of the invention after application of the composition can be observed by any method including direct and indirect methods of detecting such activity. Quantitative, qualitative, and semiquantitative methods of determining such activity are ail contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation of the physiological properties of a living organism are also applicable.
The antiviral activity of a compound of the invention can be measured using standard screening protocols that are known. For example, the antiviral activity of a
130 compound can be measured using the following general protocols.
Respiratory syncytial virus (RSV) antiviral activity and cytotoxicity assays
Anti-RSV activity
Antiviral activity against RSV is determined using an in vitro cytoprotection assay in Hep2 cells. In this assay, compounds inhibiting the virus réplication exhibit cytoprotective effect against the virus-induced cell killing that can be quantified using a cell viability reagent. The method used is similar to methods previously described în publîshed literature (Chapman et al., Antimicrob Agents Chemother. 2007,57(9):334653.)
Hep2 cells are obtained from ATCC (Manassas, VI) and maintained in MEM media supplemented with 10% fêtai bovine sérum and penicillin/streptomycin. Cells are passaged twice a week and kept at subconfluent stage. Commercial stock of RSV straîn A2 (Advanced Biotechnologies, Columbia, MD) is tîtered before compound testing to détermine the appropriate dilution of the virus stock that generates désirable cytopathic effect in Hep2 cells.
For antiviral tests, Hep2 cells are seeded into 96-well plates 24 hours before the assay at a density of 3,000 cells/well. On a separete 96well plate, compounds to be tested are serially diluted in cell culture media. Eight concentrations in 3-fold serial dilution incréments are prepared for each tested compound and 100 uL/well of each dilution is transferred in duplicate onto plates with seeded Hep2 cells. Subsequently, appropriate dilution of virus stock previously determined by titration is prepared in cell culture media and 100 uL/well is added to test plates containing cells and serially diluted compounds. Each plate includes three wells of infected untreated cells and three wells of uninfected cells that served as 0% and 100% virus inhibition control, respectively. Following the infection with RSV, testing plates are incubated for 4 days in a tîssue culture incubator. After the incubation, RSV-înduced cytopathic effect is determined using a Cell TiterGlo reagent (Promega, Madison, WI) followed by a 1A/^
131 luminescence read-out. The percentage inhibition is calculated for each tested concentration relative to the 0% and 100% inhibition controls and the EC50 value for each compound is determined by non-linear régression as a concentration inhibiting the
RSV-induced cytopathic effect by 50%. Ribavirin (purchased from Sigma, St. Louis,
MO) is used as a positive control for antiviral activity.
Cytotoxicity
Cytotoxicity of tested compounds is determined in uninfected Hep2 cells in parallel with the antiviral activity using the cell viability reagent in a similar fashion as described before for other cell types (Cihlar et al., Antimicrob Agents Chemother. 2008,52(2):655-65.). The same protocol as for the détermination of antiviral activity is used for the measurement of compound cytotoxicity except that the cells are not infected with RSV. Instead, fresh cell culture media (100 uL/well) without the virus is added to tested plates with cells and prediluted compounds. Cells are then incubated for 4 days followed by a cell viability test using CellTiter Glo reagent and a luminescence read-out. Untreated cell and cells treated with 50 ug/mL puromycin (Sigma, St. Louis, MO) are used as 100% and 0% cell viability control, respectively. The percent of cell viability is calculated for each tested compound concentration relative to the 0% and 100% controls and the CC50 value is determined by non-linear régression as a compound concentration reducing the cell viability by 50%.
Compound EC501 uM CC50 / uM
1 0.48 >100
10 0.18 47
12 6.5 >100
13 34 >100
14 2.7 92
15 0.15 >100
16 3.3 >100
RSV RNP Préparation
132
RSV ribonucleoprotein (RNP) complexes were prepared from a method modified from Mason et a/(l). HEp-2 cells were plated at a density of 7.1 x 104 cells/cm in MEM + 10% fêtai bovine sérum (FBS) and allowed to attach overnight at 37°C (5% CO2). Following attachment, the cells were infected with RSV A2 (MOI=5) in 35 mL MEM + 2% FBS. At 20 hours post-infection, the media was replaced with MEM + 2% FBS supplemented with 2 pg/mL actinomycin D and retumed to 37°C for one hour. The cells were then washed once with PBS and treated with 35 mL of PBS + 250 pg/mL lyso-lecithin for one minute, after which ail liquid was aspirated. The cells were harvested by scrappîng them into 1.2 mL of buffer A [50 mM TRIS acetate (pH 8.0), 100 mM potassium acetate, 1 mM DTT and 2 pg/mL actinomycin D] and lysed by repeated passage through an 18 gauge needle (10 times). The cell lysate was placed in îce for 10 minutes and then centrifuged at 2400g for 10 minutes at 4°C. The supematant (SI) was removed and thepellet (Pl) was disrupted in 600 uL of Buffer B [10 mM TRIS acetate (pH 8.0), 1 OmM potassium acetate and 1.5 mM MgCl2] supplemented with 1% Triton X-100by repeated passage through an 18 gauge needle (10 times). The resuspended pellet was placed in ice for 10 minutes and then centrifuged at 2400g for 10 minutes at 4°C. The supematant (S2) was removed and the pellet (P2) was disrupted in 600 uL of Buffer B supplemented with 0.5% deoxycholate and 0.1% Tween 40. The resuspended pellet was placed in ice for 10 minutes and then centrifuged at 2400g for 10 minutes at 4°C. The supematant (S3) fraction, containing the enriched RSV RNP complexes, was collected and the protein concentration determined by UV absorbance at 280 nm. Aliquoted RSV RNP S3 fractions were stored at -80°C.
RSV RNP Assay
Transcription réactions contaïned 25pg of crude RSV RNP complexes in 30 pL of reaction buffer [50 mM TRIS-acetate (pH 8.0), 120 mM potassium acetate, 5% glycerol, 4.5 mM MgCl2, 3 mM DTT, 2 mM ethyleneglycol-bis(2-aminoethylether)tetraacetic acid (EGTA), 50 pg/mL BSA, 2.5 U RNasin (Promega), ATP, GTP, UTP, CTP and 1.5 uCi [α-32Ρ] NTP (3000 Ci/mmol)]. The radiolabled nucléotide used in the transcription assay was selected to match the nucléotide analog being evaluated for y/'
133 inhibition of RSV RNP transcription. Cold, compétitive NTP was added at a final concentration of one-half its Km (A I P= 20 μΜ, GTP= 12.5 μΜ, UTP= 6 μΜ and CTP= μΜ). The three remaining nucléotides were added at a final concentration of 100 μΜ.
To détermine whether nucléotide analogs inhibited RSV RNP transcription, compounds were added using a 6 step serial dilution in 5-fold incréments. Following a 90 minute incubation at 30°C, the RNP reactions were stopped with 350 pL of Qiagen RLT lysis buffer and the RNA was purified using a Qiagen RNeasy 96 kit. Purified RNA was denatured in RNA sample loading buffer (Sigma) at 65°C for 10 minutes and run on a 1.2% agarose/MOPS gel containing 2M formaldéhyde. The agarose gel was dried and exposed to a Storm phosphorimager screen and developed using a Storm phosphorimager (GE Healthcare). The concentration of compound that reduced total radiolabled transcripts by 50% (IC50) was calculated by non-linear régression analysis of two replicates.
Reference
1) Mason, S., Lawetz, C., Gaudette, Y., Do, F., Scouten, E., Lagace, L., Simoneau, B. and Liuzzi, M. (2004) Polyadenylation-dependent screening assay for respiratory syncytial virus RNA transcriptase activity and identification of an inhibitor. Nucleic Acids Research, 32, 4758-4767.
Compound IC50 / uM
6 3.6
17 1.5
18 1.6
19 1.5
20 0.8
Description of the Parainfluenza Cytoprotection Assay
The Parainfluenza Cytoprotection assay uses Vero cells and Parainfluenza 3 strain C 243. Briefly virus and cells are mixed in the presence of test compound and incubated for 7 days. The virus is pre-titered such that control wells exhibit 85 to 95% loss of cell viabîlity due to virus réplication. Therefore, antiviral effect or
134 cytoprotection is observed when compounds prevent virus réplication. Each assay plate contains cell control wells (cells only), virus control wells (cells plus virus), compound toxicity control wells (cells plus compound only), compound colorimétrie control wells (compound only), as well as experimental wells (compound plus cells plus virus). Cytoprotection and compound cytotoxicity are assessed by MTS (CellTiter®96 Reagent, Promega, Madison WI) dye réduction. The % réduction in viral cytopathic effects (CPE) îs determined and reported; IC5o (concentration inhibiting virus réplication by 50%), TC50 (concentration resulting in 50% cell death) and a calculated TI (therapeutic index TC5()/ IC5o) are provided along with a graphical représentation of the antiviral activity and compound cytotoxicity when compounds are tested in doseresponse. Each assay includes ribavirin as a positive control.
Cell Préparation
Veto cells (Kidney, African green monkey, Cercopithecus aethiops) were obtained from the American Type Culture Collection (ATCC, Rockville, Maryland) and are grown in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% fêtai bovine sérum (FBS), 2.0 mM L-Glutamine, 100 units/ml Penicillin and 100 ug/ml Streptomycin (“growth medium”). Cells are sub-cultured twice a week at a splît ratio of 1:10 using standard cell culture techniques. Total cell number and percent viability déterminations are performed using a hemacytometer and trypan blue exclusion. Cell viability must be greater than 95% for the cells to be utilized in the assay. The cells are seeded in 96-well tissue culture plates the day before the assay at a concentration of 1 x 104 cells/well.
Virus Préparation
The virus used for this assay is Parainfluenza 3 strain C 243. This virus was obtained from the American Type Culture Collection (ATCC) and was grown in Vero cells for the production of stock virus pools. For each assay, a pre-titered aliquot of virus is removed from the freezer (-80°C) and allowed to thaw slowly to room température in a biological safety cabinet. The virus is resuspended and diluted into kA-'
135 tissue culture medium such that the amount of virus added to each well is the amount determined to give between 85 to 95% cell killing at 6-7 days post-infection.
MTS Staining for Cell Viability
At assay termination (7 days post-infection), the assay plates are stained with the soluble tetrazolium-based dye MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium; CellTiter®96 Reagent, Promega) to détermine cell viability and quanti fy compound toxicity. MTS is metabolized by the mitochondrial enzymes of metabolically active cells to yield a soluble formazan product, allowing the rapid quantitative analysis of cell viability and compound cytotoxicity. This reagent is a stable, single solution that does not require préparation before use. At termination of the assay, 20-25 pL of MTS reagent is added per well and the microtîter plates are then incubated for 4-6 hrs at 37°C, 5% CO? to assess cell viability. Adhesive plate sealers are used in place of the lîds, the sealed plate is inverted several times to mix the soluble formazan product and the plate is read spectrophotometrically at 490/650 nm with a Molecular Devices Vmax or SpectraMax Plus plate reader.
Data Analysis
Using an în-house computer program % Cytopathîc Effect (CPE) Réduction, %Cell Viability, IC25, IC5t), IC95, TC25, TC50, and TC95 and other indices are calculated and the graphical results summary is displayed. Raw data for both antiviral actîvity and toxicity with a graphical représentation of the data are provided in a printout summarizing the individual compound actîvity. The Table below shows the actîvity of selected compounds against Parainfluenza 3 virus.
Compound IC50 / uM TC50 / uM
1 1.71 >30
14 5.23 >30
The spécifie pharmacological and biochemical responses observed in the assays described may vary according to and depending on the particular active compound selected AJ
136 or whether there are présent pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or différences in the results are contemplated in accordance with practice of the présent invention.
Ail publications, patents, and patent documents cited herein above are incorporated by reference herein, as though individually incorporated by reference.
The invention has been described with reference to various spécifie and preferred embodiments and techniques. However, one skilled in the art will understand that many variations and modifications may be made while remaining within the spirit and scope of the invention,

Claims (33)

  1. What is claimed is
    1. The compound of Formula I for use in treating a Paramyxoviridae infection, wherein the compound of Formula I has the structure:
    R
    Formula I or a pharmaceutically acceptable sait or ester, thereof; wherein:
    each R1 is H or halogen;
    each R2, R3 or R5 is independently H, ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Ci-Cg)alkyl, (C4-Cg)carbocyclylalkyl, (Ci-Cg)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl or (C2-Cg)substituted alkynyl;
    or any two R , R or R on adjacent carbon atoms when taken together are O(CO)O- or when taken together with the ring carbon atoms to which they are attached form a double bond;
    R6 is ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, -C(-O)RN, -C(=O)OR11, C(=O)NR1!R12, -C(=O)SRh, -S(O)Rh, -S(O)2R“, -SiOXOR11), -S(O)2(ORh), -SO2NRllR12, halogen, (C|-Cs)alkyl, (Ci-Cgjcarbocyclylalkyl, (Ci-Cg)substituted alkyl, (C2 Q)alkenyl, (C2-C8)substituted alkenyl, (C2-Cg)alkynyl, (C2-Cg(substituted alkynyl, or aryl(CrC\)alkyl;
    each n is independently 0, 1, or 2;
    138 each Ra îs independently H, (Ci-Cg)alkyl, (C2-C|<)alkenyl, (C2-Cs)alkynyl, aryl(Ci-C8)alkyl, (C.t Cg)carbocyclylalkyl. -C(=O)Rli, -C(=O)ORn, -C(=O)NRllR12, C(=O)SRl,,-S(O)RH,-S(O)2R,-S(O)(OR11), -StOHOR1‘), or-S02NRllR12;
    R7 is H, -C(=O)R11, -C(=O)ORH, -C(=O)NR1,R,2î -C(=O)SR“, -S(0)Rn, 5 S(0)2R11, -S(O)(OR]l), -SiOJ^OR1'), -SO2NR1,R,2) or
    Y
    Il s w2 each Y or Y1 is, independently, O, S, NR, +N(0)(R), N(0R), +N(0)(0R), or N-NR2;
    W1 and W2, when taken together, are -Y3(C(Ry)2)3Y3-; or one of W1 or W2
    10 together with either R3 or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of the Formula la:
    M2
    Formula la wherein:
    15 each Y2 is independently a bond, O, CR2, NR, +N(O)(R), N(OR), +N(O)(OR),
    N-NR2, S, S-S, S(0), or S(0)2;
    each Y3 is independently O, S, or NR;
    M2 isO, 1 or 2;
    each R* is independently Ry or the formula:
    139 wherein:
    each Ml a, Mlc, and Mld is independently 0 or l;
    Ml2c is 0, l, 2, 3,4, 5, 6, 7, 8, 9, 10,11 or 12;
    each Ry is independently H, F, Cl, Br, I, OH, R, -C(=y')R, -C(=Y])OR, C(=Y!)N(R)2, -N(R)2, -+N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(OR), -S(O)2(OR), OC(=y')R, -OC(=Y')OR, -OC(=Y1)(N(R)2), -SC(=Y')R, -SC(=Y')OR, SC(=Y')(N(R)2), -N(R)C(=Y')R, -N(R)C(=Y')OR, -N(R)C(=Yl)N(R)2, -so2nr2, -CN, -N3, -NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
    each R is independently H, (C]-C8) alkyl, (C]-C8) substituted alkyl, (C2C8)alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl, (C2-C«) substituted alkynyl, C6-C2o aryl, C6-C2o substituted aryl, C2-C2o heterocyclyl, C2-C2o substituted heterocyclyl, arylalkyl or substituted arylalkyl;
    W3 is W4 or Ws; W4 is R, -C(Y*)Ry, -C(Y’)W5, -SO2Ry, or -SO2W5; and W5 is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups;
    each R8 is halogen, NRllR12, NtR'^OR11, NRI1NR11R12, N3, NO, NO2, CHO, CN, -CH(=NRn), -CH=NNHR11, -CH=N(OR11), -CH(OR)2, -C(=O)NRllR12, -C(=S)NRllR12, -C(=O)OR11, (Ci-Cg)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C4~C8)carbocyclyIalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=O)(Ci-Cs)alkyl, -S(O)n(C]-C8)alkyl, aryl(Ci-C8)alkyl, OR11 or SR11;
    each R9 or R10 is independently H, halogen, NRnR12, N(RH)0R11, NR1INRUR12, N3, NO, NO CHO, CN, -CH(=NR11), -CH=NHNR11, -CH=N(ORU), -CHtOR11)^ -C(=O)NRllR12, -C(=S)NRr12, -C(=O)ORn, R11, OR11 or SR11; and tAZ~
    140 each Rli or R12 is independently H, (C[-C8)alkyl, (C2-C8)alkenyl, (C2C8)alkynyl, (C4-C8)carbocyclyl alkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(C|-C8)alkyl, -S(O)n(C|-C8)alkyl or aryl(Cj-C8)alkyl; or R11 and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or -NRa-; and wherein each (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(C[-C8)alkyl of each R , R , R , R , R or R is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or OR1*; and wherein one or more of the nonterminal carbon atoms of each said (Ci-Cg)alkyl may be optionally replaced with -O-, S- or -NRa-.
  2. 2. The compound for use according to claim 1 wherein the compound of Formula I is represented by Formula II:
    R3 R2
    Formula II or a pharmaceutically acceptable sait or ester, thereof;
    wherein:
    each R1 is H or halogen;
    each R2 is ORa or halogen;
    each R3 or R5 is independently H, ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Ci-Cs)alkyl, (C4-C8)carbocyclylalkyl, (Ci-C8)substituted alkyl, (C2-C8)alkenyl, (C2-C8)substîtuted alkenyl, (C2-C8)alkynyl or (C2-C8)substituted alkynyl;
    141 or any two R2, R3 or R5 on adjacent carbon atoms when taken together are O(CO)O- or when taken together with the ring carbon atoms to which they are attached form a double bond; and
    R6 is ORa, N(Ra)2, N3, CN, S(O)nRa, -C(=O)R' ‘, -C(=O)ORU, -C(=O)NR11R12, C(=O)SRU, -S(O)Rn, -S(O)2R, -S(O)(ORh), -S(O)2(ORH), ~SO2NRhR12, halogen, (Ci-Cg)alkyl, (C4--Cgjcarbocyclylalkyl, (C]-Cg)substituted alkyl, (C2--Cg)alkenyl, (Ci -C'sjsubstitutcd alkenyl, (C2-Cs)alkynyl, or (C2-C8)substituted alkynyl.
  3. 3. The compound for use according to claims 1 or 2 wherein the compound of Formula I or II is represented by Formula III:
    Formula III or a pharmaceutically acceptable sait or ester, thereof;
    wherein:
    each R2 is ORa or F;
    each R3 is ORa; and
    R6 is ORa, N(Ra)2, N3, CN, S(O)nRa, -C(=O)Rh, -C(=O)ORn, -C(=O)NRR12, C(=O)SR11, -S(O)R, -SfOhR11, -S(O)(OR11), -S(O)2(OR1!), -SOîNR11^2, halogen, (Ci-Cs)alkyl, (C4--Cg/carbocyclylalkyl, (Ci Q/substituted alkyl, (C2-C8)alkenyl, (C2-C8)substituted alkenyl, (C2-C8)alkynyl, or (Cj-CgJsubstituted alkynyl. Ά'
    142
  4. 4. The compound for use according to any one of claims 1-3 wherein R6 is CN, methyl, ethenyl, or ethynyl.
  5. 5. The compound for use according to any one of claims 1-4 wherein R2 is ORa.
  6. 6. The compound for use according to any one of claims 1-5 wherein R is OH, -OC(=O)R1[, or-OC(=O)ORn.
  7. 7. The compound for use according to any one of claims 1 -6 wherein R8 is NRlIR12or ORl[.
  8. 8. The compound for use according to any one of claims 1-7 wherein R8 is NH2.
  9. 9. The compound for use according to any one of claims 1 -7 wherein R8 is OH.
  10. 10. The compound for use according to any one of claims 1-9 wherein R9 is H.
  11. 11. The compound for use according to any one of claims 1-9 wherein R9 is NH2.
  12. 12. The compound for use according to any one of claims 1-11 wherein R7 is H.
  13. 13. The compound for use according to any one of claims 1-3 wherein the compound of Formula I -III is selected from the group consisting of:
    143
    HO F OH ? , ΐ 5 »
    144
  14. 14. The compound for use according to any one of daims 1 -13 further comprising a pharmaceutically acceptable carrier or excipient.
  15. 15. The compound for use according to anyone of daims 1-14 further
    10 comprising administering a therapeutically effective amount of at least one other thereapeutic agent or composition thereof selected from the group consisting of a cortîcosteroid, an anti-inflammatory signal transduction modulator, a β2adrenoreceptor agonist bronchodilator, an anticholinergic, a mucolytîc agent, hypertonie saline and other drugs for treating Paramyxoviridae virus infections; or 15 mixtures thereof.
    145
  16. 16. The compound for use according to claim 15 wherein the at least one other thereapeutic agent is ribavirin, palivizumab, motavizumab, RSV-IGIV (RespiGam®), MEDI-557, A-60444, MDT-637 or BMS-433771 or mixtures thereof.
  17. 17. The compound for use according to any one of daims 1-16 wherein at least one therapeutic agent or mixtures thereof is administered by inhalation.
  18. 18. The compound for use according to claim 17 wherein at least one therapeutic agent or mixtures thereof is administered by nebulization.
  19. 19. The compound for use according to any oneof daims 1-18 wherein the Paramyxoviridae infection is caused by a Paramyxovirina virus.
  20. 20. The compound for use according to any one of daims 1-18 wherein the Paramyxoviridae infection is caused by a Respirovirus virus.
  21. 21. The compound for use according to any one of daims 1-18 wherein the Paramyxoviridae infection îs caused by a type 1 or 3 Human paraînfluenza virus.
  22. 22. The compound for use according to any one of daims 1-18 wherein the Paramyxoviridae infection is caused by a Pneumovirinae virus.
  23. 23. The compound for use according to any oneof daims 1-18 or 22 wherein the Paramyxoviridae infection is caused by a Human respiratory syncytial virus.
  24. 24. The compound for use according to any one of daims 1-23 wherein a Paramyxoviridae polymerase is înhîbited.
  25. 25. A compound of Formula I that is
    146
    7 >
    or a pharmaceutically acceptable sait or ester thereof.
  26. 26.
    A compound of Formula I represented by Formula IV:
    147
    Formula IV or a pharmaceutically acceptable sait or ester, thereof;
    wherein:
    5 each Rl is H or halogen;
    each R3 or Rs is independently H, ORa, N(Ra)2, N3, CN, NO2, S(O)nRa, halogen, (Ci-Cs)alkyl, (C^Cgjcarbocyclylalkyl, (Cj-Cgjsubstituted alkyl, (C2-Cs)alkenyl, (C2 Q)substitutcd alkenyl, (C2-Cg)alkynyl or (C2-Cg)substituted alkynyl;
    R6 is ORa, N(Ra)2, N3, CN, S(O)nRa, -C(=O)R11, -C(=O)ORn, -C(=O)NR11R12, 10 C(=O)SRn, -S(O)Rh, -SiOhR11, -S(O)(ORn), -S(O)2(ORh), -SO2NR11R12, halogen, (Ci-Cgjalkyl, (C4-Cg)carbocyclylalkyl, (C|-Cs)substituted alkyl, (C2-Cs)alkenyl, (C2-C8)substituted alkenyl, (C2-Cg)alkynyl, or (C2-C8)substituted alkynyl;
    each n is independently 0, 1, or 2;
    each Ra is independently H, (Cj-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl,
    15 aryl(Ci-C8)alkyl, (C4-Cs)carbocyclylalkyl, -C(=O)RH, -C(=O)OR, -C(=O)NRllR12, C(=O)SRl], -S(O)RU, -S(O)2Rh, -S(O)(ORn), -S(O)2(ORh), or SO.NRi:r'?;
    R7 is H, -C(=O)R! ', -C(=O)ORn, -C(=O)NR] ‘R12, -C(=O)SR] ‘, -S(O)Rn, S(O)2R11, -S(OXOR1’), -SfOhfOR11), SO2NRR12, or y
    II w2
    148 each Y or Y! is, independently, O, S, NR, +N(O)(R), N(OR), *N(O)(OR), or
    N-NR2;
    Wl and W2, when taken together, are Y’(C(Ry)2)',Y3-; or one of W1 or W2 together with either R3 or R4 is -Y3- and the other of W1 or W2 is Formula la; or W1 and W2 are each, independently, a group of the Formula la:
    wherein:
    each Y2 is independently a bond, O, CR2, NR, +N(O)(R), N(OR), *N(O)(OR),
    N-NR2, S, S-S, S(O), or S(O)2;
    each Y3 is independently O, S, or NR;
    M2 is 0, 1 or 2;
    each R* îs independently Ry or the formula:
    each Μ1 a, Μ1 c, and Midis independently 0 or 1 ;
    M12cis0, 1,2,3,4, 5,6, 7, 8,9, 10, 11 or 12;
    each Ry is independently H, F, Cl, Br, I, OH, R, -C(=Yl)R, -C(=Y‘)OR, C(=YI)N(R)2, -N(R)2, -+N(R)3, -SR, -S(O)R, -S(O)2R, -S(O)(0R), -S(O)2(OR), OC(=Yl)R, -OC(=Y*)OR, -OC(=Y'XN(R)2), -SCi Y'lR, -SC(=Y])OR, SC(=Y1)(N(R)2), -N(R)C(=Y1)R,-N(R)C(=Y')OR, -N(R)C(=Y1)N(R)2, -so2nr2,
    149
    -CN, -N3, -NO2, -OR, or W3; or when taken together, two Ry on the same carbon atom form a carbocyclic ring of 3 to 7 carbon atoms;
    each R is independently H, (Ci-C8) alkyl, (Ci-C8) substituted alkyl, (C2C8)alkenyl, (C2-C8) substituted alkenyl, (C2-C8) alkynyl, (C2-C8) substituted alkynyl, Cô~C2o aryl, Cô-C2o substituted aryl, C2-C20 heterocyclyl, C2-C2o substituted heterocyclyl, arylalkyl or substituted arylalkyl;
    W3 is W4 or W5; W4 is R, -C(Y')Ry, -C(Yl)W5, -SO2Ry, or -SO2W5; and Ws is a carbocycle or a heterocycle wherein W5 is independently substituted with 0 to 3 Ry groups;
    each R8 is halogen, NRnR12, NiR^OR11, NRNRr‘\ N3j NO, NO2, CHO, CN, -CHf-NR1 '), -CH=NNHR* ', -CH=N(OR‘ ’), -CH(OR] ’)2, -C( O)NR! 'R12, -C(=S)NR11R12, -C(=O)ORh, (Ci-Ce)alkyl, (C2-Cs)alkenyl, (C2-C8)alkynyl, (C4-C8)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, C(=O)(C]-C8)alkyl, -S(O)„(Ci-C8)alkyl, aryl(C1-C8)alkyl, OR11 or SR;
    each R9 is independently H, halogen, NRnR12, NÎR’^OR11, NR11NR11R12, N3, NO, NO2, CHO, CN, -CH(=NR1'), -CH=NHNRn, -CH N(OR), -CH(ORh)2, -C(=O)NRhR12, -C(=S)NRhR12, -C(=O)ORh, R11, OR11 or SR11;
    each R[1 or R12 is independently H, (C[-C8)alkyl, (C2-C8)alkenyl, (C2C8)alkynyl, (C4-C8)carbocyclylalkyl, optionally substituted aryl, optionally substituted heteroaryl, -C(=O)(Ct-C8)alkyl, -S(O)n(Ci-C8)alkyl or aryl(Ci-C8)alkyl; or R11 and R12 taken together with a nitrogen to which they are both attached form a 3 to 7 membered heterocyclic ring wherein any one carbon atom of said heterocyclic ring can optionally be replaced with -O-, -S- or -NRa-; and wherein each (C[-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl or aryl(Ci-C8)alkyl of each R3, R5, R6, R11 or R12 is, independently, optionally substituted with one or more halo, hydroxy, CN, N3, N(Ra)2 or ORa; and wherein one or more of the non-terminal carbon atoms of each said (Ci-C8)alkyl may be optionally replaced with -O-, -S- or NRa-.
    150
  27. 27. The compound of claim 26 wherein each R1, R5, and R7 is H and R3 is
    0Ra.
  28. 28. The compound of claim 26 or 27 wherein R6 is is CN, methyl, ethenyl,
    5 or ethynyl.
  29. 29. The compound of any one of claims 26-28 wherein R8 is NH2 and R9 is
    H.
  30. 30. The compound of claim 26 that is pharmaceutically acceptable sait or ester thereof.
  31. 31. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 25-30 and a pharmaceutically acceptable carrier.
    151
  32. 32. The composition of claim 31 further comprising administering a therapeutically effective amount of at least one other thereapeutic agent or composition thereof selected from the group consisting of a cortîcosteroid, an anti-inflammatory signal transduction modulator, a p2-adrenoreceptor agonist bronchodîlator, an
    5 anticholinergic, a mucolytic agent, hypertonie saline and other drugs for treating Paramyxoviridae virus infections; or mixtures thereof.
  33. 33. The composition of claim 32 wherein the at least one other thereapeutic agent is ribavirin, palivizumab, motavizumab, RSV-IGIV (RespiGam®), MEDI-557, A10 60444, MDT-637 or BMS-433771 or mixtures thereof.
OA1201300013 2010-07-22 2011-07-22 Methods and compounds for treating paramyxoviridae virus infections. OA16300A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/366609 2010-07-22

Publications (1)

Publication Number Publication Date
OA16300A true OA16300A (en) 2015-04-24

Family

ID=

Similar Documents

Publication Publication Date Title
US11492353B2 (en) Methods and compounds for treating Paramyxoviridae virus infections
CA2998189C (en) Methods for treating arenaviridae and coronaviridae virus infections
EP2794611B1 (en) Pyrazolo[1,5-a]pyrimidines as antiviral agents
AU2011302310A1 (en) 2&#39; -fluoro substituted carba-nucleoside analogs for antiviral treatment
KR102068856B1 (en) 2&#39;- substituted carba-nucleoside analogs for antiviral treatment
OA16300A (en) Methods and compounds for treating paramyxoviridae virus infections.