top liss

Upload: valdisrein

Post on 04-Jun-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Top Liss

    1/8

    Synthesis ofN-(Chlorophenyl)-2-hydroxynicotinanilides as

    Potential Anti-inflammatory Agents

    Chien-Ming Huanga,b

    ( ), Yong-Hong Hsieha

    ( ),

    Wen-Hsin Huanga* ( ) and An-Rong Lee

    a* ( )

    aSchool of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan, R.O.C.

    bDivision of Pharmacy, Cheng-Hsin Rehabilitation Medical Center, Pei-Tou, Taipei 112, Taiwan, R.O.C.

    (Received April 10, 2007; Accepted April 16, 2007)

    ABSTRACT

    Leflunomide 1 and its non-enzymically active metabolite, malononitrilamide (MNA, 2) are

    clinical use for treating rheumatoid arthritis (RA). Study indicated that the active pharmaco-

    phore, a-keto amide with the enolic hydroxyl group, was fully responsible for the immunosup-

    pressive effects of malononitrilamide 2 leading a salicylamide derivative 3 developed. Previously,

    we have conducted isosterically structural modification mainly based on salicylamide derivative 3,

    which-hydroxy-enamide-containing portion remains untouched, to successfully synthesize a se-

    ries ofN-(4-substituted phenyl)-2-hydroxynicotinanilides. After pharmacological screenings,

    compounds bearing electron-withdrawing groups such as 4-Cl, 4-Br, and 4-NO2 significantly

    showed potent anti-inflammatory activity.1 Currently, a series of compounds 5-14, which are the

    chloro variants on phenyl moiety, mainly based on N-(substituted phenyl)-2-hydroxynicotin-

    anilide 4-14 were further synthesized in high yields. After systematic pharmacological screenings

    of compounds 4-14 and controls, compound 14 exhibited potent in-vivoanti-inflammatory activity

    comparable to that of compound 5 and leflunomide 1, whereas the other compounds have compa-

    rable activity to compound 4 and malononitrilamide 2 by both in vitro suppressing nitric oxide

    (NO) production under the LPS-elicited macrophage Raw 264.7 cell and thein-vivocarrageenan-

    induced paw edema assay, except for compounds 8 and 11.

    Key words: Nicotinailides; Anti-inflammation; Leflunomide.

    INTRODUCTION

    Rheumatoid Arthritis (RA) is primarily one in-

    flammatory disease of the most common diseases of

    the elderly that causes pain, swelling, stiffness, and

    loss of function in the debilitating joints often com-

    plicated with a wide range of other symptoms such

    as feeling tired, running a fever, or generally not

    feeling well.2

    RA treatments can help relieve pa-

    tients pain, reduce swelling, slow down or stop joint

    damage, increase ability to function, and improve

    sense of well being. In addition to treatment with

    conventional non-steroidal anti-inflammatory drugs

    (NSAIDs), steroids, substantial advances in RA pa-

    tients may include antirheumatic drugs, called dis-

    ease-modifying antirheumatic drugs (DMARDs),

    Taiwan Pharmaceutical Journal,2007,59, 39-46 39

  • 8/13/2019 Top Liss

    2/8

    such as cyclosporine and methotrexate, which can

    slow damage of the diseases. However, the signifi-

    cant adverse effects of DMARDs have made themintolerable in practical use, resulting in frequent with-

    drawal.3,4

    The emergence of the de novo pyrimidine-nu-

    cleotide biosynthetic pathway for lymphocyte acti-

    vation and proliferation was thus recognized and

    confirmed by the elucidation of the mechanism of

    action of leflunomide 1,5 an immunosuppressive

    agent, and which being non-enzymically converts to

    a bioactive metabolite malononitrilamide (MNA) 2,

    a-cyano--hydroxy-enamide, involves the inhibi-

    tion of the de novo pyrimidine pathway acting at

    dihydroorotate dehydrogenase (DHODH).

    A study5 indicated that a -cyano--hydroxy-

    enamide2, a-keto amide with the enolic hydroxyl

    group fixed in a cis-configuration to the amide moi-

    ety, is the active pharmacophore of leflunomide me-

    tabolite fully responsible for the immunosuppres-

    sive effects. Bertolini et al. proposed that aZ-form of

    metabolite2, which exhibited an intra-molecularH-bonding stabilization based on molecular model-

    ing techniques, has -cyano--hydroxy-enamide

    moiety as novel rational pharmacophore of the ac-

    tive metabolite of leflunomide.6

    A lead compound3,7 a salicylamide derivative

    which has the minimum structural and physico-

    chemical feature, -hydroxy-enamide moiety, wassynthesized and exhibited immunosuppressive ac-

    tivity superior to that of leflunomide.

    In previous, we have conducted isosterically

    structural modification mainly based on salicyl-

    amide derivative 3, containing a -hydroxy-enamide

    portion remains untouched, to successfully synthe-

    size a series ofN-(4-substituted phenyl)-2-hydroxy-

    nicotinanilides. Among them,N-phenyl-2-hydroxy-

    nicotinanilide4 andN-(4-substituted phenyl)-2-hy-

    droxynicotinanilides bearing electron-withdrawing

    groups such as 4-Cl (5), 4-Br, and 4-NO2 signifi-

    cantly showed potent anti-inflammatory activity,1

    which follow Topliss rule for lead optimization.8,9

    In this study, we further mainly focused on the

    structural extensive modifications of chloro sub-

    stituents on phenyl moiety ofN-phenyl-2-hydroxy-

    nicotinanilide4 in order to search potent immuno-

    modulating agents with fewer and lower adverse ef-

    fects for potently ameliorating rheumatoid arthritisand other autoimmune diseases. A series of title

    compounds, mono-, di- and tri-chloro substituents of

    N-phenyl-2-hydroxynicotinanilides5-14, were eval-

    uated anti-inflammatory activity by both in vitro

    suppressing nitric oxide (NO) production under the

    LPS-elicited macrophage Raw 264.7 cell and the

    in-vivocarrageenan-induced paw edema assay.

    RESULTS AND DISCUSSION

    Chemistry

    The N-(substituted phenyl)-2-hydroxynicotin-

    anilides 4-14were prepared using 2-hydroxynico-

    tinic acid as starting material treated with thionyl

    chloride in the presence of dichloromethane for re-

    fluxing over 3 h. The residue was then reacted with

    the corresponding anilines in high yields (almost

    80%) and recrystallization from butanol afforded the

    40 Taiwan Pharm. J. Vol. 59, No. 1, 2007 Huanget al.

    Fig. 1. Structure of leflunomide 1, malononitril-

    amide 2, salicylamide derivative 3, 2-hy-

    droxy-N-phenylnicotinamide 4, and 2-hy-

    droxy-N-(4-chlorophenyl)-nicotinamide5.

  • 8/13/2019 Top Liss

    3/8

    desired title compounds 4-14. The active intermedi-ate, 2-hydroxynicotinoyl chloride, was successfully

    prepared by thermodynamically controlling the re-

    action in the presence of dichloromethane media in-

    stead of dioxane due to a dominant side product, 2-

    chloronicotinyl chloride, formed as if the reaction

    over 70 C.

    Conformation of compounds 4-14 similar as

    -cyano--hydroxy-enamide 2 could exhibit in a

    manner of solvent-dependence, the (Z)-isomer isdominant due to marked stabilization by forming a

    strong intra-molecular hydrogen bonding, which

    was considered crucial in the contribution of biolog-

    ical activity.6,7

    Biological Evaluations

    In a previous report,1 a series ofN-(4-substi-

    tuted phenyl)-2-hydroxynicotinanilides, isosterically

    structural modification mainly based on salicyl-

    amide derivative 3, containing a -hydroxy-enamide

    portion remains untouched, were successfully syn-

    thesized by introduction of electron-withdrawing

    substituents such as CF3, OCF3, F, Cl, Br, I, CN, and

    NO2at the para postion in the phenyl moiety, which

    show compounds bearing electron-withdrawing groups

    such as 4-Cl, 4-Br, and 4-NO2significantly showed

    potent anti-i nflammatory act ivity, and follow the

    Topliss rule for lead optimization. The Topliss tree

    for aromatic substitution assumes that the lead com-pound has been synthesized and cont ains a mono-

    substituted aromatic ring. The analog that should be

    synthesized after unsubstituted derivative is the 4-

    chloro compound. The chloro substituent is more

    electron-withdrawing and hydrophobic than the H

    atom and therefore the and values are positive as

    Hammetts quantitative correlation.

    Moreover, a series of title compounds, N-(sub-

    stituted phenyl)-2-hydroxynicotinanilides5-14bear-ing mono- di- and tri-chloro substituents on phenyl

    moiety, were synthesized and evaluated anti-inflam-

    matory activity by both in vitro suppressing nitric

    oxide (NO) production under the LPS-elicited macro-

    phage Raw 264.7 cell and the in-vivo carrageenan-

    induced paw edema assay to investigate their struc-

    ture-activity relationships. The anti-inflammatory

    results, expressed as IC50 (M) for suppressing NO

    production level to macrophages and percent inhibi-

    tion of the response to carrageenan-induced inflam-

    mation, are presented in Table 1.

    Examination of these data indicated that activ-

    ity is significant overall. None of compounds exhib-

    ited more potent inhibition of NO production in LPS-

    elicited macrophages than compound5 (4-Cl, IC50

    2.38 0.20M) and compound4 (unsubstituent of

    H at phenyl moiety, IC50 9.98 1.25M). The na-

    ture of the substituents at the phenyl moiety of the ti-

    Novel 2-Hydroxynicotinanilide Derivatives Taiwan Pharm. J. Vol. 59, No. 1, 2007 41

    Scheme 1

    Reagents and condition: (a) SOCl2/CH2Cl2, reflux 3 h; (b) respective aniline/dioxane, reflux 3 h.

  • 8/13/2019 Top Liss

    4/8

    tle compounds does not seem to increase the activity

    noticeably even those compounds are made by fol-

    lowing the Topliss principle.

    From thein-vivocarrageenan-induced paw ede-

    ma study (Table 1), compounds 8 (2,3-(Cl)2 substi-

    tuents) and 11 (2,6-(Cl)2 substituents) showed the

    least anti-inflammatory activity, but compound 14

    (2,4,6-(Cl)3 substituents) has a comparable activity

    (89%, at 3 h, and 67%, at 5 h) to that of compound 5

    (4-Cl substituent, 91%, at 3 h, and 65%, at 5 h) and

    of leflunomide1(85%, at 3 h, and 57%, at 5 h), at an

    intraperitoneal 20 mg/kg dose. The other compounds

    have comparablein-vivoanti-inflammatory activity

    to compounds 4 (unsubstituent of H, 66%, at 3 h, and

    45%, at 5 h) and MNA 2 (73%, at 3 h, and 63%, at 5

    h), at an intraperitoneal 20 mg/kg dose.

    Most of compounds significantly show consis-

    tent anti-inflammatory activity betweenin-vitrosup-

    pressing nitric oxide (NO) production under the LPS-

    elicited macrophage Raw 264.7 cell and the in-vivo

    carrageenan-induced paw edema assay, whereas

    compound14(2,4,6-(Cl)3substituents) was extraor-

    dinary active in the screening model against carrage-

    enan-induced inflammation but exhibited less active

    inhibition of NO production in LPS-elicited macro-

    phages. This ambiguous result might be compound

    14 displayed a water-insoluble in in-vitro cellular

    study.

    CONCLUSION

    We have conducted the extensive chloro sub-

    stituents of structural modification mainly on the

    phenyl ring derived from isosterically heteroatomic

    display of a salicylamide derivative3, a lead of anti-

    rheumatic drugs, while its -hydroxy-enamide-con-

    taining portion remains untouched, to successfully

    synthesize 2-hydroxynicotinamides 4-14. By virtue

    of their potent anti-inflammatory activity, our syn-

    thetic compounds might have potential to be em-

    ployed as nove l anti-rheumatic anti-inflammatory

    agents. Finally, the information gleaned from this

    42 Taiwan Pharm. J. Vol. 59, No. 1, 2007 Huanget al.

    Table 1. Anti-inflammatory effects of 2-hydroxy-N-(substituted phenyl)-nicotinamides 4-14 and controls

    Paw edema (mL) after carrageenan challengea

    (percentage inhibition, %)*Experimental candidate R

    3 h 5 h

    NO

    (M)

    Control 1.93 0.23 2.32 0.19

    Leflunomide (1) 0.29 0.10 (85) 1.00 0.15 (57) 129.90 2.21

    MNA (2) 0.52 0.08 (73) 0.86 0.12 (63) 157.60 6.11

    4 H 0.66 0.11 (66) 1.28 0.17 (45) 9.98 1.25

    5 4-Cl 0.17 0.06 (91) 0.81 0.11 (65) 2.38 0.20

    6 2-Cl 1.25 0.17 (35) 1.41 0.21 (39) 155.34 1.91

    7 3-Cl 0.45 0.13 (77) 0.83 0.15 (64) 54.36 2.07

    8 2,3-(Cl)2 1.89 0.25 (2)#

    2.08 0.19 (10)#

    222.09 1.91

    9 2,4-(Cl)2 0.52 0.13 (73) 0.88 0.18 (62) 60.01 2.48

    10 2,5-(Cl)2 0.89 0.11 (54) 1.03 0.12 (56) 86.68 2.98

    11 2,6-(Cl)2 1.97 0.31 (-2)

    #

    2.13 0.25 (8)

    #

    314.62 3.5912 3,4-(Cl)2 0.95 0.14 (51) 1.29 0.11 (44) 94.23 1.56

    13 3,5-(Cl)2 0.70 0.13 (64) 0.96 0.19 (59) 74.73 2.16

    14 2,4,6-(Cl)3 0.22 0.16 (89) 0.76 0.13 (67) 261.41 3.45

    aEach value represents the mean S.E.M. of 5 animals.

    Statistically significant different from control as *p< 0.05; statistically significant different from positive controls

    (leflunomide and MNA) as#p< 0.05.

  • 8/13/2019 Top Liss

    5/8

    study could suggest ways to develop new drugs use-

    ful in the treatment of inflammatory or immunologi-

    cal disorders such as RA.

    MATERIALS AND METHODS

    Chemistry

    Melting points were taken in open capillary

    tubes on a Buchi-530 melting point apparatus and

    are uncorrected. UV-vis spectra were recorded on a

    Shimazu UV-160A UV-Visble recording spectro-

    photometer. IR spectra were recorded on a Perkin-

    Elmer FTIR 1610 series infrared spectrophotometer

    in KBr discs. 1

    H- and 13

    C-NMR spectra were deter-

    mined on a Varian Gemini-300 NMR instrument.

    Chemical shifts () were reported in parts per mil-

    lion (ppm) relative to tetramethylsilane (TMS) as an

    internal standard, and coupling constants (J) were

    given in hertz (Hz). Fast atom bombardment (FAB)

    mass spectra were recorded using a Finnigan MAT

    95S (GC/MS) mass spectrometer. All reactions were

    routinely monitored by TLC on Merck F254 silicagel plates. Merck silica gel (70-230 mesh) was used

    for chromatography. Elemental analyses for carbon,

    hydrogen and nitrogen were performed in the Instru-

    ment Center of the National Science Counsel at the

    Nat ional Taiwan Uni versit y usi ng Perkin-Elmer

    CHN-2400. All the solvents and reagents were ob-

    tained from commercial sources and purified before

    use if necessary.

    General synthetic procedure of N-(substi-

    tuted phenyl)-2-hydroxynicotinanilides 4-14

    2-Hydroxynicotinic acid (1.27 g, 10 mmol) was

    warmed with thionyl chloride (2 mL) in dichloro-

    methane (60 mL) at 50 C for 3 h. The excess thionyl

    chloride was removedin vacuo. The residue was di-

    rectly reacted with substituted anilines (12 mmol) in

    dioxane (30 mL) for 3 h. The reaction mixture was

    purified from chromatography (hexane/e thyl ace-

    tate). Recrystallization of desired products from

    butanol afforded the title compounds 4-14.

    2-Hydroxy-N-phenylnicotinamide (4) and 2-Hydroxy-N-(4-chlorophenyl)nicotinamide (5) were

    synthesized according to our previous published.1

    2-Hydroxy-N-(2-chlorophenyl)nicotinamide

    (6)

    2.04 g (82% yield); TLC Rf= 0.42 (hexane:ethyl

    acetate = 1:4); mp 224-226 C. UV max (DMSO)

    nm (log ): 340 (4.11). IR (KBr) cm-1

    : 3321 (OH),

    2940 (CH), 1683 (C=O), 1595 (C=C). EI-MS m /z

    (%): 248 (M+, 20), 122 (100). 1H-NMR (DMSO):

    6.58 (1H, t,J= 6.6 Hz, pyridyl H-5), 7.12 (1H, t,J=

    7.9 Hz, phenyl H-4), 7.35 (1H, t,J= 7.9 Hz, phenyl

    H-5), 7.52 (1H, d,J= 7.9 Hz, phenyl H-6), 7.84 (1H,

    dd, J= 6.6, 2.1 Hz, pyridyl H-4), 8.49 (1H, dd,J=

    6.6, 2.1 Hz, pyridyl H-6), 8.55 (1H, d, J= 7.9 Hz,

    phenyl H-3), 12.56 (1H, s, NH), 12.73 (1H, s, OH).13

    C-NMR (DMSO) : 108.2, 121.2, 123.3, 123.9,

    126.0, 129.0, 130.7, 136.9, 141.9, 146.4, 163.3,

    163.8. Anal. Calcd. for C12H9ClN2O2: C, 57.96; H,3.65; N, 11.27. Found: C, 58.06; H, 3.54; N, 11.33.

    2-Hydroxy-N-(3-chlorophenyl)nicotinamide

    (7)

    2.08 g (83% yield); TLC Rf= 0.36 (hexane:ethyl

    acetate = 1:4); mp 239-241 C. UV max (DMSO)

    nm (log): 338 (4.13). IR (KBr) cm-1: 3265 (OH),

    2929 (CH), 1690 (C=O), 1585 (C=C). EI-MS m /z

    (%): 248 (M+, 40), 122 (100). 1H-NMR (DMSO):

    6.57 (1H, t,J= 7.1 Hz, pyridyl H-5), 7.16 (1H, t,J=

    8.1 Hz, phenyl H-5), 7.37 (1H, d, J= 8.1 Hz, phenyl

    H-4), 7.44 (1H, d,J= 8.1 Hz, phenyl H-6), 7.82 (1H,

    dd,J= 7.1, 2.1 Hz, pyridyl H-4), 7.98 (1H, s, phenyl

    H-2), 8.45 (1H, dd, J= 7.1, 2.1 Hz, pyridyl H-6),

    12.32 (1H, s, NH), 12.79 (1H, s, OH). 13C-NMR

    (DMSO) : 108.3, 119.6, 120.6, 121.1, 124.8, 131.9,

    134.7, 141.2, 141.8, 146.2, 163.3, 163.9. Anal.

    Calcd. for C12H9ClN 2O2: C, 57.96; H, 3.65; N,

    Novel 2-Hydroxynicotinanilide Derivatives Taiwan Pharm. J. Vol. 59, No. 1, 2007 43

  • 8/13/2019 Top Liss

    6/8

    11.27. Found: C, 57.80; H, 3.76; N, 11.12.

    2-Hydroxy-N-(2,3-dichlorophenyl)nicotin-amide (8)

    2.38 g (84% yield); TLC Rf= 0.42 (hexane:ethyl

    acetate = 1:4); mp 339-340 C. UV max (DMSO)

    nm (log ): 341 (4.08). IR (KBr) cm-1: 3205 (OH),

    2865 (CH), 1697 (C=O), 1588 (C=C). EI-MS m /z

    (%): 283 (M+

    , 40), 122 (100). 1

    H-NMR (DMSO):

    6.58 (1H, t,J= 5.8 Hz, pyridyl H-5), 7.36~7.38 (2H,

    m, phenyl H-4, 6), 7.84 (1H, d, J= 5.8 Hz, pyridyl

    H-4), 8.47~8.55 (2H, m, phenyl H-5, pyridyl H-6),

    12.31 (1H, s, NH), 12.75 (1H, s, OH). 13C-NMR

    (DMSO) : 108.4, 120.9, 121.6, 126.3, 129.7, 131.9,

    133.2, 138.9, 142.1, 146.6, 163.5, 163.8.Anal. Calcd.

    for C12H8Cl2N2O2: C, 50.91; H, 2.85; N, 9.89. Found:

    C, 50.96; H, 3.02; N, 9.79.

    2-Hydroxy-N-(2,4-dichlorophenyl)nicotin-

    amide (9)

    2.29 g (81% yield); TLC Rf= 0.39 (hexane:ethyl

    acetate = 1:4); mp 366-368 C. UV max (DMSO)nm (log ): 340 (4.15). IR (KBr) cm

    -1: 3201 (OH),

    2918 (CH), 1683 (C=O), 1584 (C=C). EI-MS m /z

    (%): 283 (M+, 35), 122 (100). 1H-NMR (DMSO):

    6.58 (1H, t,J= 7.0 Hz, pyridyl H-5), 7.47~7.59 (2H,

    m, phenyl H-5, 6), 7.84 (1H, d, J= 7.0 Hz, pyridyl

    H-4), 8.46 (1H, d,J= 7.0 Hz, pyridyl H-6), 8.57 (1H,

    s, phenyl H-3), 12.14 (1H, s, NH), 12.78 (1H, s, OH).13

    C-NMR (DMSO) : 108.4, 120.8, 121.9, 122.6,

    128.9, 132.1, 133.3, 138.7, 142.5, 146.4, 163.7, 163.9.

    Anal. Calcd. for C12H8Cl2N2O2: C, 50.91; H, 2.85;

    N, 9.89. Found: C, 50.81; H, 2.95; N, 9.86.

    2-Hydroxy-N-(2,5-dichlorophenyl)nicotin-

    amide (10)

    2.25 g (79% yield); TLC Rf= 0.36 (hexane:ethyl

    acetate = 1:4); mp 301-302 C. UV max (DMSO)

    nm (log ): 341 (4.13). IR (KBr) cm-1

    : 3310 (OH),

    2953 (CH), 1697 (C=O), 1590 (C=C). EI-MS m /z

    (%): 283 (M+, 20), 122 (100). 1H-NMR (DMSO):

    6.59 (1H, t,J= 7.1 Hz, pyridyl H-5), 7.18 (1H, dd,J

    = 8.5, 2.0 Hz, phenyl H-4), 7.55 (1H, d, J= 8.5 Hz,phenyl H-3), 7.86 (1H, d, J= 7.1 Hz, pyridyl H-4),

    8.48 (1H, d,J= 7.1 Hz, pyridyl H-6), 8.67 (1H, d,J=

    2.0 Hz phenyl H-6), 12.18 (1H, s, NH), 12.76 (1H, s,

    OH). 13C-NMR (DMSO) : 108.3, 120.7, 122.3,

    122.4, 125.5, 131.9, 133.3, 138.2, 142.2, 146.6, 163.7,

    163.8.Anal. Calcd. for C12H8Cl2N2O2: C, 50.91; H,

    2.85; N, 9.89. Found: C, 50.87; H, 2.95; N, 9.78.

    2-Hydroxy-N-(2,6-dichlorophenyl)nicotin-

    amide (11)

    2.41 g (85% yield); TLC Rf= 0.34 (hexane:ethyl

    acetate = 1:4); mp 269-270 C. UV max (DMSO)

    nm (log): 333 (3.99), 294 (3.26), 262 (3.56). IR

    (KBr) cm-1

    : 3336 (OH), 2988 (CH), 1676 (C=O),

    1615 (C=C). EI-MS m/z (%): 283 (M+

    , 80), 122

    (100). 1H-NMR (DMSO): 6.56 (1H, t,J= 7.0 Hz,

    pyridyl H-5), 7.33 (1H, t, J= 8.1 Hz, phenyl H-4),

    7.53~7.55 (2H, m, phenyl H-3, 5), 7.84 (1H, dd, J=

    7.0, 2.0 Hz, pyridyl H-4), 8.43 (1H, dd, J= 7.0, 2.0Hz, pyridyl H-6), 11.73 (1H, s, NH), 12.79 (1H, s,

    OH). 13

    C-NMR (DMSO) : 108.1, 120.8, 129.8,

    130.2, 134.2, 134.2, 141.9, 146.4, 163.0, 164.0.Anal.

    Calcd. for C12H8Cl2N2O2: C, 50.91; H, 2.85; N, 9.89.

    Found: C, 50.84; H, 2.93; N, 9.88.

    2-Hydroxy-N-(3,4-dichlorophenyl)nicotin-

    amide (12)

    2.31 g (82% yield); TLC Rf

    = 0.27 (hexane:ethyl

    acetate = 1:4); mp 295-296 C. UV max (DMSO)

    nm (log): 340 (4.22). IR (KBr) cm-1

    : 3302 (OH),

    2943 (CH), 1682 (C=O), 1591 (C=C). EI-MS m /z

    (%): 283 (M+, 25), 122 (100). 1H-NMR (DMSO):

    6.58 (1H, t,J= 7.0 Hz, pyridyl H-5), 7.49~7.59 (2H,

    m, phenyl H-5, 6), 7.82 (1H, dd, J= 7.0, 2.0 Hz,

    pyridyl H-4), 8.14 (1H, s, phenyl H-2), 8.44 (1H, dd,

    J= 7.0, 2.0 Hz, pyridyl H-6), 12.36 (1H, s, NH),

    12.82 (1H, s, OH). 13

    C-NMR (DMSO) : 108.3,

    44 Taiwan Pharm. J. Vol. 59, No. 1, 2007 Huanget al.

  • 8/13/2019 Top Liss

    7/8

    120.9, 121.3, 122.3, 126.6, 132.2, 132.6, 139.8, 141.9,

    146.2, 163.4, 163.9.Anal. Calcd. for C12H8Cl2N2O2:

    C, 50.91; H, 2.85; N, 9.89. Found: C, 50.89; H, 3.01;N, 9.77.

    2-Hydroxy-N-(3,5-dichlorophenyl)nicotin-

    amide (13)

    2.46 g (87% yield); TLC Rf= 0.36 (hexane:ethyl

    acetate = 1:4); mp 286-287 C. UV max (DMSO)

    nm (log ): 339 (4.21). IR (KBr) cm-1: 3423 (OH),

    3073 (CH), 1681 (C=O), 1594 (C=C). EI-MS m /z

    (%): 283 (M+

    , 30), 122 (100). 1

    H-NMR (DMSO):

    6.58 (1H, t, J= 6.8 Hz, pyridyl H-5), 7.30 (1H, s,

    phenyl H-4), 7.77~7.83 (2H, m, phenyl H-2, 6), 7.84

    (1H, dd,J= 6.8, 2.1 Hz, pyridyl H-4), 8.44 (1H, dd,J

    = 6.8, 2.1 Hz, pyridyl H-6), 12.40 (1H, s, NH), 12.82

    (1H, s, OH). 13

    C-NMR (DMSO) : 108.4, 119.4,

    120.8, 124.3, 135.6, 142.1, 146.4, 163.6, 163.9.Anal.

    Calcd. for C12H8Cl2N2O2: C, 50.91; H, 2.85; N, 9.89.

    Found: C, 50.87; H, 2.94; N, 9.79.

    2-Hydroxy-N-(2,4,6-trichlorophenyl)nicotin-amide (14)

    2.38 g (75% yield); TLC Rf= 0.31 (hexane:ethyl

    acetate = 1:2); mp 271-272 C. UV max (DMSO)

    nm (log ): 334 (4.01), 294 (3.35), 265 (3.64). IR

    (KBr) cm-1

    : 3416 (OH), 3075 (CH), 1698 (C=O),

    1615 (C=C). EI-MS m/z (%): 317 (M+, 20), 122

    (100). 1H-NMR (DMSO): 6.57 (1H, t,J= 6.9 Hz,

    pyridyl H-5), 7.77 (2H, s, phenyl H-3, 5), 7.85 (1H,

    d,J= 6.9 Hz, pyridyl H-4), 8.43 (1H, d, J= 6.9 Hz,

    pyridyl H-6), 11.73 (1H, s, NH), 12.79 (1H, s, OH).13

    C-NMR (DMSO) : 108.2, 120.5, 129.6, 133.4,

    133.7, 135.1, 142.1, 146.5, 163.1, 163.9.Anal. Calcd.

    for C12H7Cl3N2O2: C, 45.39; H, 2.22; N, 8.82.

    Found: C, 45.36; H, 2.37; N, 8.67.

    Pharmacology10

    Cell culture

    The mouse BALB/c macrophage cell line, RAW

    264.7 (identical to ATCC number TIB-71), was ob-

    tained from Bioresource Collection and Research

    Center, Taiwan. The cells were maintained accord-ing to the reported method, being cultured in 50 cm2

    plastic flasks (Nunc, Roskilde, Denmark) with the

    medium renewed every 3 days.

    Determination of cell viability by an MTT assay

    To evaluate the cell viability, a methylthiazo-

    letetrazolium bromide (MTT) assay was conducted

    by the standard method as described previously. In-

    cubation was performed after pretreating with a

    combination of the polyhydroxyflavonoids, in con-

    centrations of 25, 50, 100, and up to 200 M in

    dimethyl sulfoxide, and LPS (100 ng/mL) in normal

    saline for 24 h. Untreated cells were used as the con-

    trol.

    Nitrite quantification

    The cells were cultured with or without a pre-

    treatment by a polyhydroxyflavonoid of 25, 50, 100,

    and up to 200M as already described. The produc-

    tion of NO was determined by measuring the accu-

    mulated level of nitrite in the culture supernatantwith the Griess reagent in LPS-stimulated macro-

    phage cells. A quantity of 100 l of a sample aliquot

    were mixed with 100l of the Griess reagent (0.1%

    N-(1-naphthyl)ethylenediamine, 1% sulfanilamide,

    and 2.5% phosphoric acid) in a 96-well plate and

    then incubated at 25C for 10 min. The absorbance

    at 550 nm was measured with an ELISA reader (MR

    700, Dynatech Laboratories, Alexandria, VA, USA).

    NaNO2was used as the standard to calculate the ni-

    trite concentration.

    Determination of anti-inflammatory activities by

    the carrageenan-induced hind paw edema test on

    rats

    Male albino Wistar rats (180-215 g) were housed

    and cared for under the guidelines of the Institu-

    tional Animal Care and Use Committee at the Na-

    tional Defense Medical Center, Taiwan. The rats

    were assigned to groups, one of them being the con-

    Novel 2-Hydroxynicotinanilide Derivatives Taiwan Pharm. J. Vol. 59, No. 1, 2007 45

  • 8/13/2019 Top Liss

    8/8

    trol. In order to induce inflammation, 50l of a 1%

    carrageenan solution in normal saline was injected

    into the right hind paw subplantar tissue, accordingto the modified method of Winter et al.11 The devel-

    opment of paw edema was measured plethysmo-

    graphically (Basile 7140 plethysmometer, Ugo,

    Varese, Italy) and recorded prior to this administra-

    tion. One hour before the carrageenan challenge, a

    sample preparation (20 mg/kg) was injected i.p. into

    a rat in a test group. Normal saline was injected in

    the same way into animals in the control group. Af-

    ter the carrageenan challenge, each paw volume

    (mL) was measured hourly up to 5 h. The volumes of

    the injected and of the contralateral paws were mea-

    sured, using a plethysmometer (Ugo Basile, Italy), at

    3 h after test compounds administered (i.e., 4 h after

    induction of inflammation). The percentage of paw

    edema and the inhibition of inflammation were cal-

    culated by the previously reported protocol.

    Statistical analysis

    Each experimental data value is expressed as

    the mean SEM. The statistical significance of dif-ferences was assessed with an analysis of variance

    (ANOVA), followed by Tukeys test or Students

    test between two groups. Differences with p values

    of less than 0.05 are considered statistically signifi-

    cant.

    ACKNOWLEDGEMENTS

    We gratefully acknowledge the research grant

    supported from the Cheng-Hsin Rehabilitation Med-

    ical Center (CHRMC 94-34), the Republic of China.

    REFERENCES

    1. Huang, C. M.; Hsieh, Y. H.; Huang, W. H.; Lee, A.

    R. Synthesis of N-(4-substituted phenyl)-2-hy-

    droxynicotinanilides as potential anti-inflamma-

    tory agents. Chin. Pharm. J.2006,58, 105-113.

    2. Schattenkirchner, M. The use of leflunomide in

    the treatment of rheumatoid arthritis: an experi-

    mental and clinical review. Immunopharmacol-

    ogy 2000,47, 291-298.

    3. van der Heide, A.; Jacobs, J. W.; Bijlsma, J. W. J.

    The effectiveness of early treatment with sec-

    ond-line antirheumatic drugs. A randomized,

    controlled trial. Ann. Intern. Med. 1996, 124,

    699-707.

    4. El Desoky, E. S. Pharmacoatherapy of rheuma-

    toid arthritis: an overview. Curr. Ther. Res.2001,

    62, 92-112.

    5. Silva, H. T.; Morris, R. E. Leflunomide andmalononitrilamides. Am. J. Med. Sci. 1997,313,

    289-301.

    6. Papageorgiou, C.; Zurini, M.; Weber, H.-P.;

    Borere, X. Leflunomides bioactive metabolite

    has the minimal structural requirements for the ef-

    ficient inhibition of human dihydrooratate

    dehydrogenase. Bioorg. Chem. 1997, 25, 233-

    238.

    7. Bertolini, G.; Aquino, M.; Biffi, M.; et al. A new

    rational hypothesis for the pharmacophore of theactive metabolite of leflunomide, a potent immu-

    nosuppressive drug. J. Med. Chem. 1997, 40,

    2011-2016.

    8. Topliss, J. G. Utilization of operational schemes

    for analog synthesis in drug design. J. Med.

    Chem.1972,15, 1006-1011.

    9. Topliss, J. G. A manual method for applying the

    Hansch approach to drug design. J. Med. Chem.

    1977,20, 463-469.

    10. Huang, W. H.; Lee, A. R.; Yang, C. L. Anti-

    oxidative and anti-inflammatory activities of

    polyhydroxyflavonoids ofScutellaria baicalensis

    GEORGI. Biosci. Biotechnol. Biochem.2006,70,

    2371-2380.

    11. Winter, C. A.; Risley, E. A.; Nuss, C. W. Car-

    rageenan-induced edema in hind paw of the rats as

    an assay for anti-inflammatory drugs. Proc. Soc.

    Exp. Biol. Med. 1962,111, 544-547.

    46 Taiwan Pharm. J. Vol. 59, No. 1, 2007 Huanget al.