CN100471858C - Inhibitors of cyclin-dependent kinases, compositions and uses related thereto - Google Patents

Inhibitors of cyclin-dependent kinases, compositions and uses related thereto Download PDF

Info

Publication number
CN100471858C
CN100471858C CNB2004800420356A CN200480042035A CN100471858C CN 100471858 C CN100471858 C CN 100471858C CN B2004800420356 A CNB2004800420356 A CN B2004800420356A CN 200480042035 A CN200480042035 A CN 200480042035A CN 100471858 C CN100471858 C CN 100471858C
Authority
CN
China
Prior art keywords
compound
cell
represent
acid
group
Prior art date
Legal status (The legal status 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 status listed.)
Expired - Fee Related
Application number
CNB2004800420356A
Other languages
Chinese (zh)
Other versions
CN1922180A (en
Inventor
N·博科维奇
A·F·克卢格
C·奥尔曼
K·K·穆尔蒂
S·拉姆
王忠国
黄建星
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agennix USA Inc
Original Assignee
Agennix USA 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 Agennix USA Inc filed Critical Agennix USA Inc
Publication of CN1922180A publication Critical patent/CN1922180A/en
Application granted granted Critical
Publication of CN100471858C publication Critical patent/CN100471858C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention pertains to novel cyclin dependent kinase inhibitors (cdks) of formula (I) wherein the substituents are as defined in the claims and specifically, but not exclusively, as inhibitors of cdk/cyclin complexes. As described herein, the inhibitors of this invention are capable of inhibiting the cell-cycle machinery and consequently may be useful in modulating cell-cycle progression, ultimately controlling cell growth and differentiation. Such compounds would be useful for treating subjects having disorders associated with excessive cell proliferation.

Description

The kinase inhibitor of cyclin dependent and composition thereof and associated uses
I. invention field
The present invention relates to the method for the compound of kinases (cdk) inhibitor as cyclin dependent, the medicinal compositions that contains this compounds, preparation medicinal compositions at large, or with the method for this compounds for treating cancer or proliferative disease or other disease, and the intermediate and the method that are used to prepare this compounds.
II. background of invention
A most important and basic process in the biology is the cell fission by the cell cycle mediation.This process guarantees to control the progeny cell that generation has the particular organisms function.It is the phenomenon of highly regulating and control, to replying with the cell signal generation of external source in the various kinds of cell.Tumour promotes and the complex network of suppressor gene product is the key component of this cell signal process.The overexpression tumour promotes composition or the loss of tumor suppression product subsequently will cause cell proliferation and tumour to produce (Pardee out of control, Science 246:603-608,1989). the kinases of cyclin dependent plays a crucial role in regulating cell cycle machine (machinery).These mixtures are grouped into by following two kinds of one-tenth: catalytic subunit's (kinases) and regulator subunit (cyclin).Up to now, nine kinds of kinase subunit units (the kinases 1-9 of cyclin dependent) and several regulator subunit (cyclin A-H, K, N and T) have been differentiated.Every kind of kinases and specificity are regulated the mating partner gang, form the active catalytic part.Regulate each conversion of cell cycle by the kinase complex of special cyclin dependent: regulate G1/S by the kinases 2/ cyclin E of cyclin dependent, kinases 4/ cyclin D1 of cyclin dependent and the kinases 6/ cyclin D2 of cyclin dependent; Regulate S/G2 by the kinases 2/ cyclin A of cyclin dependent and the kinases/cyclin A of cyclin dependent; G2/M regulates by the kinases/cyclin D of cyclin dependent.These kinase whose synergistic activity guiding individual cells are guaranteed every crowd of offspring's vigor (Sherr, Cell 73:1059-1065,1993 by reproduction process; Draetta, Trends Biochem.Sci.15:378-382,1990).
More and more evidences shows that the relevant dysfunction of the kinases of tumor development and cyclin dependent is relevant.The adjusting albumen of overexpression cyclin relevant with several cancers (Jiang, Proc.Natl.Acad.Sci.USA 90:9026-9030,1993 with kinases hyperaction subsequently; Wang, Nature 343:555-557,1990).The kinase whose endogenous of recent findings cyclin dependent, high degree of specificity protein inhibitor on cell proliferation have material impact (Kamb etc., Science 264; 436440,1994; Beach, Nature 336:701-704,1993).These inhibitor comprise p16INK4 (the kinases 4/D1 inhibitor of cyclin dependent), p21CIP1 (kinase inhibitor of non-specific cyclin dependent) and p27KIP1 (the kinases 2/E inhibitor of specificity cyclin dependent).Nearest crystalline structure with kinases 2/A bonded p27 cyclin dependent shows how these protein repeatedly interact by the kinase complex with cyclin dependent, thereby effectively suppress kinase activity (Pavletich, Nature 382:325-331,1996).These protein carry out specificity by the kinase complex with their corresponding cyclin dependents and interact, and help regulate the cell cycle.The cell that lacks these inhibitor is easily grown out of control and the formation tumour.These evidences cause people actively to seek the micromolecular inhibitor medicine of cdk family.
III. summary of the invention
The present invention describes effective inhibitor compound of the kinases enzyme that is called cyclin dependent.The invention provides at least a The compounds of this invention by treating significant quantity or the method for its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer treatment cancer or other proliferative disease or other disease.The present invention also provides by treating effective at least a The compounds of this invention and another kind of anticancer or the combination medicine treatment cancer of anti-proliferative drugs or the method for other proliferative disease or other disease.
In certain embodiments, the invention provides compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula I structure:
Wherein
R 8Representative replaces or unsubstituted heterocycle or replacement or unsubstituted morpholino, replacement or unsubstituted piperazinyl or replacement or unsubstituted cyclohexyl;
F represents (CH 2) n, wherein n is the integer of 1-6, in some embodiments, n is 1;
The Q representative replaces or the amino substituting group of unsubstituted secondary amino group substituting group, replacement or unsubstituted uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, n is 1.
In some embodiments, the Q among the formula I represents uncle amino substituting group, for example dialkylamine.In some embodiments, the Q among the formula I represents replacement or unsubstituted nitrogen heterocyclic ring for example morpholine, piperidines, piperazine or tetramethyleneimine.In some embodiments, Q represents nitrogenous heteroaryl ring, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, R 8Representative:
Figure C200480042035D00062
Wherein Z is O or NR "; And
R " represent H or low alkyl group.
In some embodiments, the compound with formula I structure does not comprise one or more following compounds:
Figure C200480042035D00071
In some embodiments, formula I compound comprises the compound that one or more are listed in table.For example, formula I compound can comprise one or more compound B-11-B20 and C2.
As mentioned above; in some embodiments, suitable substituents can comprise alkyl, oxo base, acyl amino, hydroxyl, carbonyl, alkylsulfonyl, ester, acid amides, NR at every turn independently when occurring ", hydroxyalkyl, alkoxyalkyl, aryl, heterocyclic radical, cycloalkyl or oligomeric (ethylene glycol).In some embodiments, when Q represented the secondary amino group substituting group, suitable substituents comprised alkyl, alkoxyalkyl, hydroxyalkyl and hydroxy alkoxy alkyl.Those skilled in the art should recognize easily that cited substituting group is not limit, and can use a lot of other suitable substituents.
In certain embodiments, the invention provides compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula II structure:
Wherein
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represents O, S or NR ";
R ' represents H, low alkyl group or metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R 5Represent H, P (=O) (OR ') 2Or M nQ;
R 6Represent H, OH or M nQ, condition is R 5And R 6In the middle of have only one to represent H;
R 7Represent H, halogen, hydroxyl, low alkyl group or lower alkoxy;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=O) and C (=S)), NR ", O, S, S (O) or S (O 2);
In the time of in being present in B, n represents the integer of 1-4, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or is unsubstituted: amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, R 8Representative replaces or unsubstituted morpholino, piperazinyl or cyclohexyl.
In some embodiments, R " represent H.
In some embodiments, R 5Represent M nQ.
In some embodiments, the M that is connected on the Q represents CH 2, S (O 2), C (=S) or C (=O).
In some embodiments, the M that is connected on the Q represents CH 2
In some embodiments, be connected M on the Q represent C (=O).
In some embodiments, be connected the NR that the M representative on the Q replaces ".
In some embodiments, the Q representative replaces or unsubstituted nitrogen heterocyclic ring.
In some embodiments, the Q representative replaces or unsubstituted uncle's amino.
In some embodiments, R 8Representative replaces or unsubstituted morpholino, piperazinyl or cyclohexyl.In some embodiments, R " represent H.And in some embodiments, the M of at least one appearance represents CH 2, the NR that replaces ", perhaps, represent CH when being connected Q when going up 2, S (O 2), C (=S) or C (=O).
In some embodiments, the Q representative replaces or unsubstituted nitrogenous heteroaryl ring.In some embodiments, the Q representative replaces or unsubstituted nitrogen heterocyclic ring.In some embodiments, the Q representative replaces or unsubstituted uncle's amino.In some embodiments, the Q representative replaces or unsubstituted secondary amino group.
In certain embodiments, the present invention relates to have compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer of following structure:
Figure C200480042035D00091
C1。
In certain embodiments, the present invention relates to have compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer of following structure:
Figure C200480042035D00092
C5。
In some embodiments, the invention provides one or more compounds described herein of purifying or synthesized form.
In certain embodiments, the invention provides compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula II structure:
Figure C200480042035D00101
Wherein
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represents O, S, S (O 2), C (=O), C (=S), CH 2Or NR ";
R ' represents H, low alkyl group or metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R " ' represent H or choose the low alkyl group that replaces wantonly;
R 5Represent M nJK;
R 6Represent H, OH or M nQ;
R 7Represent H, halogen, hydroxyl, low alkyl group or lower alkoxy;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', N (R ") 2Or N (R ') SO 2R ";
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=O) and C (=S)), NR ", O, S, S (O) or S (O 2);
In the time of in being present in B, n represents the integer of 1-7, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or is unsubstituted: nitrogenous heteroaryl ring, secondary amino group substituting group, the amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, R 8Representative replaces or unsubstituted morpholino, piperazinyl or cyclohexyl.In some embodiments, R " represent H.
In some embodiments, the M that is connected on the Q represents CH 2, the NR that replaces ", S (O 2), C (=S) or C (=O).
In some embodiments, R 8Representative replaces or unsubstituted morpholino, piperazinyl or cyclohexyl.
In some embodiments, R " represent H.
In some embodiments, R 5Represent M nQ.
In some embodiments, the M that is connected on the Q represents CH 2, S (O 2), C (=S) or C (=O).
In some embodiments, be connected M on the Q be C (=O).
In some embodiments, the M that is connected on the Q represents CH 2
In some embodiments, be connected the NR that the M representative on the Q replaces ".
In some embodiments, the Q representative replaces or the amino substituting group of unsubstituted uncle.
In some embodiments, the Q representative replaces or unsubstituted nitrogen heterocyclic ring.
In some embodiments, substituting group comprises alkyl, oxo base, acyl amino, hydroxyl, carbonyl, alkylsulfonyl, ester, acid amides, NR at every turn independently when occurring ", hydroxyalkyl, alkoxyalkyl, aryl, heterocyclic radical, cycloalkyl or oligomeric (ethylene glycol).
In certain embodiments, the invention provides compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula II structure:
Figure C200480042035D00111
Wherein
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represents O, S, S (O 2), C (=O), C (=S), CH 2Or NR ";
R ' represents H, low alkyl group or metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R " ' represent H or choose the low alkyl group that replaces wantonly;
R 5Represent H, P (=O) (OR ') 2, M nJK or M nQ;
R 6Represent H, OH or M nQ, condition is R 5And R 6In the middle of have only one to represent H;
R 7When occurring, represent H, halogen, hydroxyl, low alkyl group or lower alkoxy independently at every turn;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', N (R ") 2Or N (R ') SO 2R ";
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=O) and C (=S)), NR ", O, S, S (O), S (O 2) or CH 2
In the time of in being present in B, n represents the integer of 1-7, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or is unsubstituted: nitrogenous heteroaryl ring, secondary amino group substituting group, the amino substituting group of uncle or nitrogen heterocyclic ring;
Condition is: do not comprise the compound with formula IIa structure:
Figure C200480042035D00121
Wherein
W and Z represent O or NR independently ";
R ' represents H, low alkyl group or metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R 5Represent H, P (=O) (OR ') 2Or M nQ;
R 6Represent H, OH or M nQ, condition is R 5And R 6In the middle of have only one to represent H;
R 7When occurring, represent H, halogen, low alkyl group or lower alkoxy independently at every turn; M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=S) and C (=O)), NR ", O, S, S (O), S (O 2);
N represents the integer of 1-5; And
Q represents nitrogenous heteroaryl ring, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, the Q among the formula IIa represents for example dialkylamine of the amino substituting group of uncle.In some embodiments, the Q among the formula IIa represents replacement or unsubstituted nitrogen heterocyclic ring for example morpholine, piperidines, piperazine or tetramethyleneimine.In some embodiments, Q represents nitrogenous heteroaryl ring, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, in formula II,
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represent C (=O), C (=S), S (O 2) or CH 2
R ' represents H, low alkyl group, metal counter ion or alkaline-earth metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R " ' represent H or choose the low alkyl group that replaces wantonly;
R 5Represent H, P (=O) (OR ') 2, M nJK or M nQ;
R 6Represent H, OH or M nQ, condition is R 5And R 6In the middle of have only one to represent H;
R 7Represent H, halogen, hydroxyl, low alkyl group or lower alkoxy;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', N (R ") 2Or N (R ') SO 2R ";
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=S) and C (=O)), NR ", O, S, S (O) or S (O 2);
In the time of in being present in B, n represents the integer of 1-4, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or is unsubstituted: nitrogenous heteroaryl ring, secondary amino group substituting group, the amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, Q represents for example dialkylamine of the amino substituting group of uncle, or replacement or unsubstituted nitrogen heterocyclic ring for example morpholine, piperidines, piperazine or tetramethyleneimine.
In some embodiments, in formula II,
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represents O, S, S (O 2), C (=O), C (=S), CH 2Or NR ";
R ' represents H, low alkyl group, metal counter ion or alkaline-earth metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R " ' represent H or choose the low alkyl group that replaces wantonly;
R 5Represent H, P (=O) (OR ') 2, M nJK or M nQ;
R 6Represent H, OH or M nQ, condition is R 5And R 6In the middle of have only one to represent H;
R 7Represent H, halogen, hydroxyl, low alkyl group or lower alkoxy;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', N (R ") 2Or N (R ') SO 2R ";
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=S) and C (=O)), NR ", O, S, S (O) or S (O 2);
In the time of in being present in B, n represents the integer of 1-4, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or unsubstituted secondary amino group substituting group.
In some embodiments, in formula II,
B represents M nR 8
Ar represents aryl or heteroaryl ring;
V represents O, S or N-CN;
W represents O, S, S (O 2), C (=O), C (=S), CH 2Or NR ";
R ' represents H, low alkyl group, metal counter ion or alkaline-earth metal counter ion at every turn independently when occurring;
R " when occurring, represent H or low alkyl group independently at every turn;
R " ' represent H or choose the low alkyl group that replaces wantonly;
R 5Represent M nJK, condition is R 5Not CH 2COOH;
R 6Represent H, OH or M nQ;
R 7Represent H, halogen, hydroxyl, low alkyl group or lower alkoxy;
R 8Represent replacement or unsubstituted alkyl, alkenyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', N (R ") 2Or N (R ') SO 2R ";
M when occurring, represent independently at every turn replace or unsubstituted methylene radical (comprise C (=S) and C (=O)), NR ", O, S, S (O) or S (O 2);
In the time of in being present in B, n represents the integer of 1-4, when being present in R 5When middle, n represents the integer of 0-6, when being present in R 6When middle, n represents the integer of 1-3; And
The Q representative replaces or is unsubstituted: nitrogenous heteroaryl ring, secondary amino group substituting group, the amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, in formula II, Q replaces or unsubstituted nitrogenous heteroaryl ring, and R 8Can represent and replace or unsubstituted morpholino, piperazinyl or cyclohexyl.In formula II, R " can represent H.
M can also represent CH 2In some embodiments, in formula II, W represents CH 2, and the NR of at least one M representative replacement ".
In some embodiments, in formula II, the Q representative replaces or unsubstituted secondary amino group.In some embodiments, in formula II, the Q representative replaces or unsubstituted uncle's amino.In some embodiments, in formula II, the Q representative replaces or unsubstituted nitrogen heterocyclic ring.
In some embodiments, in formula II, the Q representative replaces or is unsubstituted: nitrogenous heteroaryl ring, the amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, in formula II, R 5Represent M nQ, and Q representative replacement or unsubstituted: nitrogenous heteroaryl ring, the amino substituting group of uncle or nitrogen heterocyclic ring.
In some embodiments, in formula II, the Q representative replaces or unsubstituted uncle's amino.
In some embodiments, in formula II, the Q representative replaces or unsubstituted nitrogen heterocyclic ring.
In some embodiments, in formula II, R 5Represent M nQ, and the Q representative replaces or unsubstituted secondary amino group.
In some embodiments, in formula II, R 5Represent M nQ, and the Q representative replaces or unsubstituted nitrogenous heteroaryl ring.
In some embodiments, in formula II, R 8Representative replaces or unsubstituted morpholino, piperazinyl or cyclohexyl.
In some embodiments, in formula II, R " represent H.
In some embodiments, in formula II, W represents CH 2
In some embodiments, in formula II, when being connected with Q, M is CH 2, S (O 2), C (=S) or C (=O).
In some embodiments, in formula II, when being connected with Q, M is CH 2
In some embodiments, in formula II, the M that is connected with Q is CH 2, S (O 2), C (=S) or C (=O).
In some embodiments, in formula II, V is O, and M represents NH, and R 8Have following structure:
Figure C200480042035D00161
Wherein Z represents O or NR ".
In some embodiments, AR represents benzyl ring, and the R that is occurred 6And R 7Represent H.
In certain embodiments, the invention provides compound or its prodrug, isomer, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula V structure:
Figure C200480042035D00162
Wherein
R 8Representative replaces or unsubstituted heterocycle;
The Q representative replaces or is unsubstituted: secondary amino group substituting group, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
As mentioned above, in some embodiments, R 8Can represent morpholino or piperazinyl ring.
Some embodiments in, as mentioned above, Q can represent piperazine, morpholine, piperidines, pyridine, pyrroles, oxazole, isoxazole, imidazoles or pyrazoles.
Some embodiments comprise compound or its prodrug, isomer, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer that is selected from A34, A36, A37, A44, A46 and A76-A82.
Some embodiments comprise compound or its prodrug, isomer, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer that is selected from A47, A49, A51 and A82.
In some embodiments, W represents O, S (O 2), C (=O), C (=S), S, CH 2Or NR ".
As mentioned above, in some embodiments, the W representative replaces or is unsubstituted: nitrogenous heteroaryl ring, the amino substituting group of uncle or nitrogen heterocyclic ring.
In certain embodiments; when occurring, suitable substituents can independently comprise alkyl, oxo base, acyl amino, hydroxyl, carbonyl, alkylsulfonyl, ester, acid amides, NR at every turn ", hydroxyalkyl, alkoxyalkyl, aryl, heterocyclic radical, cycloalkyl or oligomeric (ethylene glycol).In certain embodiments, wherein Q represents the secondary amino group substituting group, and suitable substituents comprises alkyl, alkoxyalkyl, hydroxyalkyl and hydroxy alkoxy alkyl.Those skilled in the art will recognize easily that the substituting group of enumerating not is limit, can use many other suitable substituents.
Some embodiment can comprise the medicinal compositions that comprises pharmaceutically acceptable vehicle and any kind compound disclosed herein, and some embodiment comprises the method for the treatment of excess proliferative disease, and this method comprises and gives animal any kind compound disclosed herein.
In certain embodiments, compound disclosed herein may be used on suppressing in the method for cell proliferation, and these class methods comprise makes cell contact with the compound that discloses type herein; Or be applied in the method for treatment virus infection (for example infection that causes by human immunodeficiency virus (HIV)), these class methods comprise and give the Mammals compound of type openly herein.Some embodiment comprises the method for the alopecia that treatment or prevention of chemotherapy or radiotherapy cause, these class methods comprise openly the compound of type and one or more chemotherapy or radiotherapy are united and given Mammals herein.Compound disclosed herein also can be used for preparing medicine.
The compounds of this invention also can be used for treating for example excess proliferative illness of illness.Can be with compound to human or animal's administration.The compounds of this invention can be used for suppressing cell proliferation, for example by proliferating cells is contacted the described effect that realizes with compound.By compound to the Mammals administration, The compounds of this invention also can be used for treating virus infection.
The medicine that uses The compounds of this invention to form can be used as treatment or prevention illness (for example excess proliferative illness), virus infection, the alopecia of chemotherapy inductive, with the medicine of the kinase activity diseases associated of cyclin dependent etc.Medicine can be used for treating any illness described herein.
It is utilizable using the whole bag of tricks of The compounds of this invention.For example, can the employing method suppress the kinases of cyclin dependent, comprise any compound to host's administering therapeutic significant quantity of this treatment of needs.Can also the employing method treat the illness relevant, comprise compound to host's administering therapeutic significant quantity of this treatment of needs with the kinases of cyclin dependent.The invention still further relates to the method for treatment people or other animal.In some embodiments, can use the composition that comprises one or more The compounds of this invention for the treatment of significant quantity to treat people or other animal.
In certain embodiments, the invention provides a kind of new medicinal compositions, said composition comprises formula (I) or (II) compound or any other compound disclosed herein of pharmaceutically acceptable carrier and treatment significant quantity, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.
In another embodiment, the invention provides a kind of new treatment cancer or the method for other proliferative disease or other disease, this method comprises that the host that this treatment needs are arranged treats the formula of significant quantity (I) or (II) compound or any other compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.
In another embodiment, the invention provides a kind of new treatment cancer or the method for other proliferative disease or other disease, this method comprises that the host that this treatment needs are arranged treats significant quantity: (a) formula (I) or (II) compound or any other compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer; (b) at least a compound that is selected from anticarcinogen and antiproliferative agents.
As described herein, inhibitor of the present invention can suppress the cell cycle machine, thereby can be used for regulating cell cycle progression, finally controls cell growth and differentiation.This compounds can be used for treating the patient and excessive cell proliferation diseases associated, for example cancer, psoriatic, relate to the Immunological diseases that do not need white corpuscle propagation; Can be used for treating restenosis and other smooth muscle cell disease etc.This compounds also can be used for suppressing I type human immunodeficiency virus (HIV-I) and transcribes (Wang etc., J.Virology 75:7266-7279 (2001).
Also described The compounds of this invention herein and can be used for preparing medicine, this medicine can be used for treating those diseases that disease is for example discussed herein.
IV. accompanying drawing summary
Fig. 1 represent following situation be exposed in the compound A-13 7 influenced: (a) analyze the cell cycle analysis of the HCT-116 cell that carries out by PIFACS; (b) induce the PARP division.
Fig. 2 (a) dose response; (b) 7 pairs of HCT-116 tumour cells of time-histories explanation compound A-13 produce the irreversible effect of clone (clongeneic) survival.
Fig. 3 graphic compound B16 produces the irreversible effect of clone's survival to the HCT-116 tumour cell, represents with time-histories.
Fig. 4 represents to compare with normal (IMR90) cell of the stagnation that is exposed to same compound, and the viability that is exposed to stagnation tumour (HCT-116) cell of compound A-13 7 descends.
Fig. 5 represents with all cpds of the present invention and carries out the result that the test of HCT-116 xenotransplantation tumour obtains.
Fig. 6 represents to carry out the result that A2780 xenotransplantation tumour test obtains with compound A-13 7, by (a) time-histories of tumour size under various dosage; (b) table of the remarkable tolerance (salient metrics) in test expression.
Fig. 7 represents to carry out the result that PC3 xenotransplantation tumour test obtains with compound A-13 7, by (a) time-histories of tumour size under various dosage; (b) the significance scale in the test is represented.
Fig. 8 represents to carry out the result that A2780 xenotransplantation tumour test obtains with compound B-11 6, by (a) time-histories of tumour size under various dosage; (b) the significance scale in the test is represented.
Fig. 9 represents CDK 2The chips incorporate of/cyclin E and loading CM5-inhibitor obtains result's example.K DAccording to these data computation, equal 8,0+/-2,8nM.
V. exemplary describes in detail
The present invention relates to the kinase inhibitor (cdks) of new cyclin dependent, especially but be not only cdk/ cyclin mixture inhibitor.As described herein, inhibitor of the present invention can suppress the cell cycle machine, thereby can be used for regulating cell cycle progression, finally controls cell growth and differentiation.This compounds can be used for treating the patient and excessive cell proliferation diseases associated, for example treats cancer, psoriatic, relates to the Immunological diseases that do not need white corpuscle propagation; Can be used for treating restenosis and other smooth muscle cell disease etc., more be discussed in detail below.
In one embodiment, the invention provides compound, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula I structure:
Figure C200480042035D00201
Wherein
R 8Representative replaces or unsubstituted heterocycle or replacement or unsubstituted morpholino, replacement or unsubstituted piperazinyl or replacement or unsubstituted cyclohexyl;
F represents (CH 2) n, wherein n is the integer of 1-6, in some embodiments, n is 1;
The Q representative replaces or the amino substituting group of unsubstituted secondary amino group substituting group, replacement or unsubstituted uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, the Q among the formula I represents uncle amino substituting group, for example dialkylamine.In some embodiments, the Q among the formula I represents replacement or unsubstituted nitrogen heterocyclic ring for example morpholine, piperidines, piperazine or tetramethyleneimine.In some embodiments, Q represents nitrogenous heteroaryl ring, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In some embodiments, R 8Representative:
Wherein Z is O or NR "; And
R " represent H or low alkyl group.
In some embodiments, the compound with formula I structure does not comprise one or more following compounds:
Figure C200480042035D00212
In some embodiments, formula I compound comprises the compound that one or more are listed in table.For example, formula I compound can comprise one or more compound B-11-B20 and C2.
As mentioned above; in some embodiments, suitable substituents can comprise alkyl, oxo base, acyl amino, hydroxyl, carbonyl, alkylsulfonyl, ester, acid amides, NR at every turn independently when occurring ", hydroxyalkyl, alkoxyalkyl, aryl, heterocyclic radical, cycloalkyl or oligomeric (ethylene glycol).In some embodiments, when Q represented the secondary amino group substituting group, suitable substituents comprised alkyl, alkoxyalkyl, hydroxyalkyl and hydroxy alkoxy alkyl.Those skilled in the art should recognize easily that cited substituting group is not limit, and can use a lot of other suitable substituents.
In another embodiment, the present invention also provides the compound with formula II structure, comprises its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer:
Figure C200480042035D00221
Wherein
B represents M nR 8
Ar represents for example phenyl ring of aryl or heteroaryl ring;
V represents O, S or N-CN, preferred O or S;
W represents O, S, S (O 2), C (=O), C (=S), CH 2Or NR ";
When occurring at every turn, independent H, low alkyl group or metal counter ion for example alkali or the alkaline-earth metal counter ion represented of R ';
When occurring at every turn, R " independent H or the low alkyl group represented, preferred H;
R " ' represent H or the optional low alkyl group that replaces, preferably have the substituting group that is selected from ester, acid amides, acyl amino or acyloxy;
R 5Represent H, P (=O) (OR ') 2, M nJK or M nQ;
R 6Represent H, OH or M nQ, optimum condition are R 5And R 6In one and have only one to represent H;
When occurring at every turn, R 7Independent H, halogen, hydroxyl, low alkyl group, for example methyl or lower alkoxy, for example methoxyl group represented;
R 8Represent replacement or unsubstituted alkyl, thiazolinyl, alkynyl, alkoxyl group, aryl, heteroaryl, cycloalkyl, heterocyclic radical or amine;
J represent C (=O), C (=S) or SO 2
K represents OR ', NR " or N (R ') SO 2R ";
When occurring, M independently represents and replaces or unsubstituted methylene radical (for example by replacements such as low alkyl group, oxo base, hydroxyls), NR at every turn ", O, S, S (O) or S (O 2), preferred NR " or CH 2, or when being connected, represent CH with W or Q 2, S (O 2), C (=S) or C (=O);
When occurring in B, n represents the integer of 0-10, preferred 1-7 or even 1-4, at R 5In when occurring, represent the integer of 0-6, at R 6In when occurring, represent the integer of 1-3; With
The Q representative replaces or be unsubstituted: nitrogenous heteroaryl ring is pyrroles, tetrazolium, oxazole, oxadiazole, isoxazole, imidazoles or pyrazoles for example; The secondary amino group substituting group is monoalkylamine, aromatic yl alkyl amine, heteroarylalkyl amine for example; The amino substituting group of uncle is dialkylamine for example; Or nitrogen heterocyclic ring for example morpholine, piperidines, piperazine, pyridine or tetramethyleneimine.
In certain embodiments, represent N (R ') SO as K 2R " time, R " represent low alkyl group.
In certain embodiments, work as R 5Be M nDuring JK, R 5Be not CH 2COOH.
In certain embodiments; when occurring at every turn; suitable substituents independently comprises alkyl, oxo base, hydroxyl, alkoxyl group, hydroxyl-alkoxyl group, carbonyl, alkylsulfonyl, ester, acid amides, NR ", alkylogen, acyl amino, or replacement or unsubstituted aryl, heteroaryl, heterocyclic radical, cycloalkyl, oligomeric (ethylene glycol) etc.Aryl and heteroaryl can use any suitable substituents to comprise that any above-mentioned listed those substituting groups are conspicuous to those skilled in the art.
In certain embodiments, R 8Represent any following substituting group: alkyl, thiazolinyl, alkynyl, alkoxyl group, hydroxyl-alkoxyl group, aryl, amine or heteroaryl.In certain embodiments, any aforementioned substituting group can be chosen wantonly by any described substituting group and replace, or even by halogen ,-CN, N 3, NO 2Or haloalkyl replaces.Other suitable substituents for example also can comprise cyclohexyl ,=O, carbonyl, alkylsulfonyl, carboxyl, sulfonic group (sulfoxyl), acid amides, heterocycle, ester or ether.
In certain embodiments, when at R 5The middle appearance and and R 8During connection, M is the NR for replacing at least once ".
In some embodiment, comprise in any above embodiment R 8Have following form:
Figure C200480042035D00241
Wherein Z represents O or NR ".In certain embodiments, R 8Represent morpholino or cyclohexyl.In some this type of embodiment, M nBe NR ", preferred NH.In certain embodiments, V is O.
In certain embodiments, W represents CH 2In some this type of embodiment, M is the NR for replacing at least once ".
In certain embodiments, R wherein " ' existing and low alkyl group for replacing, this low alkyl group is selected from following substituting group and replaces by 1-3 (preferred 1) is individual: low alkyl group, low-grade halogenated alkyl, NR 8R 8a, NR " C (O) R 8,=O, COR 8, CO 2R 8, NR " CO 2R 8, C (O) NR 8R 8a, NR " C (O) NR 8R 8a, NR " C (S) NR 8R 8a, C (S) NR 8R 8a, NR " SO 2NR 8R 8a, SO 2NR 8R 8a, NR ' ' SO 2R 8a, SO 2R 8a, NR ' ' SO 2R 8a, by 0-5 R " ' C that replaces 3-10Carbocyclic ring and contain 1-4 and be selected from the heteroatomic of O, N and S by 0-3 R 85 yuan of-10 yuan of heterocycles, wherein R replacing 8Represent H, C 1-4Haloalkyl, NR 8aR 8a, NR " C (O) OR 8a, NR " C (O) R 8a, COR 8a, CO 2R 8a, CONR 8aR 8a, NHC (O) NR 8aR 8a, NHC (S) NR 8aR 8a, SO 2NR 8aR 8a, SO 2R 8a, C 1-4Alkyl, phenyl, benzyl, choose by 0-3 R " ' C of replacement 2-10The C of alkenyl substituted 5-10Alkyl, by 0-3 R " ' C that replaces 2-10Alkynyl ,-(CF 2) mCF 3, by 0-5 R " ' C that replaces 3-10Carbocyclic ring, and by 0-3 R " ' replace contain 1-4 heteroatomic 5 yuan to the 10 yuan heterocycles that are selected from O, N and S; When occurring at every turn, R 8aIndependent representative is selected from the group of H, low alkyl group, phenyl and benzyl.
In certain embodiments, R " ' contain amino-acid residue for example Xie Ansuan or glycine residue, for example R " ' low alkyl group residue for being replaced by amino-acid residue by acid amides or ester bond.
In preferred embodiments, R 5W and R 6(ortho position) adjacent one another are on Ar, the key adjacent (ortho position) that preferred discord is connected with three ring parent nucleus.
In certain embodiments, V represents S or N-CN.In certain embodiments, Ar represents hetero-aromatic ring.
In some embodiment of formula II, W represents O, S or NR ".In certain embodiments, R 5Represent H, P (=O) (OR ') 2Or M nQ.In certain embodiments, when occurring at every turn, R 7Independent for example methyl or the lower alkoxy methoxyl group for example of halogen, hydroxyl, low alkyl group of representing.In certain embodiments, when appearing at R 5The time, n represents the integer of 0-5, preferred 1-5, more preferably 2-4.
In some embodiment of formula II, W represents O, CH 2, C (=O), C (=S) or SO 2In certain embodiments, R 5Represent M nJK or M nQ.In certain embodiments, R 6And R 7Represent H.In certain embodiments, M represent C (=O) or CH 2In certain embodiments, preferred n is 1, and in other embodiments, n can be 0.In certain embodiments, preferred J be C (=O), K is OR ' or N (R ') SO 2R ".In certain embodiments, N (R ') SO 2R " be NHSO 2R ".
In certain embodiments, the Q representative replaces or unsubstituted nitrogenous hetero-aromatic ring.In certain embodiments, the Q representative replaces or unsubstituted hetero-aromatic ring, for example contains 5 yuan or 6 yuan of rings of at least two nitrogen-atoms.In certain embodiments, Q can be for replacing or unsubstituted tetrazolium or oxadiazole.In certain embodiments, Q can be replacement or unsubstituted pyridine, piperidines or piperazine.
In certain embodiments, Q represents the secondary amino group substituting group.In some this type of embodiment, the substituting group on the secondary amino group substituting group is selected from alkyl, alkoxyalkyl, hydroxyalkyl and hydroxy alkoxy alkyl.
In some embodiment of formula II, W represent C (=O), SO 2Or C (=S), R 6And R 7Represent H, R 5Represent M nQ, wherein n represents 0, and the Q representative replaces or unsubstituted nitrogenous hetero-aromatic ring.In certain embodiments, W represents CH 2, R 6And R 7Represent H, R 5Represent M nQ, wherein n represents 0, and the Q representative replaces or unsubstituted nitrogenous hetero-aromatic ring.
In certain embodiments, W represents S, O or NR ", R 6And R 7Represent H, R 5Represent M nJK, wherein n is the integer of 1-3, J be C (=O), K is OR ' or N (R ') SO 2R ".
In certain embodiments, W represents S, O or NR ", R 6And R 7Represent H, R 5Represent M nQ, wherein n is the integer of 1-3, Q is for replacing or unsubstituted 5 member heterocyclic ring containing nitrogens.In this type of embodiment, preferred n is 1.In certain embodiments, Q contains at least two nitrogen-atoms.
In certain embodiments, W represents S, O or NR ", R 6And R 7Represent H, R 5Represent M nQ, wherein n represents the integer of 1-3, and Q is for replacing or unsubstituted 6 member heterocyclic ring containing nitrogens.In some this type of embodiment, n is 2, M nRepresent CH 2C (=O).
In certain embodiments, W represents O, S or NR ", R 6And R 7Represent H, R 5Represent M nQ, wherein M is CH 2, n is the integer of 1-3, Q is for replacing or unsubstituted nitrogen heterocyclic ring.
In certain embodiments, wherein Q represents the nitrogen heterocyclic ring that replaces, for example piperazine, morpholine, piperidines, pyridine, thiazole, oxadiazole, tetrazolium, pyrroles etc., suitable substituents comprises replacement or unsubstituted alkyl, amino-alkyl, alkoxyl group, aralkyl (for example benzyl), aryl (for example phenyl) and heteroaryl Li such as oxazolyl, piperazinyl, pyridyl, pyrryl.In some this type of embodiment, wherein Q contains the nitrogen that does not link to each other with M, and this nitrogen is for example replaced by such substituting group.
In certain embodiments, the present invention relates to have compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer of following structure:
Figure C200480042035D00261
C1。
In certain embodiments, the present invention relates to have compound or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer of following structure:
Figure C200480042035D00262
C5。
In some embodiment of formula II, the invention provides compound with formula IIa structure, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer:
Figure C200480042035D00271
Wherein
W and Z independently represent O or NR ";
When occurring at every turn, independent H, low alkyl group or metal counter ion, for example alkali or the alkaline-earth metal counter ion represented of R ';
When occurring at every turn, R " independent H or the low alkyl group represented, preferred H;
R 5Represent H, P (=O) (OR ') 2Or M nQ;
R 6Represent H, OH or M nQ, optimum condition are R 5And R 6In one and have only one to represent H;
When occurring at every turn, R 7Independent for example methyl or the lower alkoxy methoxyl group for example of hydrogen, halogen, low alkyl group of representing;
When occurring, M independently represents and replaces or unsubstituted methylene radical (for example by replacements such as low alkyl group, oxo base, hydroxyls), NR at every turn ", O, S, S (O) or S (O 2), preferred CH 2, or when being connected, represent CH with W or Q 2, S (O 2), C (=S) or C (=O);
When being present in R 5The time, n represents the integer of 1-5, and preferred 2-4 is when being present in R 6The time, n represents the integer of 1-3; With
Q represents for example amino substituting group of pyrroles, oxazole, isoxazole, imidazoles or pyrazoles, uncle for example morpholine, piperidines, piperazine or tetramethyleneimine of dialkylamine or replacement or unsubstituted nitrogen heterocyclic ring for example of nitrogenous heteroaryl ring.
In certain embodiments, Q represents for example dialkylamine of the amino substituting group of uncle.In certain embodiments, Q represents replacement or unsubstituted nitrogen heterocyclic ring for example morpholine, piperidines, piperazine or tetramethyleneimine.In certain embodiments, Q represents nitrogenous hetero-aromatic ring, the amino substituting group of uncle or replacement or unsubstituted nitrogen heterocyclic ring.
In certain embodiments, the compound with formula II structure does not comprise the compound with formula IIa structure.
Exemplary formula II and IIa compound comprise those compounds shown in the table B.
The present invention also provides has the compound that is selected from following structure: A3, A7 to A29, A31, A33 to A37, A40, A41, A44 to A47, A49, A51, A56, A57, A65, A69 to A82, C1, C2 and C5 comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In certain embodiments, the invention provides compound, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with structure A37.
In another embodiment, the invention provides the prodrug or the pharmacy acceptable salt of isolated compound A37 metabolite.A prodrug or the pharmacy acceptable salt that preferred this embodiment is compd A 68 or C5.
In another embodiment, the invention provides and have the compound that is selected from following structure: B1 to B20, C1, C2 and C5, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In a preferred embodiment, the invention provides compound, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with B16 or C5 structure.In another embodiment, the invention provides compound, comprise its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with B3 structure.
In another embodiment, the invention provides the prodrug or the pharmacy acceptable salt of isolated compound B16 metabolite.A prodrug or the pharmacy acceptable salt that preferred this embodiment is a compd B 3.
In certain embodiments, the invention provides compound or its prodrug, isomer, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer with formula V structure:
Wherein
R 8Representative replaces or unsubstituted heterocycle; With
The Q representative replaces or is unsubstituted: amino substituting group of uncle or nitrogen heterocyclic ring.
In other embodiments of the present invention, compound is an exemplary compounds shown in table C, D and the E, the present invention includes isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or the steric isomer of wherein describing compound.
In another embodiment, the invention provides a kind of new medicinal compositions, said composition contains for example C5 of formula I, the II of acceptable carrier pharmaceutically and treatment significant quantity or IIa compound or any compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In a preferred embodiment, this medicinal compositions comprises the following compound that is selected from of treatment significant quantity: A1 to A82, B1 to B20 and C1 to C5, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In another embodiment, this medicinal compositions comprises the compound A-13 7 for the treatment of significant quantity or the prodrug or the pharmacy acceptable salt of B16 metabolite, the metabolite that preferably has A68 or C5 structure.
In some embodiments, the invention provides the compound described herein of one or more purifying or synthesized form.
In another embodiment, the invention provides the method that a kind of new treatment cancer or other proliferative disease or other disease comprise following any disease or illness, this method comprises that the host that this treatment of needs is arranged treats the formula I of significant quantity, II or IIa compound or any compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In certain embodiments, at least a compound that is selected from cancer therapy drug and anti-proliferative drugs can with formula I, II or IIa compound or any compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer Combined Preparation.In a preferred embodiment, this type of methods of treatment comprises suitably treats the following compound of being selected from of significant quantity: A1 to A82, B1 to B20 and C1 to C5, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.Term Combined Preparation used herein comprises such therapy: wherein two kinds of medicines give the patient with the unitary agent Combined Preparation in addition for example with independently preparation while or different time administration, or as the part of treatment plan.
In another embodiment, the invention provides a kind of method of preparing medicinal compositions, said composition contains the formula I that treats significant quantity, II or IIa compound or any compound disclosed herein, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer, with optional pharmaceutically acceptable carrier. in a preferred embodiment, what this medicinal compositions comprised the treatment significant quantity is selected from following compound: A1 to A82, B1 to B20 and C1 to C5, or its isomer, prodrug, tautomer, pharmacy acceptable salt, N-oxide compound or steric isomer.In another embodiment, this medicinal compositions comprises the compound A-13 7 for the treatment of significant quantity or the prodrug or the pharmacy acceptable salt of B16 metabolite, the metabolite that preferably has A68 or C5 structure.
In some embodiments again, medicinal compositions of the present invention is used for the treatment of disease, and for example cancer and other proliferative disease or other disease comprise following any disease or illness.
In some embodiment of the group that wherein use to replace of the present invention, suitable substituents for example can comprise halogen, hydroxyl, carbonyl (for example ketone, aldehyde, carboxyl, ester, acyl group), thiocarbonyl (for example thioester, thioacetate, thiocarboxylic), alkoxyl group, phosphoryl (for example phosphonic acid ester, phosphinate), phosphoric acid ester, phosphonic acid ester, phosphinate, amino, amino-alkyl, amido, amidine, imines, cyano group, nitro, azido-, sulfydryl, alkylthio, ether ,-CF 3, alkyl, thiazolinyl, alkynyl, cycloalkyl, alkoxyl group, silyl, alkylsulfonyl (for example sulfuric ester, sulfonamido, sulfamyl, sulphonate), heterocyclic radical, aralkyl (for example benzyl) or aromatics or heteroaromatic moiety (for example phenyl, oxazolyl, piperazinyl, pyridyl, pyrryl).Itself also can be substituted this type of substituting group or not be substituted.
Ii. definition
Following term used herein and expression have specified implication.The compounds of this invention can contain the carbon atom of asymmetric replacement, and therefore separable is optically-active form or racemic modification.It is known in the art how preparing optically active form, for example by resolution of racemates or synthetic with the opticity raw material.Except the specific stereochemistry or isomer formula that particularly point out, will comprise all chiralitys, diastereomer, racemic modification and all geometrical isomers of structure.Be useful on the method for preparing The compounds of this invention and wherein preparation intermediate be considered as a part of the present invention.
The present invention will comprise that all appear at the atom isotope on the The compounds of this invention.Isotropic substance comprises that those have the same atoms ordinal number but the atom of different mass number.In general non-limiting example, the isotropic substance of hydrogen comprises tritium and deuterium.The isotropic substance of carbon comprises 12C and 14C.
Term " alkyl " will comprise side chain and the straight chain radical of saturated aliphatic alkyl with clear and definite number carbon atom.The example of alkyl includes but not limited to methyl, ethyl, n-propyl, sec.-propyl, normal-butyl, sec-butyl, the tertiary butyl, n-pentyl and sec.-amyl sec-pentyl secondary amyl.In addition; this term comprises the alkyl that does not replace and replace, and the latter refers to that moieties has one or more quilts but is not limited to the hydrogen substituting group of following group displacement: halogen, hydroxyl, carbonyl, alkoxyl group, ester, ether, cyano group, phosphoryl, amino, imino-, amido, sulfydryl, alkylthio, thioester, alkylsulfonyl, nitro, heterocyclic radical (heterocyclo), aryl or heteroaryl.Those skilled in the art also will understand when suitable, replace part and itself also can be substituted.Term " low alkyl group " refers to that those have the alkyl of 1 to 6 carbon atom, and preferred 1 to 4 carbon atom, term " lower alkoxy " refer to this class low alkyl group of being connected with Sauerstoffatom.In certain embodiments, the preferred alkyl substituting group is a low-grade alkyl substituent.
Term used herein " halo " or " halogen " refer to fluorine, chlorine, bromine and iodine.
Term " aryl " will represent that the aromatics part is such as but not limited to phenyl, indanyl or naphthyl.
Term " cycloalkyl " and " bicyclic alkyl " will be represented any stable loop systems, and it can be saturated or part is unsaturated.This examples of groups includes but not limited to cyclopropyl, cyclopentyl, cyclohexyl, norborneol alkyl, dicyclo [22] nonane, adamantyl or tetralyl (1,2,3,4-tetralin).
" carbocyclic ring " used herein or " carbocylic radical " will be represented any stable 3 yuan to 7 yuan monocycles or dicyclo or 7 yuan to 13 yuan dicyclos or three rings, wherein any ring can for saturated, part is unsaturated or aromatics.This type of isocyclic example includes but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, suberyl, adamantyl, ring octyl group, [3.0] double-octane, [4.0] bicyclic nonane, [4.0] dicyclo decane (naphthalane), [2.2] double-octane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl or tetralyl (1,2,3, the 4-tetraline).
That term used herein " heterocycle " or " heterocyclic ring system " will be represented will be saturated, part is unsaturated or undersaturated 5 yuan to 7 yuan monocycles or dicyclo or 7 yuan to the 10 yuan bicyclic heterocycles (aromatics/heteroaryl) stablized, it forms and comprises any bicyclic radicals by carbon atom and 1 to 4 heteroatoms that independently is selected from N, O and S, and wherein any heterocycle and the phenyl ring of above definition condense.Nitrogen and sulfur heteroatom can be chosen wantonly oxidized.Heterocycle can be connected to form rock steady structure in any heteroatoms or carbon atom and its side group.If the compound that generates is stable, described herein heterocycle can replace on carbon or nitrogen-atoms.If refer in particular to, the nitrogen on the heterocycle can be chosen wantonly by quaternized.In certain embodiments, when heterocyclic S and O total atom number surpassed 1, then these heteroatomss need not be adjacent one another are.S total atom number on the preferred heterocycle is not more than 1.Stable 5 yuan to 7 yuan monocycles or dicyclo or 7 yuan to 10 yuan bicyclic heterocycle aromatic rings will be represented in term used herein " fragrant heterocyclic ring system ", and it is made up of carbon atom and 1 to 4 heteroatoms that independently is selected from N, O and S.S and O total atom number on the preferred fragrant heterocycle are not more than 1.The heterocyclic example includes but not limited to the 1H-indazole, the 2-Pyrrolidone base, 2H16H dithiazine base, the 2H-pyrryl, the 3H-indyl, the 4-piperidone base, the 4aH-carbazole, the 4H-quinolizinyl, 6H-1,2,5-thiadiazine base, acridyl, the azocine base, benzimidazolyl-, benzofuryl, benzothienyl (benzothiofuranyl), benzothienyl (benzothiophenyl) benzoxazolyl, benzothiazolyl, the benzotriazole base, the benzo tetrazyl, the benzoisoxazole base, the benzisothiazole base, benzimidazoline ketone group (benzimidazalonyl), carbazyl, the 4aH-carbazyl, the P-carbolinyl, chromanyl, benzopyranyl, cinnolinyl, decahydroquinolyl, 2H, 6H dithiazine base, dihydrofuran also [2,3-b] tetrahydrofuran (THF), furyl, the furazan base, imidazolidyl, imidazolinyl, imidazolyl, the 1H-indazolyl, indoline base (indolenyl), indolinyl, the indolizine base, indyl, isobenzofuran-base (isobenzofuranyl), the isochroman base, iso indazolyl, iso-dihydro-indole-group, pseudoindoyl, isoquinolyl, isothiazolyl isoxazolyl, morpholinyl, 1, the 5-phthalazinyl, octahydro isoquinolyl oxadiazole base, 1,2,3-oxadiazole base, 1,2,4-oxadiazole base, 1,2,5-oxadiazole base, 1,3,4-oxadiazole base oxazolidinyl oxazolyl oxazolidinyl pyrimidyl (perimidinyl), phenanthridinyl, the phenanthroline base, the phenarsazine base, phenazinyl, phenothiazinyl, benzene oxathiin base (phenoxathiinyl) phenoxazinyl, 2, the 3-phthalazinyl, piperazinyl, piperidyl, pteridyl, piperidone base, the 4-piperidone base, pteridyl, purine radicals, pyranyl, pyrazinyl, pyrazolidyl, pyrazolinyl, pyrazolyl, pyridazinyl, Bi Ding Bing oxazole, pyridine-imidazole, the pyrido thiazole, pyridyl (pyridinyl), pyridyl, pyrimidyl, pyrrolidyl, pyrrolinyl, pyrryl, quinazolyl, quinolyl, the 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, carbolinyl, tetrahydrofuran base, tetrahydro isoquinolyl, tetrahydric quinoline group, 6H-1,2,5-thiadiazine base, 1,2, the 3-thiadiazolyl group, 1,2, the 4-thiadiazolyl group, 1,2, the 5-thiadiazolyl group, 1,3, the 4-thiadiazolyl group, thianthrenyl, thiazolyl, thienyl, the thieno-thiazolyl, thiophene Bing oxazolyl, the Thienoimidazole base, thienyl (thiophenyl), triazinyl, 1,2, the 3-triazolyl, 1,2,4 triazolyls, 1,2, the 5-triazolyl, 1,3, the 4-triazolyl, xanthenyl.Preferred heterocycle includes but not limited to pyridyl, furyl, thienyl, pyrryl, pyrazolyl, imidazolyl, indyl, benzimidazolyl-, 1H-indazolyl, oxazolidinyl, benzotriazole base, benzoisoxazole base, oxindole base, benzoxazole quinoline base or isatin acyl group (isatinoyl).Also comprise and contain for example above heterocyclic condensed ring and spirocyclic compound.
" pharmacy acceptable salt " herein refers to the come into the open derivative of compound of this paper, wherein modifies parent compound by its acid of preparation or alkali salt.The example of pharmacy acceptable salt includes but not limited to the alkaline residue for example mineral or the organic acid salt of amine; Acidic residues is the alkali salt or the organic salt of carboxylic acid for example; Deng.Pharmacy acceptable salt for example comprises conventional non-toxic salt or the quaternary ammonium salt with the nontoxic inorganic or parent compound that organic acid forms.
For example, this type of conventional non-toxic salt comprises from mineral acid those salt of deutero-such as hydrochloric acid, Hydrogen bromide, sulfuric acid, thionamic acid, phosphoric acid, nitric acid for example; Salt with for example following organic acid preparation of usefulness: acetate, propionic acid, succsinic acid, oxyacetic acid, stearic acid, lactic acid, oxysuccinic acid, tartrate, citric acid, xitix, pamoic acid, toxilic acid, hydroxymaleic acid, toluylic acid, L-glutamic acid, phenylformic acid, Whitfield's ointment, sulfanilic acid, 2-acetoxy-benzoic acid, fumaric acid, toluenesulphonic acids, methylsulfonic acid, ethionic acid, oxalic acid, isethionic acid etc.
Pharmacy acceptable salt of the present invention can pass through the conventional chemical method, and is synthetic with the parent compound that contains alkalescence or acidic moiety.Usually, free acid that this type of salt can be by making these compounds or alkali form in water or organic solvent, or in both mixtures with the suitable alkali or the acid-respons preparation of stoichiometric quantity; Usually, preferred non-aqueous media such as ether, EtOAc, ethanol, Virahol or acetonitrile.Acceptable acid addition salts is listed in Remington ' s Pharmaceutical Sciences, and the 18th edition, Mack Publishing Company, Easton, PA, 1990, the 1445 pages, its content is attached to herein by reference.
Phrase used herein " pharmaceutically acceptable " refers in rational medical judgment scope, those be applicable to contact with humans and animals tissue and do not have too much toxicity, pungency, atopic reaction or other problem or complication, and reasonable benefit/risk the ratio compound, material, composition and/or the formulation that match.
Term used herein " prodrug " will comprise discharge the carrier of any covalent bonding of active parent drug of the present invention in vivo when this prodrug gives mammalian subject.Because known prodrug can increase the character (being solubleness, bioavailability, preparation property etc.) of the many needs of medicine, so the form release that The compounds of this invention can prodrug.Therefore, the present invention will comprise claimed compound prodrug, discharge the method for this class prodrug and contain the composition of this class prodrug.Prepare prodrug of the present invention by the functional group in the modified compound, with in routine operation or the body this modifier being dissociated into parent compound.Prodrug comprises The compounds of this invention, and wherein hydroxyl, amino or sulfydryl are respectively with when prodrug of the present invention gives mammalian subject, and it dissociates and forms any group bonding of free hydroxyl group, free amine group or free sulfhydryl groups.The example of prodrug includes but not limited to acetic ester (salt), manthanoate (salt) and benzoic ether (salt) derivative of alkohol and amine functional group in the The compounds of this invention.
Term " pure " or " purifying " are meant and do not have other organic molecule, especially impurity for example by product or degraded product in the molecule basically.In some embodiments, " pure " or " purifying " compound is to have at least 80% weight in forming, more preferably 95-99% weight, the most preferably organic compound of at least 99.8% weight (for example getting rid of water, buffer reagent, vehicle equimolecular in the pharmaceutical preparation that can be present in compound).
" replacement " will be illustrated in the selecteed appointment group displacement of one or more hydrogen that uses on the atom of expressing " replacement ", and condition is that the normal valency of specified atom does not exceed, and this replacement causes stable compound.When substituting group is that (promptly=O) during group, then 2 hydrogen on this atom are replaced for ketone or oxo.There is not ketone/oxo substituting group in the aromatics part.Exemplary substituting group comprises for example alkyl; perfluoroalkyl (for example trifluoromethyl); halogen; hydroxyl; carbonyl (carboxyl for example; alkoxy carbonyl; formyl radical or acyl group); thiocarbonyl (thioester for example; thioacetate or thiocarboxylic); alkoxyl group; phosphoryl; phosphoric acid ester; phosphonic acid ester; phosphinate; amino; amido; amidine; imines; cyano group; nitro; azido-; sulfydryl; alkylthio; sulfuric ester; sulphonate; sulfamyl; sulfonamido; alkylsulfonyl; carbocylic radical; heterocyclic radical; aralkyl; heteroaralkyl or aromatics or heteroaromatic moiety.Substituting group for example heterocyclic radical, aryl, alkyl etc. it will be understood by those skilled in the art that if suitably, can be substituted itself.
The term of The compounds of this invention " treatment significant quantity " expression effectively suppresses the amount that a class is called the kinase whose enzyme of cyclin dependent, or treatment host's the cancer or the amount of other proliferative disease or other disease symptoms.
Term used herein " anticancer " or " antiproliferative " medicine include but not limited to altretamine, busulfan, Chlorambucil, endoxan, ifosfamide, mustargen, melphalan, Tespamin, CldAdo, Fluracil, floxuridine, gemcitabine, Tioguanine, pentostatin, methotrexate, Ismipur, cytosine arabinoside, carmustine, lomustine, streptozocin, carboplatin, cis-platinum, oxaliplatin, iproplatin, four platinum, Lip river platinum, JM216, JM335, fludarabine, aminoglutethimide, flutamide, goserelin, leuproside, Magace, cyproterone acetate, tamoxifen, Anastrozole, bicalutamide, dexamethasone, stilboestrol, prednisone, bleomycin, dactinomycin, daunorubicin, Dx, idarubicin, mitoxantrone, losoxantrone, mitomycin-c, Plicamycin, taxol, docetaxel, Hycamtin, irinotecan, 9-aminocamptothecin (camptothecan), the 9-nitrocamptothecin, GS-211, JM 118, Etoposide, teniposide, vinealeucoblastine(VLB), vincristine(VCR), vinorelbine, Procarbazine, asparaginase, pegaspargase, Sostatin, estramustine and hydroxyurea.
Iii. dosage and preparation
Can produce any method treatment cancer or proliferative disease or other disease that active medicine is contacted with drug effect position in the mammalian body by giving the kinase inhibitor of cyclin dependent of the present invention.Can be by the ordinary method of any existing and medication combined use, give them with independent medicine or in the mode of each medicine associating.The chemical property of described inhibitor gives compound good dissolution characteristics herein, makes them be applicable to other preparation administration with intravenous formulations, topical formulations, oral preparations and following more detailed argumentation.They can be individually dosed, but preferably to select route of administration and standard pharmaceutical to be practiced as the basis, with the pharmaceutical carrier administration of selecting.At for example Remington ' s Pharmaceutical Sciences (Remington ' s Pharmaceutical Sciences.Mack Publishing Company, Easton, Pa., USA 1985) discussed suitable carriers (vehicle) and their preparation in the book.
On the other hand, the invention provides pharmaceutically acceptable composition, said composition contain treat significant quantity one or more The compounds of this invention for example above-claimed cpd and together the preparation one or more pharmaceutically acceptable carrier (additive) and/or thinners.As described in detail below, medicinal compositions of the present invention especially can be mixed with solid or liquid form of medication, comprise those forms that are fit to following route of administration: (1) oral administration, for example Haust (drenches) (water or non-aqueous solution or suspension), tablet, disposable large bolus injection liquid (boluses), powder, granule, tongue paste; (2) administered parenterally is for example by subcutaneous, intramuscular or intravenous injection, for example sterile solution or suspension; (3) the local use for example is used for creme, ointment or the sprays of skin; Or (4) intravaginal or internal rectum, for example vaginal suppository, creme or whipping agent.In certain embodiments, what medicinal preparations can be for non-pyrogen, patient temperature does not promptly raise.
Wetting agent, emulsifying agent and lubricant, for example sodium lauryl sulphate and Magnesium Stearate, and tinting material, releasing agent, coating material, sweeting agent, correctives and perfume compound, sanitas and antioxidant also can add composition.
The example of pharmaceutically acceptable antioxidant comprises: (1) water soluble antioxidant, for example xitix, cysteine hydrochloride, sodium pyrosulfate, Sodium Pyrosulfite, S-WAT etc.; (2) oil-soluble inhibitor, for example ascorbyl palmitate, butylated hydroxyanisol (BHA), butylated hydroxytoluene (BHT), Yelkin TTS, Tenox PG, alpha-tocopherol etc.; (3) metal chelator, for example citric acid, ethylenediamine tetraacetic acid (EDTA) (EDTA), sorbyl alcohol, tartrate, phosphoric acid etc.
Dosage nature becomes with following known facts: for example pharmacodynamic profiles of concrete medicine, the pattern and the approach of its administration; Medication person's age, health and body weight; The nature and extent of symptom; The kind of Synergistic treatment; The frequency of treatment; Effect with needs.The per daily dose of expection activeconstituents can be about 0.001 to about 1000 mg/kg body weight, and preferred dosage is about 0.1 to about 30mg/kg.
The combination dosage form per unit that is applicable to administration contains about 1mg to about 100mg activeconstituents.In these medicinal compositionss, activeconstituents generally accounts for about 0.95% (weight) by the total restatement of composition.Activeconstituents can solid dosage for example capsule, tablet and powder, or with liquid dosage form for example elixir, syrup and suspensoid oral administration.It also can the sterile liquid formulation through administered parenterally.
Preparation of the present invention comprises those preparations of suitable per os, nose, part (comprising mouthful cheek and hypogloeeis), rectum, vagina and/or administered parenterally.Can provide the unit dosage of these preparations expediently, and can be by any method preparation of knowing in pharmaceutics field. can become with the host of treatment, concrete mode of administration with the amount of the activeconstituents of the single formulation of carrier substance combined preparation.Can be generally the amount of the inhibitor that reaches curative effect with the amount of the activeconstituents of the single formulation of carrier substance combined preparation.Usually by 100%, the amount of this activeconstituents is about 1% to about 99%, preferred about 5% to about 70%, most preferably from about 10% to about 30%.
Preparing these preparations or method for compositions comprises The compounds of this invention and carrier and chooses any one kind of them or multiple auxiliary agent blended step.Generally speaking, this type of preparation prepares as making product shaping then by solid carrier or both even and thorough mixing with inhibitor of the present invention and liquid vehicle or pulverizing.
Being fit to peroral administration preparation of the present invention can be following form: capsule, cachet, pill, tablet, lozenge (use flavoring matrix, be generally sucrose and gum arabic or tragacanth), the solution or the suspensoid of powder, granule or water or on-aqueous liquid, or oil-in-water or water-in-oil liquid emulsion, or elixir or syrup, or pastille (uses inert base, for example gelatin and glycerine, or sucrose and gum arabic) and/or mouth wash shua etc., the The compounds of this invention of each self-contained predetermined amount is as activeconstituents.Inhibitor of the present invention also can the dense injection of disposable heavy dose (bolus), electuary or patch administration.
Be used for peroral administration solid dosage of the present invention (capsule, tablet, pill, dragee, powder, granule etc.), activeconstituents and one or more pharmaceutically acceptable carriers for example Trisodium Citrate or Lin Suanergai and/or following any carrier mix: (1) weighting agent or supplement are starch, lactose, sucrose, glucose, N.F,USP MANNITOL and/or silicic acid for example; (2) for example carboxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and/or gum arabic of tackiness agent; (3) wetting Agent for Printing Inks, for example glycerine; (4) disintegrating agent, for example agar, lime carbonate, potato or tapioca (flour), alginic acid, some silicate and yellow soda ash; (5) solution retarding agent paraffin for example; (6) absorption enhancer quaternary ammonium compound for example; (7) for example hexadecanol and Zerol of wetting agent; (8) for example kaolin and bentonite clay of absorption agent; (9) lubricant such as talcum powder, calcium stearate, Magnesium Stearate, solid polyethylene glycol, sodium lauryl sulphate and composition thereof; (10) tinting material.In the situation of capsule, tablet and pill, this class medicinal compositions also can comprise buffer reagent.Use the similar solids composition of this type of vehicle such as lactose and high molecular weight polyethylene glycol etc. also to can be used as the weighting agent of filling soft hard gelatin capsule.
Tablet can be suppressed or molded with one or more auxiliary agents by optional.Compressed tablet useful binders (for example gelatin or Vltra tears), lubricant, inert diluent, sanitas, disintegrating agent (for example primojel or croscarmellose sodium), tensio-active agent or dispersion agent preparation.Molded tablet can prepare through the wetting inhibitor powdered mixture of inert liquid diluent by molded in suitable machine.
Can choose wantonly with the solid dosage of tablet of the present invention and other medicinal compositions for example dragee, capsule, pill and granule indentation or with coating material and shell for example other coating material of knowing of enteric-coating material and field of pharmaceutical preparations prepare.They can be to use Vltra tears, other polymer backbone material, liposome and/or the microsphere that the various ratios that need release profiles for example are provided, and provide slowly or the sustained release formulation of activeconstituents wherein.They can filter by for example filter through holding back bacterium, or sterilize by sterilizing agent is added in the aseptic solid composite, this aseptic solid composite can faced with before being dissolved in sterilized water or some other aseptic injection medium.These compositions also can be chosen wantonly and contain opacifying agent, and can be they only or preferentially at the optional composition of GI some part with retardation mode release of active ingredients.Spendable implant compositions example comprises polymer substance and wax.If suitable, activeconstituents can also be the micro-capsule form with one or more above-mentioned vehicle together.
The liquid dosage form of peroral administration The compounds of this invention comprises pharmaceutically acceptable emulsion, micro emulsion, solution, suspension, syrup and elixir.Except that activeconstituents, liquid dosage form can contain this area inert diluent commonly used, for example water or other solvent, solubilizing agent and emulsifying agent, fatty acid ester of ethanol, Virahol, ethyl-carbonate, ethyl acetate, benzylalcohol, phenylformic acid benzyl ester, propylene glycol, 1,3 butylene glycol, oil (especially oleum gossypii seminis, peanut oil, Semen Maydis oil, plumule (germ) oil, sweet oil, Viscotrol C and sesame oil), glycerine, tetrahydrofurfuryl alcohol, polyoxyethylene glycol, anhydro sorbitol and composition thereof for example.
Except that inert diluent, oral compositions also can comprise auxiliary agent for example wetting agent, emulsifying agent and suspension agent, sweeting agent, correctives, tinting material, perfume compound and sanitas.
Except that activity inhibitor of the present invention, suspension can contain suspension agent for example ethoxylation isooctadecanol, polyoxyethylene sorbitol and Isosorbide Dinitrate, Microcrystalline Cellulose, inclined to one side aluminium hydroxide, bentonite, agar, tragacanth and composition thereof.
Can be provided for the suppository formulations of the medicinal compositions of the present invention of rectum or vagina administration, this formulation can be by comprising that with one or more The compounds of this invention and one or more suitable nonirritant excipients or carrier for example theobroma oil, polyoxyethylene glycol, suppository wax or salicylate (ester) are mixed with, it at room temperature is a solid, but be liquid under the body temperature, therefore discharge activity inhibitor at rectum or vaginal canal fusing.
The preparation of the present invention that is applicable to vagina administration comprises vaginal suppository, tampon, creme, gelifying agent, paste, whipping agent or the sprays preparation that contains known suitable carrier in this area.
The formulation that is used for part or transdermal administration The compounds of this invention comprises powder, sprays, ointment, paste, creme, lotion, gelifying agent, solution, patch and inhalation.Under aseptic condition, can be with active compound and pharmaceutically acceptable carrier and any sanitas, buffer reagent or the propellant mixing that may need.
Except that active prenyltransferase inhibitor, ointment, paste, creme and gelifying agent can contain vehicle, for example animal and plant fat, oil, wax, paraffin, starch, tragacanth, derivatived cellulose, polyoxyethylene glycol, silicone, bentonite, silicic acid, talcum powder and zinc oxide or its mixture.
Except that The compounds of this invention, powder and sprays can contain vehicle for example lactose, talcum powder, silicic acid, aluminium hydroxide, Calucium Silicate powder and polyamide powder, or the mixture of these materials.Sprays also can contain propelling agent commonly used, for example chlorofluorocarbon and volatile hydrocarbon for example butane and propane of not replacing.
Transdermal patch also has provides the control advantage that The compounds of this invention discharges in body.Can be by inhibitor of the present invention being dissolved in or being scattered in this type of formulation of preparation in the suitable medium.Also available absorption enhancer increases transdermal drug flux.Can be by rate controlling membranes be provided, or The compounds of this invention is dispersed in the speed of controlling this flow in polymer backbone or the gel.
Ophthalmic preparation, eye ointment, powder, solution etc. are also included within the scope of the present invention.
Be applicable to that the medicinal compositions of the present invention of administered parenterally comprises one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solution, dispersion, suspension or the emulsion of one or more inhibitor of the present invention and applied in any combination, or be the sterilized powder of aseptic injectable solution or dispersion facing with preceding the reformulation, said composition can contain antioxidant, buffer reagent, bacteriostatic agent, make preparation and intended recipient's the isoosmotic solute of blood or suspension agent or thickening material.
Can be used for the suitable water of medicinal compositions of the present invention and examples of non-aqueous carriers and comprise for example sweet oil, injection organic ester ethyl oleate for example of water, ethanol, polyvalent alcohol (for example glycerine, propylene glycol, polyoxyethylene glycol etc.) and suitable mixture thereof, vegetables oil.For example by use coating material for example Yelkin TTS, by keeping in the dispersion essential particle diameter and can keep suitable flowability by the use tensio-active agent,
These compositions also can contain auxiliary agent for example sanitas, wetting agent, emulsifying agent and dispersion agent.For example nipagin esters, trichloro-butyl alcohol, phenol Sorbic Acid etc. guarantee to prevent microbial process can to add various antiseptic-germicides and anti-mycotic agent.May need also that for example sugar, sodium-chlor etc. are included in the composition with isotonic agent.In addition, can for example aluminum monostearate and gelatin prolong the absorption of injectable drug form by the reagent that add to postpone absorbs.
In some cases, for prolonging the curative effect of inhibitor, need slow down absorption subcutaneous or the intramuscularly inhibitor.This can have the liquid suspension realization of relatively poor water miscible crystallization or amorphous substance by use.Thereby the absorption rate of inhibitor depends on its dissolution rate, and dissolution rate depends on crystallite size and crystallized form.Perhaps, realize making the inhibitor form of administered parenterally to postpone to absorb in the oily carrier by inhibitor being dissolved in or being suspended in.
Prepare injection prolonged action preparation form by the microencapsulation skeleton that for example in polylactide-polyglycolide, forms the theme inhibitor at biodegradable polymkeric substance.According to the ratio and the concrete character of using polymkeric substance of medicine and polymkeric substance, the speed of may command drug release.The example of other biodegradable polymers comprises poly-(ortho ester) and poly-(acid anhydride).Also can prepare long acting injection by in liposome or the micro emulsion compatible, capturing medicine with bodily tissue.
When The compounds of this invention gives humans and animals as medicine, can give them separately, or give them as the medicinal compositions of the pharmaceutically acceptable carrier that contains 0.1-99.5% (more preferably 0.5-90%) activeconstituents for example and be used in combination.
But preparation per os of the present invention, parenteral, part or rectum give.They will give by the form that is applicable to every kind of route of administration naturally.For example, they give with the form of tablet or capsule; Give by injection, inhalation, eye wass, ointment, suppository etc.; Give by injection, infusion or suction; By lotion or ointment topical administration; With give by rectum by suppository.Preferred oral administration.
The pattern of administration and topical in phrase used herein " administered parenterally " and the non-intestines of " parenteral gives " expression, usually by injection, indefiniteness comprises in intravenously, intramuscular, intra-arterial, the sheath, in the capsule, interior, intracardiac, the intracutaneous of eye socket, intraperitoneal, under tracheae, subcutaneous, epidermis, under the intraarticular, capsule, under the arachnoid membrane, in the backbone and breastbone inner injection and infusion.
Phrase used herein " whole body administration ", " whole body gives ", " peripherally administered " and " administered peripherally " expression give directly not enter compound, medicine or other material of central nervous system, subcutaneous administration for example, so that it enters patient's whole body, thereby experience metabolism and other similar procedure.
No matter the route of administration of selecting, the CDK inhibitor that uses in this subject methods can use by suitable hydrated form, and/or medicinal compositions of the present invention can be mixed with pharmaceutically acceptable formulation with it by ordinary method well known by persons skilled in the art.
The gelatine capsule agent contains activeconstituents and support powder for example lactose, starch, derivatived cellulose, Magnesium Stearate, stearic acid etc.Available similar thinner prepares compressed tablet.Tablet and capsule can be formed in the slow release product that continuous release medicine was provided in a few hours.Can give compressed tablet sugar coating or film-coat covering any offending taste and to make tablet and air isolated, or wrap casing so that tablet selectivity disintegration in gi tract.Also can be with similar solids composition as the weighting agent of filling soft hard gelatin capsule; In this relation, preferred material also comprises lactose and high molecular weight polyethylene glycol.Preferred preparation is for example Gelseal of sweet oil, Miglyol or Capmul solution or suspension of oil.Can suitably add antioxidant to prevent long-term degradation.
Peroral administration liquid dosage form can contain tinting material and correctives to increase patient's compliance.Generally speaking, water, suitable oil, salt solution, ethanol, D/W and relevant sugar soln, dibasic alcohol for example the mixture of propylene glycol or polyoxyethylene glycol or these materials are suitable carriers of stomach externally used solution.
Can with more than the compound of coming into the open be mixed with the sterile solution of its free form activeconstituents or its salt with physiological buffer or aqua sterilisa to be used for intravenous administration.Can use as needs to contain sugar carrier liquid (for example ringer lactate or other glucose solution), condition is that total sugar degree does not cause unwanted lactic acidosis level.Intravenous administration can carry out by the dense notes of disposable heavy dose (preferred every day several times) or by continuous infusion in the time that continues.The total dose of dense notes of preferred disposable heavy dose or infusion changes according to the patient's body condition basically; Generally speaking, they are generally about 25mg/kg to about 250mg/kg.
The solution of preferred administered parenterally contains water-soluble salt, the suitable stabilizers of activeconstituents and takes the circumstances into consideration to contain buffer substance.For example sodium bisulfite, S-WAT or xitix are suitable stabilizers to the antioxidant of Ying Yonging alone or in combination.Also use citric acid and salt thereof and EDTA sodium.In addition, the stomach externally used solution can contain sanitas, for example benzalkonium chloride, Tegosept M or propylben, trichloro-butyl alcohol.At this area canonical reference teaching material Remington ' sPharmaceutical Sciences, the 18th edition, Mack Publishing Company, Easton, PA has the description to suitable pharmaceutical carrier in 1990, and its content is bonded to herein by reference.
Use preparation, solution and other preparation of the compound of Table A, B and/or C to apply for that the method for describing among WO 03/033499 and/or the WO 04/092139 makes according to PCT, instruction wherein is incorporated herein by reference.
Iv. treatment is used
Because cdks generally plays a crucial role in regulating cell proliferation, compound disclosed herein can be used as reversible cytostatics, can be used for treating any lysis excess proliferative disease for example that is characterized as abnormal cell proliferation, comprise cancer, benign prostatic hyperplasia, family's adenoma characteristic of disease polyposis, neurofibromatosis, psoriatic, fungi infestation, endotoxin induction is repaiied gram, hypertrophic scar forms, inflammatory bowel, transplant rejection, the vascular smooth muscle cell proliferation relevant with atherosclerosis, psoriatic, pulmonary fibrosis, sacroiliitis, glomerulonephritis, restenosis and other narrow and restenosis in operation back after angioplasty or the vascular surgery.See for example U.S. Patent number 6,114,365 and 6,107,305.
Expect that compound disclosed herein can effectively treat proliferative or excess proliferative disease for example cancer, autoimmune disorder, virus disease, fungal disease, neurodegenerative disease and cardiovascular disorder.
More particularly, compound disclosed herein can effectively be treated various cancers and be included, but is not limited to following cancer: cancer comprises bladder cancer, mammary cancer, colorectal carcinoma, kidney, liver cancer, the lung cancer that comprises small cell lung cancer, esophagus cancer, carcinoma of gallbladder, ovarian cancer, carcinoma of the pancreas, cancer of the stomach, cervical cancer, thyroid carcinoma, prostate cancer and comprises the skin carcinoma of squamous cell cancer; Hemopoietic system lymph tumour comprises leukemia, acute lymphoblastic leukemia, acute lymphocytoblast leukemia, B-cell lymphoma, T-cell lymphoma, hodgkin's lymphoma, non Hodgkin lymphoma, trichoblast lymphoma and Burkett lymphomas; Hemopoietic system marrow tumour comprises acute and chronic lymphocytic leukemia, myelodysplastic syndrome and promyelocytic leukemia; Be derived from the tumour of a matter, comprise fibrosarcoma and rhabdosarcoma; Maincenter and peripheral nervous system tumour comprise astrocytoma, neuroblastoma, neurospongioma and schwannoma; With other tumour, comprise melanoma, spermocytoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi sarcoma.
Participate in tau protein phosphorylation (J.Biochem, 117,741-749 (1995)) owing to find cdk5 recently, point out compound disclosed herein also can effectively treat presenile dementia.
Apoptosis can be induced or suppress to compound disclosed herein.In the various human disease, apoptosis is replied unusually.Can effectively treat cancer (including but not limited to above-mentioned those kinds) as the compound described herein of apoptosis conditioning agent, virus infection (includes but not limited to simplexvirus, poxvirus, Ai-Ba Er Shi (Epstein-Barr) virus, Xin Peisi (Sindbis) virus and adenovirus), prevention AIDS develops in the infected by HIV individuality, autoimmune disorder (includes but not limited to systemic lupus erythematosus, the glomerulonephritis of autoimmunization mediation, rheumatoid arthritis, psoriatic, inflammatory bowel and autoimmune diabetes), neurodegenerative disease (includes but not limited to presenile dementia, the dementia relevant with AIDS, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, Duchenne-Arandisease and cerebellar degeneration), myelodysplastic syndrome, aplastic anemia, the ischemia injury relevant with myocardial infarction, apoplexy and reperfusion injury, irregular pulse, atherosclerosis, toxin causes or alcohol dependency hepatopathy, hemopathy (including but not limited to chronic anaemia and aplastic anemia), Skeletal system degenerative disease (including but not limited to osteoporosis and sacroiliitis) acetylsalicylic acid allergic sinusitis, cystic fibrosis, multiple sclerosis, ephrosis and carcinomas pain.
As adjustable ganglion cell RNA of the compound disclosed herein of cdks inhibitor and DNA synthetic level.Therefore, these medicines can effectively be treated virus infection (including but not limited to HIV, human papillomavirus, simplexvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus).
Compound disclosed herein also can effective chemical preventing cancer.Chemoprophylaxis is defined as by blocking-up and causes the mutagenesis incident, or cell development or suppress tumor recurrence and suppress the invasive cancer development before the deterioration that has suffered damage by blocking-up.
Compound disclosed herein also can effectively suppress tumor-blood-vessel growth and transfer.
Compound disclosed herein also can be used for preventing to follow usually the alopecia of many traditional chemical therapy schemes appearance.For example, CDK inhibitor of the present invention can be used for suppressing the cell proliferation in the hair follicle, and therefore, they are avoided with the proliferative cell is the attack of the cell toxicity medicament of target.
Compound disclosed herein also can be used as the inhibitor of following other protein kinase: for example protein kinase C, her2, raf 1, MEK1, map kinase, EGF acceptor, pdgf receptor, IGF acceptor, PI3 kinases, wee l kinases, Src, Abl, therefore can effectively treat and other protein kinase diseases associated.
The compounds of this invention also can with known anticancer therapy radiotherapy or unite use (simultaneously or sequential giving) for example with following cytostatics or cell toxicity medicament: such as but not limited to DNA drugs with function for example cis-platinum or Dx; The topoisomerase II inhibitor is Etoposide for example; The topoisomerase I inhibitor is CPT-11 or Hycamtin for example; The tubulin drugs with function is taxol, docetaxel or epothilones for example; Hormonal medicaments is tamoxifen for example; Thymidylate synthetase inhibitor is 5 FU 5 fluorouracil for example; With antimetabolite methotrexate for example.In this type of combination medicine, compound of the present invention and preparation can effectively prevent or reduce the alopecia incidence, and it is normally caused by radiotherapy or chemotherapy.
If be made into fixed dosage, this type of combined prod uses The compounds of this invention and other active medicine or at the dosage range internal therapy of its approval in following dosage range.For example, find the collaborative known cell toxicity medicament cell death inducing (J.Cell Sci., 108,2897 (1995)) of cdc2 inhibitor olomucine.When combination preparation is improper, also can sequentially give described compound and known anticancer drugs or cell toxicity medicament herein.The present invention is not subjected to the qualification of order of administration; Can before or after giving known anticancer drugs or cell toxicity medicament, give described compound herein.For example, the cytotoxic activity of the kinase inhibitor flavopiridol of cyclin dependent is subjected to influence with the order of administration of cancer therapy drug.Cancer Research,57,3375(1997)。
All embodiments described herein are applicable to all different aspects of the present invention.In due course, any described embodiment can freely merge with one or more other such embodiment.Each embodiment of The compounds of this invention, be suitable for each embodiment of the illness of such compounds for treating and use each embodiment of the methods of treatment of The compounds of this invention freely to merge each other.
Specific embodiments of the present invention has been described herein in more detail.Yet these are embodiments of illustrative, and should be not interpreted as the restriction of where face in office.
Equivalent
Those skilled in the art will be appreciated that or can only use normal experiment to determine a lot of equivalents of specific embodiments of the present invention as herein described.Such equivalent is included in the scope of claims.Those skilled in the art it should also be appreciated that the embodiment of claim described herein or all of feature make up all within the scope of the present invention.
V. synthetic
The known synthetic method in available following method and synthetic organic chemistry field, or the synthetic The compounds of this invention of its deriving method that those skilled in the art recognize that.Preferable methods includes but not limited to following those methods.Each reference of hereinafter quoting is attached to herein by reference.
The key intermediate of preparation compounds more disclosed herein is pyrazoles amino-nitrile, aminocarboxylic acid amides and amino ester.These intermediates preparation are delivered in chemical literature, at flow process A (A.O.Abdelhamid etc., J.Heterocycl.Chem.1984,21,1049), B (C.C.Cheng and R.K.Robins, J.Org.Chem.1956,21,1240), C (P.Schmidt and J.Druey, Helv.Chem.Acta 1956,39,986) summarized several method in.Also can consult Tominaga etc., J.Heterocycl.Chem.1990,27,775 and PCT application number WO 00/21926 and WO 99/54308.Multiple hydrazine and aldehyde raw material have commercially available, maybe can be by the preparation of standard organic transformation method.Below the substituent A r of Shi Yonging represents aromatic ring, is substituted formation (conform) or is converted into the corresponding aryl substituent of compounds more disclosed herein.Also can use CH by in the presence of alkali 2(CN) 2Handle PrCOCl, use PCl 5Handle the compound that generates, make product and ArNHNH then 2Reaction prepares some compounds.
Flow process A
Figure C200480042035D00441
Flow process B
Figure C200480042035D00451
Flow process C
Figure C200480042035D00452
As shown in flow process D, amino-nitrile II can be converted into pyrazolo of the present invention [3,4-d] pyrimidine.In a word, choose wantonly at suitable solvent for example in the presence of the methylene dichloride, by with suitable alkali for example triethylamine handle aminocarboxylic acid amides acidylate, use formula ArCH subsequently 2The preferred acyl chlorides of COX carboxylic acid halides is handled, and obtains carboxamido nitrile V.Perhaps, by in the presence of suitable alkali and coupling reagent, in suitable solvent, make amino-nitrile II and general formula ArCH 2CO 2The carboxylic acid coupling of H can prepare carboxamido nitrile V.(Klausnew and Bodansky, Synthesis 1972,453-463), one skilled in the art will recognize that the reagent of multiple this coupling of realization summary have been made in the coupling of amine and carboxylic acid.
Flow process D
Figure C200480042035D00461
Can by about 0 ℃ to being up to 100 ℃, in the presence of suitable alkali preferable alloy oxyhydroxide or alcoholate alkali, in solvent preferably water, alcohol or water-alcohol mixture, handle with excess hydrogen peroxide, can finish the conversion to The compounds of this invention with carboxamido nitrile V.
Perhaps, can pass through at dense, preferred 85% H of strong acid 3PO 4In, preferred about 1 hour of heating, V is converted into The compounds of this invention with the carboxamido nitrile.Flow process E represents to prepare the another kind of method of The compounds of this invention.In the preferred low-level chain triacontanol of suitable solvent, preferably under the boiling point of solvent, be the formula ArCH of low alkyl group for example with excessive wherein R 2CO 2The ester of R and the excessive rudimentary alcoholate of alkali preferable alloy are handled aminoacyl imines III, obtain The compounds of this invention.Many aryl acetates have commercially available, maybe can be by at acid catalyst H for example 2SO 4Or the p-TsOH existence down, with excessive alcohol roh, preferably as solvent prepared with ethanol or methyl alcohol in commercially available one step of Arylacetic acids esterification under refluxing.Perhaps, preferably at solvent CH for example 2Cl 2In and adopt for example DMAP of catalyzer, can use coupling to try for example DCC.
Flow process E
Figure C200480042035D00462
Toluylic acid can be by the preparation of acid or basic hydrolysis aryl acetonitrile, and aryl acetonitrile can be by preferably for example in DMF, MeOH, EtOH, water, DMSO or its mixture, handling aryl halide preparation with CN-at solvent.More examples of aryl acetate can prepare with aryl carboxylic acid under Arndt-Eistert (Meier and Zeller, Angew.Chem.Int.Ed.Engl.1975,14,32) or relevant homologation condition.
Can by with excessive formula ArCH 2CN nitrile and sodium reaction are converted into The compounds of this invention with formula IV amino ester.
Flow process F
Figure C200480042035D00471
Preferred this reaction solvent-free (neat) heating is carried out.
Pyrazolo [3,4-d] pyrimidone also can further prepare as follows, obtains other compound of the present invention.Can on the Ar group, carry out close electric aromatics substitution reaction, introduce substituting group.That the reaction of this class includes but not limited to is nitrated, acidylate (Friedel-Crafts), halo, alkylation (Friedel-Crafts), chloromethylation, sulfonation and aminomethylation (Mannich reaction).The technician in organic synthesis field is familiar with carrying out the condition of these reactions, is usually directed in the presence of catalyzer, carries out cationoid reaction with aryl or heteroaryl substrate.In the situation of nitrated or Mannich reaction, preferred catalyst is the protonic acid that can be used as solvent, and is wherein electrophile respectively by nitre or amine and the generation of carbonyl component original position.For other close electric aromatics substitution reaction, preferred catalyzer is Lewis acid, includes but not limited to FeX 3, AlX 3And ZnX 2, wherein X is a halogen.
Can be by reacting, with the amino compound derivingization that has of above preparation with electrophile carboxylic acid halides, acid anhydrides, isocyanic ester, chloro-formic ester, sulfonic acid halide, alkylogen, lactone or the ester of including but not limited to.The technician in organic synthesis field is familiar with the operational condition of these addition reactions, be usually directed to preferably in solution, 0 ℃ to room temperature, add on the nucleophilic reagent electrophile.May need to add alkali.The product that is noted that these reactions also can be gone up and some electrophile reaction at the nitrogen (N5) of pyrimidone.Compare with the anilino or the aliphatic group of needs, gained functional group (acid amides, carbamate etc.) to the less stable of alkaline hydrolysis, can be reverted to the pyrimidone that has H on the N5 by dissociating.
Make compound with amido and reagent for example halogen acyl halide, α, beta-unsaturated acyl halogen, or the reaction of halo sulfonic acid halide obtain intermediate, this intermediate can with nucleophilic reagent for example uncle or secondary amine, diamines, alkoxide, amino alcohol or thiol reactant.
Can include but not limited to amine and alcohol reaction by activation with nucleophilic reagent, obtain acid amides and ester respectively, the compound derivingization with carboxyl of above preparation.The coupled reaction of amine and carboxylic acid and carbodiimide summary (Klausnew and Bodansky have been made, Synthesis1972,453-463), one skilled in the art will recognize that and realize that this reaction can have multiple other reagent and (the Greenand Wuts of needs may be arranged the blocking group of sheltering active function groups, " Protective Groups in Organic Synthesis " second edition, John Wiley ﹠amp; Sons, 1991).Above-mentioned for prepare the description of ester with acid.Available suitable hydride reducer is reduced to amine and alcohol with these acid amides and ester.
Can be preferably in the presence of alkali, by with phosgene or the activation of phosgene equivalent, be converted into the Electron Affinities material (Tetrahedron:Asymmetry 1995,61,745; J.Org.Chem.1994,59,1937), include but not limited to amine, pure and mild sulfuryl amine reaction with nucleophilic reagent then, obtain urea, carbamate and sulfonylurea respectively, with the amino compound derivingization that has of above preparation.The operational condition that the technician in organic synthesis field is familiar with these reactions with handle the relevant danger of phosgene photoreactive gas equivalent, therefore should take all suitable safeguard procedures.
Other conversion that the preparation The compounds of this invention may need comprises reductone, aldehyde, ester, acid, reductive amination (the J.Seyden-Penne of acid amides or use aluminium and borohydride reagents, " Reductions by the Alumino and Borohydrides in OrganicSynthesis " VCH Publishers, Inc., 1991) and the following group of oxidation, include but not limited to alcohol, aldehyde, alkene, thioether, sulfoxide and heteroaryl (Milos Hudlicky, " Oxidationsin Organic Chemistry " American Chemical Society, 1990).
Can finish reduction functional group for example alkene, alkynes, nitrogen, nitro or cyano group by catalytic hydrogenation or by dissolving metal reduction.Can be by include but not limited to the displacement of CN-, amine, alkoxide, mercaptan or carbanion with nucleophilic reagent, finish further preparation and contain intermediate with the electric combining site of The compounds of this invention parent.Other compound of the present invention also can pass through with suitable boric acid or stannane coupling aryl halide or triflate preparation (Stille, J.K., Angew.Chem.Int.Ed.Engl.1986,25,508; Suzulki, A.Pure Appl.Chem.1985,57,1749).By reacting with nucleophilic reagent, obtain secondary alcohol, can be with the further derivatize of the compound with carbonyl of above preparation.This type of nucleophilic reagent includes but not limited to Grignard reagent (Grignardreagent), lithium alkylide, thiazolinyl lithium and alkynyl lithium reagent, and allyl group stannane, silane etc.Also can be by resetting for example Beckmann (Gawley, Org.React.1988,35,1) or the further compound for preparing by above-mentioned preparation of other rearrangement.
More than Zhi Bei compound also can prepare organic-magnesium or the organolithium material is finished preparation (Beak and Meyers, Acc.Chem.Res.1986,19,356-363 by direct metallized; Beak and Snieckus, Acc.Chem.Res.1982,15,306-312; Katritzky, Lam and Sengupta, Prog Heterocycl.Chem.1989,11,1-29) or by lithium-halogen exchange with aryl halide finish preparation (Parham and Bradsher, Acc.Chem.Res.1982,15,300-305).
The method of preparation formula II disclosed herein, IIa compound and some other compound is provided in flow process 1, and can be used for preparing The compounds of this invention.Substituting group Z, R 5, R 6And R 7The substituting group of describing among the representative formula II, or representative can be converted into those substituent substituting groups with standard organic transformation method.The blocking group that the P representative is suitable.The example of blocking group comprises silyl ether, the acetal of aldehyde and the ketal of ketone of carboxylicesters, alcohol.Existing summary (Greeue, T.W. to the blocking group chemical field; Wuts, P.G.M.Protective Groups in OrganicSynthesis, the 2nd edition; Wiley:New York, 1991).Is amine with catalytic hydrogenation with the nitroreduction of nitrophthalic acid dimethyl ester.With diacetyl oxide and pyridine base with this aniline acidylate.Improving under the temperature, in the suitable solvent, handling the ethanamide 2 that obtains and the mixture of methyl phenyl ketone, obtaining the triketone 3 that needs with highly basic.Those skilled in the art are known as Kilgore etc., Industrial and Engineering Chemistry 34:494-497, other method of the triketone of preparation described in 1946.Improving under the temperature, in the suitable solvent, handle this triketone with hydrazine, obtain indeno [1,2-c] pyrazolone loop systems.
Those skilled in the art are known to Lemke etc., J.Heterocyclic Chem.19:1335-1340,1982; Mosher and Soeder, J.Heterocyclic Chem.8:855-59,1971; Hrnciar and Svanygova, Collect.Czech.Chem.Commun.59:2734-40 prepares other method of indeno [1,2-c] pyrazolone described in 1994.By in suitable solvent, with the strong acid heating, make the acid amides removal of acylation, obtain aniline 4.This aniline is used the acyl chlorides acidylate under standard conditions, in the suitable solvent, obtain the product 5 that needs.
Flow process 1
Figure C200480042035D00501
The another kind of method of preparation The compounds of this invention is seen flow process 2.Use method known to those skilled in the art, available strong acid makes triketone 3 intermediate deacylations, uses suitable acyl chlorides acidylate again.Use the same terms in the aforementioned flow process 1 subsequently, triketone 6 can be converted into indeno [1,2-c] pyrazolone loop systems.
Flow process 2
Figure C200480042035D00511
The another kind of method of triketone 6 is pressed Rotberg and Oshkaya, Zh.Organ.Khim.8:84-87,1972 in the preparation flow 2; Described in the Zh.Organ.Khim.9:2548 2550,1973, with 1,3-diketone 6a and the condensation of 3-nitrophthalic acid acid anhydride.When commercially available, those skilled in the art can easily prepare 1, the 3-diketone by with essential methyl ketone acetylize or trifluoroacetylation when no.Available several different methods comprises catalytic hydrogenation, handles with zinc or iron under acidic conditions or with for example V-Brite B or the tin protochloride processing of other reductive agent, finishes making the nitro-derivative that obtains be reduced to aniline 6b.Can pass through acidylate subsequently, handle by aforementioned flow process 2 usefulness hydrazines then, make aniline 6c be converted into indeno of the present invention [1,2-c] pyrazolone.
The another kind of method of preparation indeno [1,2-c] pyrazolone loop systems is seen flow process 3.Solubilizing agent does not make the reaction of dimethylhydrazine and 3-acetylpyridine, obtains hydrazone 7.Handle this product by similar fashion described in the flow process 1, obtain the intermediate 8 that needs.
Flow process 3
Figure C200480042035D00521
Perhaps, amino morpholine of available activatory acidylate N-or piperazine ring for example carboxylamine nitrophenyl ester handle 6b.The known Rappoport that presses of those skilled in the art, J.Org.Chem.49:2948-2953, other method of the similar intermediate of preparation described in 1984.By with this intermediate of subsequent treatment of similar fashion described in the flow process 1.
W is the general synthetic route of oxygen although aforementioned flow process has been described wherein, and according to this general routes outlined content, those skilled in the art can envision and implement synthetic wherein W and not be other compound of the present invention of oxygen.For example, wherein W is selected from S, S (O 2), C (=O), C (=S), CH 2And NR ".
In the process of describing exemplary subsequently, further feature of the present invention will be apparent, provide these exemplary for illustrating the present invention, and the present invention is not limited by it.
V. embodiment
The compound of Table A, B and C can be applied for coming shown in WO 03/033499 and/or the WO04/092139 synthetic as PCT, and its instruction is incorporated herein by reference.
Mensuration scheme and result
Comprise the following biological activity and the effectiveness of those measuring method proof The compounds of this invention in greater detail by one or more mensuration:
Measure the inhibition (the results are shown in Figure 1 and table 2) of 1. usefulness iodate, third ingot and BrdU mensuration The compounds of this invention cell cycle process.
Measure the 2. 60 kinds of clone viabilities of the wide region that is derived from various people's tumours that are exposed to NCI series (panel) representative of The compounds of this invention descend (the results are shown in Table 3).
Measure 3. and producing during the clone cell survival measures, the irreversible effect of The compounds of this invention pair cell (the results are shown in Table 3, Fig. 2 and Fig. 3).
Measure 4. usefulness fluorexon (Calcein) AM and measure HCT-116 and the IMR90 cell survival decline (the results are shown in Table 3 and 6) that assessment is exposed to The compounds of this invention.
Measure 5. viabilities that are exposed to the stagnation tumour cell of The compounds of this invention and suppressed, stagnate normal cell and then be not subjected to suppress (the results are shown in Table 4 and Fig. 4).
Measure the inhibition activity (the results are shown in Table 5 and 6) of 6. The compounds of this invention in some kinases biochemical measurement.
Measure the activity (the results are shown in Figure 5,6,7 and 8) of 7. compounds in the xenotransplantation tumor model.
Measure the avidity (the results are shown in Figure 9) of 8. compounds and some target protein.
Measure the antiviral activity (the results are shown in Table 7) of 9. The compounds of this invention.
Measure 1: with iodate third ingot and BrdU analysis of cells cycle
G1, S and G2/M phase cell percentage in the cell cycle are measured by dyeing to DNA with iodate third ingot, and by flow cytometer with 2N or 4N DNA quantitative determination of complement cell count.Change with the cell distribution of this method evaluation corresponding to the cell cycle that is exposed to the Cdk inhibitor.
The iodate third ingot cell dyeing method
In T-25 flask, in the presence of test compounds, cultivate 3 batches of HCT-116 cells (100,000 cells/criticize) according to following table 1.Analyzed at 24,48 and 72 hours.Collect sticking parietal cell by trysinization, merge at the showy cell of Falcon 12 * 75 fluidic cell Guan Zhongyu, by centrifugal results.Incline substratum in the cell precipitation adds 100 μ l PI staining agents, with cell at 37 ℃ of incubation 20-25 minutes.The preferred cell counting is no more than 2 * 10 6-4 * 10 6/ ml.Then equal-volume (100 μ l) PI salt is added in the cell, then with it at 4 ℃ of following incubation 1-2 hours.Last Becton Dickinson FACScan flow cytometry analysis staining cell.The sample lucifuge.Fig. 1 shows when being exposed to compound A-13 7, and apoptosis and endoreduplication proof cell are stuck in G1 and G2 phase latter stage.Find that some other compound of the present invention comprises that similar results appears in compound B-11 6.
Mensuration is in conjunction with the BrdU of DNA
This method is measured when cell developed through the cell cycle S phase, in conjunction with the new cell per-cent of the nucleotide analog BrdU of synthetic DNA.The Cdk inhibitor developed the S phase and the influence of dna replication dna in conjunction with measuring with suppressing BrdU.
The method of BrdU mark
3 batches of HCT-116 cells of inoculation in the T25 bottle (100,000 cells/batch), make it with above test compounds incubation.Analyzed at 24,48 and 72 hours.BrdU is joined in each T-25 bottle, and storing solution is from 10mg/ml to 10 μ M final concentration, under 37 ℃, again with cell incubation 16-18 hour.Scheme (BrdU Flow kit, BD-Pharmingen catalog number 2354KK) by manufacturers is prepared as follows the cell that is used for flow cytometry analysis then:
Results from the T25 bottle (sticking wall and showy) cell, the same then Falcon 12 * 75 fluidic cell pipes that directly add add 100 μ l Cytofix/Cytoperm damping fluids subsequently and fix, change thoroughly (30 minutes, room temperature).Use 1ml Perm lavation buffer solution washed cell then, with the cell precipitation resuspending in 100 μ l Cytoperm Plus damping fluids, incubation on ice 10 minutes.Cell with the washing of 1ml Perm lavation buffer solution, at room temperature, repeated to solidify 10 minutes in 100 μ lCytofix/Cyto Perm damping fluids more then.Cell washs with 1ml Perm lavation buffer solution then.Next step carried out washing step again with 1ml Perm lavation buffer solution to expose bonded BrdU in 1 hour subsequently with 100 μ l DNA enzymes processing cell under 37 ℃.Prove with α-BrdU-FITC antibody (the antibody Perm lavation buffer solution diluents of 50 μ l 1:50) and to have bonded BrdU.The cell lucifuge, at room temperature incubation 20-30 minute.Behind the incubation, with 1ml Perm lavation buffer solution washed cell, resuspending is in the PBS solution of 300 μ l 2%FBS, and the upflowing cell instrument is analyzed.The result lists to suppress 50%BrdU bonded compound concentration (μ M) in table 2.
Measure 2: estimate of the inhibition of Cdk inhibitor to NCI series human tumor cell line
The evaluation of with 60 kinds of clone series compound being carried out in state-run cancer research institute provide a large amount of in relevant kinds of tumors kind and genetic background the information of usefulness.Comprise the clone that is derived from leukemia, melanoma, lung cancer, colorectal carcinoma, the cancer of the brain, ovarian cancer, mammary cancer, prostate cancer and kidney in this series.This serial purposes provides the measurement compound and transforms the usefulness that takes place in the relevant gene in the alternative cell many with tumour, and these genes comprise p53, Her2/Neu and relate to metabolic those genes and cause multiple chemical sproof those genes.The activity of the data evaluation compound of these clones that available employing following proposal obtains.
The result that NCI series is measured lists (NCI series) in table 3, with two kinds of tolerance representatives that data can be provided: (a) with IC50 (μ M) less than except the 10nM, calculate full cell series average-curve median (Mean-Graph Mid-point)-average IC50, equal 10nM in this evaluation; (b) compound suppresses the active IC50 (μ M) of Zorubicin resistant cell line (ADR-res).
Other compound of the present invention shows following activity in NCI measures: (i) compound A-13 7: the IC50<100 μ M of average-curve median<50nM and the growth of inhibition ADR-res cell; (ii) compound B-11 6: the IC50<10 μ M of average-curve median<50nM and the growth of inhibition ADR-res cell.
External cancer screening method
Cell is grown in RPMI-1640 10% FCS, inoculate on 96 hole microtiter plates, inoculum density is 5,000 to 40,000 cells/well.At 37 ℃, 5%CO 2Down, with plate incubation 24 hours.Preparation contains twice needs the substratum of the compound of final concentration (crossing over 5 dosage of 4 orders of magnitude (log)), adds 100 μ l in each hole that fills 100 μ l substratum and cell, obtains the final concentration that needs.And then with each plate incubation 48 hours.
Influence with sulfo group rhodamine (Sulforhodamine) B (SRB) the test determination compound pair cell viability of measuring total protein.With cold TCA fixed cell to final concentration 10%, under 4 ℃, incubation 60 minutes.Abandoning supernatant, each plate wash with water 5 times, dry.In each hole, add 4% (w/v) SRB/1% acetic acid solution, at room temperature with each plate incubation 10 minutes.Each plate dries with 1% acetate washing 5 times.With 10mM trizma alkali dissolution bonded dyestuff, on card reader, the 515nM place reads absorption value.
Measure 3: use the HCT-116 cell to produce the scheme that the clone cell survival is measured
With the compound concentration that causes the viability irreversible loss behind this measuring method mensuration exposure certain hour.Usually, cellular exposure in compound 1,2 or 5 days, is transferred in the growth medium of no compound then.How in the future incubation adds up the colony number that reclaims continuously in the substratum of no compound, estimation survivaling cell number.
This survival measurement result to all cpds of the present invention is listed with the compound concentration (IC50) (μ M) that suppresses the recovery of 50% colony in table 3 (producing the clone).Fig. 2 represents the time-histories relation of the irreversible inhibition of cytoactive and this inhibition of 7 pairs of HCT-116 cells of compound A-13, is exposed to compound 24 hours, its IC50<50nM.In same measured, compound B-11 6 shows its IC50<100nM, and 30 to 60min, IC50 reaches 100nM (Fig. 3).
Measurement is exposed to the method for cell survival behind the compound
Substratum (RPMI-1640,10% FCS, penicillin/streptomycin) is preheated to 37 ℃ in water-bath.At 37 ℃, 5% CO 2Down, the incubation cell also makes its growth.Reclaim the cell that divides in the cachamalplate by trysinization, count with hematimeter.In 15cm tissue culture ware, 25ml substratum, inoculation 1 * 10 4Individual cell.Every kind of test compounds is provided with 14 plates, is incubated overnight under 37 ℃.With substratum diluted chemical compound is become 7 concentration, with the substratum in the substratum replacement cell that contains test compounds.The testing compound of each concentration is provided with the control board of two plates and two no compounds.The same plate incubation 24,48 or 74 hours is removed substratum, replace, with plate incubation 7 days, wash again with PBS with fresh culture.Colony, is counted with distilled water wash twice with crystal violet solution (0.4% Viola crystallina, 20% ethanol) dyeing 5 minutes.
Measure 4: do not exist serum protein and serum protein to exist down, give birth to fluorexon AM Deposit power evaluation of measuring Cdk inhibitor
Measure the effectiveness that (molecular probe) determines to be lost by cell survival the Cdk inhibitor of measurement with fluorexon AM.Fluorexon AM is the substrate of born of the same parents' lactonase, and it only divides in viable cell, produces available fluorescent plate and reads quantitative fluorescence-causing substance.This fluorescent signal and viable count are proportional, and the therefore corresponding cell signal loss that is exposed to the Cdk inhibitor is lost relevant with viability.This mensuration can be distinguished cell cycle arrest from viability loss, wherein cell may be still alively, therefore is fit to very much estimate the Cdk inhibitor.In this mensuration, effective cell poisons the remarkable loss that compound can cause cell survival.
In human colorectal cancer clone, HCT-116, normal people inoblast, IMR90, measure cell IC 50The IC that protein is regulated 50Also in HCT-116, measure.
The result that this viability is measured lists (HCT-116 (viability/protein is regulated) and IMR-90) in table 3.Other compound of the present invention sees Table 6 to the mensuration of HCT-116 cell survival IC50 (μ M, nonprotein is regulated).
Measure the same to the similar cell survival of other clone.The IC50 (μ M) that finds other compound of the present invention is: (i) compound A-13 7:HCT-116 (<50nM), HCT-116 protein regulate (<500nM), A2780 (<10nM), IMR90 (<50nM); (ii) compound B-11 6:HCT-116 (<10nM), HCT-116 protein regulate (<500nM), A2780 (<10nM), IMR90 (<100nM).Fluorexon AM viability is measured scheme.
Reclaim HCT-116 or the IMR90 cell that divides in the cachamalplate by trysinization, 1,000 or 4,000 cell is seeded in respectively in the 24 hole wares, at 37 ℃, 5% CO 2Under be incubated overnight.In RPMI-1640,10% FCS, cultivate the HCT-116 cell, and in α-minimum essential medium, 10% FCS, cultivate the IMR90 cell.After the incubation that spends the night made adhesion, the substratum in each hole of sucking-off added and to contain 0 to 250nM concentration, crosses over the substratum of the test compounds of 7 dosage altogether.Plate is put into incubator once more, cultivate 72 hours (3 days) again.The used substratum of IC50 of measuring the protein adjusting is that RPMI-1640,10% FCS add acid α-glycoprotein (Sigma G-9885) of 1mg/ml and 45mg/ml human serum albumin (SigmaA3782).After 72 hours, cell will pay special attention to remove all residual buffer liquid with 1 * PBS washed twice with the test compounds incubation.
50 μ g fluorexon sample aliquot (molecular probe catalog number C3100) are dissolved in 50 μ l DMSO prepare 5 μ M fluorexon AM solution.Fluorexon dissolves back (following 10 minutes of room temperature) fully, and it is diluted to 10ml PBS.Fluorexon/PBS (0.5ml) is added each hole.Under 37 ℃ (lucifuge),, read at fluorescent plate and to read fluorescent signal (exciting 485/20, emission 530/25) on the meter plate incubation 75 minutes.
Measure 5: stagnate raji cell assay Raji
The active cell that promotes of kinases (Cdk) that needs cyclin dependent is by the asynchronous process of cell division cycle.In substratum, normal non-transformed cell propagation needs the existence of somatomedin, removes it by removing serum deprivation, causes the Cdk loss of activity, and the result works as cell and enters G stationary phase 0The time, withdraw from the cell cycle.Therefore, according to the mechanistic view of life viewpoint but not bound by theory, compare with the corresponding cell of its conversion, the effectiveness of Cdk inhibitor in stagnating normal cell should significantly reduce.
Stagnating normal cell (IMR90) and stagnating the viability measurement result of carrying out on the tumour cell (HT-116) and in following table 4, list with some compound of the present invention.Fig. 4 represents the active increase of the inhibition of compound A-13 7 pairs of stagnations normal cell (IMR90) and tumour cell (HT-116) viability.Find 7 couples of IC50<50nM that stagnate the HCT-116 cell of compound A-13, and to stagnating the IC50 of IMR90 cell 10 μ M.Compound B-11 6 confirms the IC50<50nM to stagnation HCT-116 cell, and to stagnating the IC50 of IMR90 cell 10 μ M.
Come assessing compound to render a service by serum starvation stagnation HCT-116 and IMR90 cell
For each testing compound concentration, the HCT-116 cell is triplicate, in RPMI 1640 substratum that contain 10% foetal calf serum, 24 hole wares, to inoculate, inoculum density is 200 or 2,000 cells/well, at 37 ℃, 5% CO 2Under be incubated overnight.Remove the substratum that contains in 2, the 000 cells/well plates, cell with the serum free medium washing once adds the 1m1 serum free medium in the cell.To contain serum under existing cell and do not have the plate incubation 6 days again of the cell of serum.
For each compound concentration to be measured, the IMR90 cell is triplicate, in containing the MEM-α substratum of 10% foetal calf serum, 24 hole wares, to inoculate, inoculum density is 2,000 or 20,000 cells/well, at 37 ℃, 5% CO 2Under be incubated overnight.Remove the substratum of 20,000 cells/well wares, cell with the serum free medium washing once adds serum free medium in the cell.To contain serum under existing cell and do not have the plate incubation 3 days again of the cell of serum.
Estimate the cell cycle arrest of HCT-116 and IMR90 cell by serum starvation
For cell after guaranteeing to remove serum deprivation breaks away from the cell cycle really, in each experiment, measure the percentage of representative by the BrdU positive cell of those processes of S phase.For this experiment, with only in viable cell the fluorogenic substrate SNARF-1 of activated born of the same parents' lactonase estimate cell survival simultaneously.By flow cytometer BrdU combination and SNARF-1 division are estimated together, on unicellular basis, provided stagnating the evaluation of cell survival.For carrying out this analysis, make cell dyeing as follows with SNARF-1, prepare above-mentioned BrdU then in conjunction with mensuration.
HCT-116 and IMR90 cell are seeded in containing in the blood serum medium (being respectively RPMI-1640 or MEM-α with 10% FCS) of T25 bottle by following density.Grow after 24 hours, remove substratum, behind the washed cell, add serum free medium.
5,000 cells of HCT-116+FCS
100,000 cells of HCT-116-FCS
100,000 cells of IMR90+FCS
200,000 cells of IMR90-FCS
Make IMR90 cell regeneration long 3 days, made HCT-116 cell regeneration long 6 days, use the BrdU burst process then.At room temperature, 50 μ g SNARF-1 (molecular probe catalog number C1272) sample aliquot is dissolved in 50 μ l DMSO, kept 10 minutes, be diluted to 10mlPBS then.With SNARF-1 further dilution be 1:64,000, in each cell pipe, add 200 μ l then, cell is in the presence of serum or serum-free culture, and with BrdU burst process 20 hours in the pipe.With cell 37 ℃ of following incubations 30 minutes, then with 3ml PBS washing.
Fix these cells then, prepare by above-mentioned measurement BrdU combination.On the FACScan flow cytometer, measure live (FL-2) and the BrdU positive (FL-1) cell percentage.
Evaluation is exposed to the viability of stagnating HCT-116 and IMR90 cell behind the The compounds of this invention
At the existence of compound, 37 ℃ of 5% CO 2Down, cell incubation 72 hours (3 days) is as follows, measure the effectiveness of compound to cycle and stagnation cell.With cycle and stagnation HCT-116 cell and 6 the dosage series of cycle IMR90 cellular exposure in 5 to 250nM scopes.For stagnating normal cell, dosage range increases to 50nM to 25 μ M, and the activity of compensation expection reduces.
Be exposed to the influence of 72 hours pair cell viabilities of compound with fluorexon AM evaluation of measuring.Fluorexon AM is the fluorogenic substrate of activated born of the same parents' lactonase in viable cell only.Therefore this substrate division provides and measures and the proportional viability of cell count.
By being dissolved in 50 μ lDMSO, 50 μ g sample aliquot (molecular probe catalog number C3100) prepare fluorexon AM storing solution.At room temperature, should manage incubation about 10 minutes, guarantee that fluorexon dissolved fully.Fluorexon is diluted among the 10ml PBS, prepares whole solution, this solution is wanted lucifuge.
With the substratum sucking-off in the cell, use 1ml PBS washed cell twice then, thoroughly remove PBS in the cell by sucking-off.With transfer pipet 0.5ml fluorexon/PBS solution is moved in each hole.Plate at 37 ℃ of following incubations 75 minutes (lucifuge), is read (exciting 485/20, emission 530/25) and gone up reading at fluorescent plate.
Measure 6: biochemical kinase inhibition is measured
Enzyme: the Cdc2/ cell periodic protein B is available from market.Cdk2/his-cyclin E Short-termUse the Sf9 cell expressing.Cdk2/ cyclin A, cdk4/ cyclin D1 and cdk6/ cyclin D2 use the Sf9 cell expressing.Protein kinase A (catalytic subunit is from OX-heart) and protein kinase C (the mixing isozyme of rat brain) are available from market.
Substrate: histone h1 is available from market.GST-Rb is and Rb protein 37 9-928 residue N-terminal condensed glutathione S-transferase.
Measure: by measure with the TCA precipitation measure bonding histone H1 [γ- 32P] the Triphosaden radioactivity measures Cdc2/ cell periodic protein B activity.Cdc2/ cell periodic protein B kinases and histone h1 are available from market.Measure liquid eventually and contain 50mM Tris.HCl, 10mMMgCl 2, 1mM dithiothreitol (DTT), 50 μ M Triphosadens, 2 μ Ci 32P, 10% methyl-sulphoxide (from compound), pH7.5,20 μ g histone h1s, 6U enzyme, 50 μ L volumes.The compound that adds each concentration of 1nM to 10 μ M.Add enzyme, begin reaction, under 30 ℃, carry out 20min, add 20 μ L reaction terminating liquid (237mM disodium ethylene diamine tetraacetate, 105mM Triphosaden, pH8.0) termination reactions then.Add 35 μ L 70% (w/v) trichloroacetic acid precipitation albumen, (Millipore catches precipitation on Inc.), and is with 25% (w/v) trichoroacetic acid(TCA) that it is wetting at 96 hole glass fibre filter plates.Filter is with 25% (w/v) trichoroacetic acid(TCA) washing 10 times, add 100 μ l scintillators (Microscint 20, PackardInstruments) after, measure combination with scintillation counting 32The amount of P.Amount by there being binding radioactivity down with compound does not contain under the compound amount of binding radioactivity and measures relative reactivity divided by only containing DMSO in the control experiment.Before the calculating, all results deduct containing 50mM EDTA and do not contain the background radiation that records in the compound experiment.Data substitution standard equation is measured compound 50% inhibition concentration (IC50):
P=min+(max-min)(1/(1+(IC50/[I]) s)) (1)
Wherein the P=1-relative reactivity is to suppress relatively, and [I] is compound concentration, and max is respectively minimum and maximum relative inhibition (1 and 0) with min, and s is alleged Hill slope.
Measure Cdk2/ cyclin E, Cdk2/ cyclin A, Cdk4/ cyclin D1 and Cdk6/ cyclin D2 activity with the captive test of Triptide agarose.At Sf9 expressed in insect cells enzyme, be heterodimer, substrate (GST-Rb) is and the Rb retinoblastoma protein 379-928 residue condensed glutathione-S-transferase of expressing in intestinal bacteria (E.coli).This mensuration liquid contains 50mM Tris.HCl, 10mMMgCl 2, 1mM dithiothreitol (DTT), 50 μ M Triphosadens, 2 μ Ci[γ- 33P] Triphosaden, 10% methyl-sulphoxide (from compound), pH7.5,40 μ g GST-Rb and enzymes, 100 μ L volumes.The compound that adds each concentration of 1nM to 10 μ M.Make to be reflected at and carry out 15min under 30 ℃, add 70 μ L reaction terminating liquid (237mM disodium ethylene diamine tetraacetate, 105mM Triphosaden, pH8.0) termination reactions then.By making it combine 110min with Triptide sepharose 4B (Amersham), catch GST-Rb, suspension filters by glass fibre filter.The pearl that is trapped with the phosphate buffered saline (PBS) washing that contains 0.3% (w/v) tween 20 5 times, add 100 μ l scintillators after, measure combination with scintillation counting 33The amount of P.Amount by there being binding radioactivity down with compound does not contain under the compound amount of binding radioactivity and measures relative reactivity divided by only containing DMSO in the control experiment.Before the calculating, all results deduct containing the 50mM disodium ethylene diamine tetraacetate and do not contain the background radiation that records in the compound experiment.By data substitution equation (1) being recorded 50% inhibition concentration (IC of compound 50).
In order to histone h1 is the TCA precipitation test mensuration protein kinase C and the protein kinase A of substrate.For protein kinase A, measure liquid eventually and contain 50mM Tris, 10mM MgCl 2, 1mM dithiothreitol (DTT), pH7.5,12 μ M Triphosadens, 10% (v/v) methyl-sulphoxide (from compound), 20 μ g histone h1s, 2 μ Ci[γ- 32P] Triphosaden, 0.2U protein kinase A, 100 μ l measure liquid.Protein kinase C is measured liquid and is contained 50mM Tris, 10mMMgCl 2, 1mM dithiothreitol (DTT), 0.8mM CaCl 2, pH7.5,5 μ M Triphosadens, 10% (v/v) methyl-sulphoxide (from compound), 20 μ g histone h1s, 2 μ Ci[γ- 32P] Triphosaden, 0.01U protein kinase C, 50 μ l measure liquid.Add enzyme, begin to measure, under 30 ℃, make reaction carry out 10min, add 0.4 volume 237mM disodium ethylene diamine tetraacetate, 105mM Triphosaden, pH8.0 termination reaction then.Add 0.5 volume 75% (w/v) trichoroacetic acid(TCA), protein precipitation from termination reaction is caught by the filtration of 96 hole glass fibre filtration units (Millipore).Filter adds 100 μ l scintillators with 25% (w/v) trichoroacetic acid(TCA) washing 10 times, with scintillation counting measure bonded [ 32P] amount of phosphoric acid ester.By data substitution equation (1) being recorded 50% inhibition concentration (IC of compound 50).
Above measurement result is listed at table 5 and table 6.
Measure 7: the xenotransplantation tumor model
Medicine.Synthetic The compounds of this invention is prepared to be used for intravenously administrable with physiologically acceptable carrier.Obtain CPT-11 (
Figure C200480042035D00611
Pharmacia) medicine prepares with 5% glucose-water (D5W).All preparations are new system weekly, and volume injected is adjusted to body weight (0.2ml/20g mouse).
Mouse/raising.The female nu/nu mouse that derives from Charles River is raised in the little isolation cage of peace and quiet, provided and freely intake and through radiating standard rodent (PurinaPico-Lab Rodent Diet 20).
Measure maximum tolerated dose (MTD).With mouse pairing in 8 ages in week, be one group of marshalling by 5-8 animal, carry out preliminary toxicity research with unknown test compounds.Animal is handled with test compounds intravenous injection every day, continuous 10 days, weighs weekly twice.The clinical sign of any medicine related reactions of running check induced mice.The accepted toxicity of cancer therapy drug in mouse is defined as by NCI: average group weight reduce be no more than 20% and the toxicity mortality ratio of treated animal be no more than 10%.
Standard scheme.With single 1mm 3Tumour segment (tumour brie) s.c. implants in athymic nude mice (6-7 is male or female age in the week) body, or the cell in 5-10 * 106 tissue culture source is implanted association's abdomen.Monitor animal tumor growth twice during beginning weekly, then as the approaching about 100mm of implant 3During pre-determined volume, monitoring every day.When tumor growth during to calculated weight 62-221mg, with the animal pairing, weave into suitable experimental therapy group (8-10 animal/group), begin to treat with test compounds.Positive controls is by control group administration in the past.Calculate tumor weight, weigh in for twice weekly, often observe the ADR that animal occurs.Scheme requires tumor quality to reach the peaceful and comfortable immediately death of any animal of 1000mg.
Measure the length of tumour and width to measure tumour by digital calliper.Tumor weight is estimated with following formula:
Tumor weight (mg)=(w 2X l)/2
W=tumour width wherein, the l=length of tumor is in mm.These are worth the also (mm of available volume unit 3) expression.
Experimental treatment can cause tumor section to disappear (PR) or disappear fully (CR).PR be wherein during studying continuous three days tumour sizes be initial (the 1st day) size 50% or less than initial size but greater than 0.0mg, and when not having measurable tumor epithelial cell in continuous three days, CR appears.Healing is defined as its tumour and is contracted to 0mg, and the animal that keeps this state to finish until experiment.
It is the percentile calculation result of tumour cell of being killed after the decision initial therapy (LCK) that cell kills logarithm (Log cell kill), can be used as quantitative measurment usefulness:
The Log cell kills (LCK)=(T-C)/(3.32) (Td)
Wherein T=treatment group mouse size reaches required mean time of 1000mg, C=control group tumour size reaches the mean time of 1000mg, Td=is in exponential phase of growth, the tumour of estimating with the linear regression analysis of control group tumour semilog growth curve reaches the time of twice, and 3.32=colony increases the required multiple that adds of 1-log10 unit.On behalf of cell, each LCK unit kill 1-log10 unit's (for example 1LCK=90% kills, and 2LCK=99% kills etc.).We think when the LCK of compound value〉1 the time, be equivalent to 90% tumour cell kills, they have remarkable activity.
Tumor growth suppresses (TGI) to be described within a certain period of time, and compounds for treating suppresses the calculation result of tumor growth amount.It is expressed as:
%TGI=100(1-T/C)
Wherein T is specifying the sky, the average tumor size of compounds for treating group, and C is the average tumor size of vehicle Control group on the same day.
Be poisoned to death and be defined as compounds for treating and death that non-disease conditions causes.If the tumour size does not reach 1000mg behind the final compounds for treating, animal is dead in 1 week, can think death due to the toxicity.The relevant death record of non-tumour after this point is arranged, but do not think toxicity death.
By following convention definition tumor regression: if tumor weight alleviate to<initial weight 50% (<50mg), disappearing is defined as part (PR).If at experimental session, tumor weight is reduced to less than measurable weight, disappears to be defined as (CR) fully.Cure and be defined as tumor free animal when the observation period finishes.
Result: Fig. 6 represents the result that several compound of the present invention obtains in HCT116 xenotransplantation tumor model.Fig. 6 represents the result that compound A-13 7 obtains in A2780 xenotransplantation tumor model.Fig. 7 represents the result that compound A-13 7 obtains in PC3 xenotransplantation tumor model.Fig. 8 represents the result that compound B-11 6 obtains in A2780 xenotransplantation tumor model.
Measure 8: measure the avidity between target molecule and the compound
For confirming to be used for to propose the suitability of the specific compound of purposes herein, characterize this compound and its known binding partners (if any) the character that combines come in handy.But this should not be considered as limiting the scope of the invention.
Can measure the avidity of compound and their corresponding binding partners, for example use BIACORE TMThe mensuration system (Biacore AB, Uppsala, SE).Other system that produces similar quantitative result for example Affinity Sensors (Cambridge, UK) Kai Fa those systems will be apparent to those skilled in the art.
In an exemplary process, analysis of compounds R combines with its known binding partners CDK2/ cyclin E's.This analysis under 22 ℃, on BIACORE 2000 SPR-Biosensor, (the albumen quality and grade is carried out in electrophoretic buffer Calbiochem) to contain 20mM HEPES (pH7.4), 150mM NaCl, 1mM DTT and 0.005% polysorbas20.With 10 μ M compound R solution at pH8.0, by the dextran surface coupling of acid amides coupling chemistry with CM5 sensor chip (research grade).Be characterizing compounds R and protein combining of CDK2/ cyclin E for example, PPF is diluted in electrophoretic buffer, obtain 9 different protein concentrations, make each concentration in succession by this sensor surface then, the time of passing through of each concentration is 5min, makes electrophoretic buffer pass through this surface in 5min by identical velocity of flow subsequently.Under 30 μ l/min flow velocitys, measure the association of the CDK2/ cyclin E mixture on CM5-compound R-loading chip and dissociate.After each experiment, before next sample load sample, inject 3M hydrochloric acid continuously by twice
Figure C200480042035D0064154934QIETU
(20 seconds, 30 μ l/min) make chip regeneration.
(Biacore AB, Uppsala SE) analyze data with Bioevaluation software 3.1 editions.Curve is normalized to the background of injecting beginning and obtaining with contrast surface.Measure respectively and associate and the speed of dissociating, or all use Langmuir 1:1 combination model.In order to following Equation for Calculating avidity (K D):
K D=k dissociates/the k association
Available other target protein for example Cdk9, Cdk4 etc. carries out similar operations by above method.For example, HIV and/or AIDS can effectively be treated or prevent to the inhibitor of Cdk9.
Fig. 9 represents the example as a result that obtained by CDK2/ cyclin E and CM5-compound R-loading chips incorporate.K with these data computation DEqual 8,0+/-2,8nM.
Measure 9: antiviral activity
In peripheral blood lymphocytes (PBMCs), measure the usefulness of these compounds, estimate the activity of some compound of the present invention the acute infection cell with low generation (low passage), isolating HIV-1ROJO infection clinically.Use these normal cells can estimate the therapeutic index of these compounds.The fresh PBMCs of two donors is merged, stimulated 48-72 hour with PHA-P.Culturing cell in the presence of IL-2 then causes by mitogenetic signal and to keep cell fission.Infection multiplicity by 0.1 adds virus.After the infection,, estimate usefulness then with cell cultures 7 days.By the reverse transcriptase activity horizontal survey virus replication in the supernatant liquor, cytotoxicity MTS test determination.Measuring anti-HIV usefulness of these compounds and Cytotoxic result for twice lists in table 7.
All reference, patent and the publication of quoting herein integral body by reference is attached to this paper.
Table 1.
Be used to measure 1 compound concentration scope.
Compound concentration 0 5nM 10nM 25nM 50nM 100nM 250nM
Table 2.
Some compound of the present invention at above-mentioned BrdU in conjunction with the result in measuring.
Table 3
The result of some compound of the present invention in following above-mentioned cell in vitro determination of activity: carry out viability and produce clone cell survival mensuration with the HCT-116 cell, carry out that viability is measured and the active measurement of two anti-NCI cell series (" average-curve M ID value " of anti-Zorubicin resistant cell line and IC50) with the IMR90 cell.
Table 4.
The result of some compound of the present invention in above-mentioned stagnation raji cell assay Raji (IC50, nM).
Figure C200480042035D00671
Table 5.
Some compound of the present invention is in the above-mentioned biochemical result (IC50, μ M) who suppresses in the mensuration
Compound Cdk2/ cyclin E Cdk2/ cyclin A Cdk4/ cyclin D The Cdc2/ cell periodic protein B Cdk6/ cyclin D2 PKA PKC c-Abl
A <0.1 <0.1 <1 <1 <1
B <0.01 <0.1 <10 <0.1
C <0.1 <0.1 <1 <1 >10 >10 >10
D <0.1 <0.1 <1 <1
E <0.01 <0.01 <0.01 <0.1 <0.01 <10 <10 <10
F <0.1 <0.1 <0.01 <0.1 <0.01
G <0.1 <0.1 <0.01 <0.1 >10 >10
H <0.1 <0.1 <0.1 <0.1
I <0.1 <0.1 <0.01 <0.1
J <0.1
K <0.1
L <0.1 <0.1 <0.1
M <0.1
N <0.1 <0.01
O <0.1
P <0.1
Q <0.01 <0.1 <0.01 <0.1 <0.1
Table 6.
The result of other compound in above-mentioned biochemical inhibition and HCT-116 viability mensuration (nonprotein adjusting).
Figure C200480042035D00691
Figure C200480042035D00701
Figure C200480042035D00721
Table 7
Some compound antiviral activity result of the present invention.IC50: 50% virus replication by reversed transcriptive enzyme horizontal survey in the supernatant liquor suppresses; TC50:50% cytotoxicity (MTS); TI:TC50/IC50.
Compound IC50(μM) TC50(μM) TI
A32 <0.01 <0.1 >10
A61 <0.01 <0.1 >10
A64 <0.01 <0.1 >10
C3 <0.01 <0.1 >10
C4 <0.1 <0.1 >1
AZT <0.01 >1.0 >100
Table A
Figure C200480042035D00741
Figure C200480042035D00771
Figure C200480042035D00781
Figure C200480042035D00791
Figure C200480042035D00801
Table B
Figure C200480042035D00802
Figure C200480042035D00811
Figure C200480042035D00821
Figure C200480042035D00831
Figure C200480042035D00841
Figure C200480042035D00851
Figure C200480042035D00861
Figure C200480042035D00871
Figure C200480042035D00881
Figure C200480042035D00891
Figure C200480042035D00901
Figure C200480042035D00911
Figure C200480042035D00921
Figure C200480042035D00951
Figure C200480042035D00961
Figure C200480042035D00971
Table C
Figure C200480042035D00991
Table D
Other compound of the present invention that the possible suitable feature of feature selection produces from following table.For example from following selection, produce compd A 77: nothing-morpholino-aryl-OCH 2(CO)-piperazine-CH 3
Left-handed cyclophane base or ring substituents N dextrorotation substituting group
Substituting group heteroaryl feature
CH 3Morpholino aryl OCH 2The NHM alkyl
Sec.-propyl piperazine thiophene OCH 2(CO) NMM alkoxyl group
CH 3CH 2O (CO) CH 2SO 2Morpholino alcohol
No OCH 2(CO) OCH 2Piperazine replaces amine
Nipecotic acid
The pyrazoles ester
Tetramethyleneimine CH 2CH 2OCH 3
CH 2CH 2OH
CH 2NH 2
CH 2NHCH 2CH 2CH 3
CH 2NHCH 3
CH 2NHCHCH 3CH 3
CH 3
CHCH 3CH 3
COOCH 2CH 3
Do not have
Table E
Other compound of the present invention that the possible suitable feature of feature selection produces from following table.For example from following selection, produce compd B 3: nothing-morpholino-aryl-CH 2-piperazine-CH 2CH 2OH.
Left-handed cyclophane base or ring substituents N dextrorotation substituting group
Substituting group heteroaryl feature
CH 3Morpholino aryl CH 2The NHM alkyl
Sec.-propyl piperazine thiophene CH 2CH 2The NMM alkoxyl group
CH 3CH 2O (CO) CH 2CH 2CH 2CH 2Morpholino alcohol
No CH 2CH 2CH 2CH 2Piperazine replaces amine
Nipecotic acid
The pyrazoles ester
Tetramethyleneimine CH 2CH 2OCH 3
CH 2CH 2OH
CH 2NH 2
CH 2NHCH 2CH 2CH 3
CH 2NHCH 3
CH 2NHCHCH 3CH 3
CH 3
CHCH 3CH 3
COOCH 2CH 3
Replace or unsubstituted virtue
Family's carbocyclic ring
Replace or unsubstituted virtue
Family's heterocycle
Do not have

Claims (9)

1. the compound or its tautomer or the pharmacy acceptable salt form that have formula I structure:
Figure C200480042035C00021
2. the compound of claim 1, wherein said pharmacy acceptable salt form has structure:
Figure C200480042035C00022
3. medicinal compositions, described composition comprise each compound of pharmaceutically acceptable vehicle and claim 1-2.
4. each compound of claim 1-2 is used for the treatment of purposes in the medicine of excess proliferative illness in preparation.
5. each compound of claim 1-2 is used for suppressing the purposes of the medicine of cell proliferation in preparation.
6. each compound of claim 1-2 is used for the treatment of purposes in the medicine for treating viral infections in preparation.
7. the purposes of claim 6, wherein said virus infection is caused by human immunodeficiency virus (HIV).
Claim 1-2 each compound or comprise the described compound compositions for the treatment of significant quantity and be used for purposes in the kinase whose medicine that host in this treatment of needs suppresses cyclin dependent in preparation.
Claim 1-2 each compound or comprise the described compound compositions for the treatment of significant quantity and be used for purposes in the medicine of host's treatment of this treatment of needs illness relevant with the kinases of cyclin dependent in preparation.
CNB2004800420356A 2003-12-23 2004-12-21 Inhibitors of cyclin-dependent kinases, compositions and uses related thereto Expired - Fee Related CN100471858C (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US53187203P 2003-12-23 2003-12-23
US60/531,872 2003-12-23
US60/560,138 2004-04-06
US60/616,480 2004-10-06

Publications (2)

Publication Number Publication Date
CN1922180A CN1922180A (en) 2007-02-28
CN100471858C true CN100471858C (en) 2009-03-25

Family

ID=37779302

Family Applications (1)

Application Number Title Priority Date Filing Date
CNB2004800420356A Expired - Fee Related CN100471858C (en) 2003-12-23 2004-12-21 Inhibitors of cyclin-dependent kinases, compositions and uses related thereto

Country Status (1)

Country Link
CN (1) CN100471858C (en)

Also Published As

Publication number Publication date
CN1922180A (en) 2007-02-28

Similar Documents

Publication Publication Date Title
CN101501054B (en) Phosphoramidate alkylator prodrugs
US20080146555A1 (en) Uses of Kinase Inhibitors and Compositions Thereof
CN101616917A (en) Pyrido [2,3-D] pyrimidine and they are as the purposes of kinase inhibitor
JP2016528251A (en) Methods for the treatment of HER2 amplifying cancer
CN110437205A (en) Pyridine alkenyl piperidine derivative and application thereof
UA125892C2 (en) An aurora a kinase inhibitor for use in the treatment of neuroblastoma
TW200914014A (en) Pharmacological adjunctive treatment associated with glaucoma filtration surgery
US20210246120A1 (en) Method of preparation and use of phosphoinositide 3-kinase inhibitors in treating cancer
JP2005511535A (en) Cyclin-dependent kinase inhibitors, compositions and uses thereof
CN100471858C (en) Inhibitors of cyclin-dependent kinases, compositions and uses related thereto
JP4791975B2 (en) Inhibitors, compositions and related uses for cyclin-dependent kinases
CN107820495A (en) Target the new E PHA4 inhibitor of EPHA4 ligand binding domain
US7893057B2 (en) Inhibitors of cyclin-dependent kinases, compositions and uses related thereto
JP2014515026A (en) Acadesine derivatives, formulations and compositions containing them, their therapeutic use, and their synthesis
CA2471543C (en) Non-nucleosidic inhibitors of reverse transcriptase as antagonists of cell proliferation and inducers of cell differentiation
US7605175B2 (en) Inhibitors of cyclin-dependent kinases, compositions and uses related thereto
COURNIA METHOD OF PREPARATION AND USE OF PHOSPHOINOSITIDE 3-KINASE INHIBITORS IN TREATING CANCER-Patent Information

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1101399

Country of ref document: HK

C14 Grant of patent or utility model
GR01 Patent grant
REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1101399

Country of ref document: HK

C17 Cessation of patent right
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20090325

Termination date: 20121221