ADAM9 inhibitors and uses thereof

文档序号:1145036 发布日期:2020-09-11 浏览:30次 中文

阅读说明:本技术 Adam9抑制剂及其用途 (ADAM9 inhibitors and uses thereof ) 是由 佘玉萍 吴永昌 杨颛丞 郭婷婷 罗佳茜 于 2018-10-29 设计创作,主要内容包括:本发明是关于式(I)的ADAM9抑制剂化合物,包含其盐、水合物和立体异构体,其组合物及其使用方法。包含使用有效量的所述化合物或组合物治疗有需要的人,并检测其结果以改善人的健康或状况。(The present invention relates to ADAM9 inhibitor compounds of formula (I), including salts, hydrates, and stereoisomers thereof, compositions thereof, and methods of use thereof. Comprising treating a human in need thereof with an effective amount of the compound or composition and measuring the results thereof to improve the health or condition of the human.)

1. A method of inhibiting ADAM9, treating cancer, and/or inhibiting cancer cell growth or metastasis, comprising treating a human in need thereof with an ADAM9 inhibitor compound of formula I, or

Use of an ADAM9 inhibitor compound of formula I for the manufacture of a medicament for inhibiting ADAM9, treating cancer and/or inhibiting the growth or metastasis of cancer cells,

Figure FDA0002473031400000011

wherein:

r is a substituted or unsubstituted C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen), or C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen) substituted with heteroatoms (nitrogen, sulfur or oxygen); and

r comprises a C5-C10 organic ring;

or a pharmaceutically acceptable salt, hydrate or stereoisomer.

2. The method or use of claim 1, wherein the organic ring is an aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

3. The method or use of claim 1 or 2 wherein R is a secondary amine or amide.

4. As in claimThe method or use of claim 1 or 2, wherein R is NHR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

5. The method or use of claim 1 or 2 wherein R is NHR1Wherein R is1Is 2-thiazole-4-yl-ethyl-isoindoline-1, 3-dione. 12W-0271

6. The method or use of claim 1 or 2 wherein R is NHCOR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

7. The method or use of claim 1 or 2 wherein R is NHCOR1Wherein R is1Is benzothiophene. 6X-0214

8. The method or use of claim 1 or 2 wherein R is:

wherein R is1Is an aryl group optionally substituted containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

9. The method or use of claim 1 or 2 wherein R is:

wherein R is1Is a substituted phenyl group.

10. The method or use of claim 1 having the formula of table one.

11. A composition comprises a compound of formula I co-formulated with different anti-cancer compounds,

Figure FDA0002473031400000023

wherein:

r is a substituted or unsubstituted C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen), or C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen) substituted with heteroatoms (nitrogen, sulfur or oxygen); and

r comprises a C5-C10 organic ring;

or a pharmaceutically acceptable salt, hydrate or stereoisomer.

12. The composition of claim 11 wherein the organic ring is an aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

13. The composition of claim 11 or 12 wherein R is a secondary amine or amide.

14. The composition of claim 11 or 12 wherein R is NHR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

15. The composition of claim 11 or 12 wherein R is NHR1Wherein R is1Is 2-thiazole-4-yl-ethyl-isoindoline-1, 3-dione. 12W-0271

16. The composition of claim 11 or 12 wherein R is NHCOR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

17. The composition of claim 11 or 12 wherein R is NHCOR1Wherein R is1Is benzothiophene. 6X-0214

18. The composition of claim 11 or 12 wherein R is:

Figure FDA0002473031400000031

wherein R is1Is an aryl group optionally substituted containing 0-3 heteroatoms (nitrogen, sulfur or oxygen).

19. The composition of claim 11 or 12 wherein R is:

Figure FDA0002473031400000041

wherein R is1Is a substituted phenyl group.

20. The composition of claim 11 or 12 having the formula of table one.

Background

ADAM 9(Disintegrin and metalloprotease domain-containing protein 9) is overexpressed in many tumors, such as pancreatic, breast, prostate and lung cancers, and high expression of ADAM9, which is associated with poor prognosis in cancer patients, can be used as a predictive marker because ADAM9 expression can help tumor cells adapt to unfavorable environments, making ADAM9 thought to promote tumor development and be a better therapeutic target than other cancer-associated metalloproteases. ADAM9 has the ability to cleave and release many molecules involved in cancer progression, participates in tumorigenesis because it can cleave and release many molecules involved in cancer progression, and ADAM9 secreted by stromal cells around tumors can promote tumor development through neovascularization.

Our previously published results show that inhibition of ADAM9 expression and its downstream signaling can significantly prolong the survival time of lung tumor mice. It is evident from clinical specimens of lung and breast cancer that patients with low ADAM9 expression levels in tumor specimens survive longer than patients with high ADAM9 expression levels. In addition, since mice deficient in ADAM9 have no apparent phenotype, it is suggested that ADAM9 may have good drug tolerance as a target protein.

In the invention, the design of a small molecule compound is disclosed, and the small molecule compound is confirmed to be capable of being used as an ADAM9 inhibitor to block the activity of ADAM9 protease.

Disclosure of Invention

The present invention provides methods and compositions for inhibiting ADAM9, treating cancer, and/or inhibiting the growth or metastasis of cancer cells. The present invention further provides ADAM9 inhibitor compounds, including salts, hydrates, and stereoisomers thereof, compositions thereof, and methods of using the same, comprising treating a human in need thereof with an effective amount of the compound or composition, and testing the results to improve the health or condition of the human.

In one aspect, the present invention provides a method of inhibiting ADAM9, treating cancer, and/or inhibiting the growth or metastasis of cancer cells, comprising treating a human in need thereof with an ADAM9 inhibitor compound of formula I, or the use of an ADAM9 inhibitor compound of formula I in the manufacture of a medicament for inhibiting ADAM9, treating cancer, and/or inhibiting the growth or metastasis of cancer cells,

wherein:

r is a substituted or unsubstituted C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen), or C1-C18 hydrocarbyl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen) substituted with heteroatoms (nitrogen, sulfur or oxygen); and

r comprises a C5-C10 organic ring;

or a pharmaceutically acceptable salt, hydrate or stereoisomer.

In the examples

-the organic ring is an aryl group comprising 0-3 heteroatoms (nitrogen, sulphur or oxygen);

-R is a secondary amine;

-R is an amide;

r is NHR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen);

r is NHR1Wherein R is1Is 2-thiazole-4-yl-ethyl-isoindoline-1, 3-dione (12W-0271);

r is NHCOR1Wherein R is1Is a substituted or unsubstituted aryl group containing 0-3 heteroatoms (nitrogen, sulfur or oxygen);

-R is NHCOR1Wherein R is1Benzothiophene (6X-0214);

-R is:

wherein R is1Is an aryl group optionally substituted containing 0-3 heteroatoms (nitrogen, sulfur or oxygen);

-R is:

wherein R is1Is a substituted phenyl group; and/or

-the formula of said compound is shown in table one;

another aspect of the present invention is to provide a composition comprising an ADAM9 inhibitor compound of formula I co-formulated with different anti-cancer compounds.

The present invention includes all combinations of the specific embodiments described herein.

Detailed Description

The following description of specific embodiments and examples is intended to be illustrative only and not to be limiting, as various non-critical parameters may be modified or varied by those skilled in the art to produce substantially similar results. The present invention provides various embodiments.

Unless otherwise indicated, "a" and "an" as used herein and in the specification means that the grammatical object in the article is one or more (i.e., at least one), "or" means "and/or", and polynucleic acid sequences are understood to include both sense and antisense sequences and the alternative backbones described herein. Further, a category is an abbreviation for all members contained in the category; for example, C1-C3 alkyl is an abbreviation for all C1-C3 alkyl groups, for example C1-C3 alkyl groups include methyl, ethyl, propyl and isomers thereof.

The following terms, phrases and symbols are generally intended to have the meanings set forth below, unless the context in which they are used indicates otherwise. The following abbreviations and terms have the indicated meanings.

The term "alkyl" as used herein refers to straight and branched chain saturated hydrocarbon groups having 1 to 18, 1 to 12 or 1 to 6 carbon atoms. Examples of alkyl groups include methyl, ethyl, 1-propyl or n-propyl ("n-Pr"), 2-propyl or isopropyl ("i-Pr"), 1-butyl or n-butyl ("n-Bu"), 2-methyl-1-propyl or isobutyl ("i-Bu"), 1-methylpropyl or sec-butyl ("s-Bu"), and 1, 1-dimethylethyl or tert-butyl ("t-Bu"). Other examples of alkyl groups include, but are not limited to, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, and 3, 3-dimethyl-2-butyl.

The lower alkyl group means an alkyl group having 1 to 8, preferably 1 to 6, and more preferably 1 to 4 carbon atoms. The lower alkenyl or alkynyl group means an alkenyl or alkynyl group having 2 to 8, 2 to 6 or 2 to 4 carbon atoms.

The term "alkenyl" as used herein refers to a straight or branched chain hydrocarbon group selected from the group consisting of a C ═ C double bond and 2 to 18, 2 to 12, or 2 to 6 carbon atoms. Examples of alkenyl groups may be selected from, but are not limited to, vinyl or vinyl, prop-1-enyl, prop-2-enyl, 2-methylprop-1-enyl, but-2-enyl, but-3-enyl, but-1, 3-dienyl, 2-methylbut-1, 3-diene, hex-1-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl and hexa-1, 3-dienyl groups.

The term "alkynyl" as used herein refers to a hydrocarbon group selected from straight and branched chain hydrocarbon groups containing at least one C.ident.C triple bond and 2-18, 2-12 or 2-6 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl (propargyl), 1-butynyl, 2-butynyl, and 3-butynyl.

As used herein, "cycloalkyl" refers to a cyclic hydrocarbon group selected from saturated and partially unsaturated, including monocyclic and polycyclic (e.g., bicyclic and tricyclic) groups. For example, the cycloalkyl group may be a cyclic hydrocarbon group having 3 to 12, 3 to 8, or 3 to 6 carbon atoms. Further, the cycloalkyl group may be a monocyclic group having 3 to 12, 3 to 8, or 3 to 6 carbon atoms. Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, and cyclododecyl. Examples of bicyclic cycloalkyl groups include, but are not limited to, a ring system having 7-12 ring atoms and whose bicyclic arrangement is selected from [4,4], [4,5], [5,6] and [6,6], or a bridged bicyclic ring selected from bicyclo [2.2.1] heptane, bicyclo [2.2.2] octane and bicyclo [3.2.2] nonane. The rings may be saturated or have at least one double bond (i.e., partially unsaturated), but are not fully conjugated and are not aromatic. Aromatic groups having an aryl group are defined otherwise in this specification.

The term "aryl" as used herein refers to a group selected from: 5-and 6-membered carbocyclic aromatic rings, for example phenyl, bicyclic and tricyclic ring systems. Wherein the bicyclic ring system can be a 7-12 membered bicyclic ring system wherein at least one ring is carbocyclic and aromatic and is selected from, but not limited to, naphthalene, indane, and 1,2,3, 4-tetrahydroquinoline. The tricyclic ring system can be a 10-15 membered tricyclic ring system in which at least one ring is a carbocyclic ring and an aromatic ring, such as fluorene.

For example, aryl is selected from 5-and 6-membered carbocyclic aromatic rings fused to a 5-to 7-membered cycloalkyl or heterocyclic ring optionally containing at least one heteroatom selected from nitrogen, oxygen and sulfur. The point of attachment is on the carbocyclic aromatic ring when the carbocyclic aromatic ring is fused to the heterocyclic ring, and may be on the carbocyclic aromatic ring or the cycloalkyl when the carbocyclic aromatic ring is fused to the cycloalkyl. Divalent radicals formed from substituted benzene derivatives and having a free valence at the ring atom are referred to as substituted phenylene radicals. Divalent radicals derived from monovalent polycyclic hydrocarbon radicals whose name ends with a "-yl (-idene)" by removing one hydrogen atom from a carbon atom having a free valence are named by adding a "-ylidene (-yl)" to the name of the corresponding monovalent radical, for example naphthyl having two points of attachment is called naphthylene. However, aryl groups do not include or overlap heteroaryl groups, as defined separately below in this specification. Thus, if one or more carbocyclic aromatic rings are fused to a heterocyclic aromatic ring, the resulting ring system is heteroaryl, rather than aryl as defined herein.

The term "halogen" as used herein refers to fluorine, chlorine, bromine or iodine.

The term "heteroalkyl" as used herein refers to an alkyl group containing at least one heteroatom.

The term "heteroaryl" as used herein refers to a group selected from:

a 5-to 7-membered aromatic monocyclic ring comprising 1,2,3 or 4 heteroatoms selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon;

an 8-to 12-membered bicyclic ring comprising 1,2,3 or 4 heteroatoms selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon, and wherein at least one ring is aromatic and at least one heteroatom is present in the aromatic ring.

For example, heteroaryl groups comprise a 5-to 7-membered heterocyclic aromatic ring fused to a 5-to 7-membered cycloalkyl ring. The aforementioned fused bicyclic heteroaryl ring systems, wherein only one ring contains at least one heteroatom, the point of attachment may be on the heteroaryl or cycloalkyl ring.

When the total number of sulfur and oxygen atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to each other. In some embodiments, the total number of sulfur and oxygen atoms in the heteroaryl group is no greater than 2, and in other embodiments, the total number of sulfur and oxygen atoms in the aromatic heterocycle is no greater than 1.

Examples of heteroaryl groups include, but are not limited to (numbering starting from the position of the linkage that assigns priority 1) pyridyl (e.g., 2-pyridyl, 3-pyridyl or 4-pyridyl), quinolinyl, pyrazinyl, 2, 4-pyrimidinyl, 3, 5-pyrimidinyl, 2, 4-imidazolyl, imidazopyridinyl, isoxazolyl, oxazolyl, thiazolyl, isothiazolyl, thiadiazolyl, tetrazolyl, thienyl, triazinyl, benzothienyl, furyl, benzofuryl, benzimidazolyl, indolyl, isoindolyl, indolinyl, phthalazinyl, pyrazinyl, pyridazinyl, pyrrolyl, triazolyl, quinolinyl, isoquinolinyl, pyrazolyl, pyrrolopyridyl (e.g., 1H-pyrrolo [2,3-b ] pyridin-5-yl), pyrazolopyridyl (e.g., 1H-pyrazolo [3,4-b ] pyridin-5-yl), benzoxazolyl (e.g. benzo [ d ] oxazol-6-yl), piperidinyl, purinyl, 1-oxa-2, 3-oxadiazolyl, 1-oxa-2, 4-oxadiazolyl, 1-oxa-2, 5-oxadiazolyl, 1-oxa-3, 4-oxadiazolyl, 1-thiophene-2, 3-oxadiazolyl, 1-thiophene-2, 4-oxadiazolyl, 1-thiophene-2, 5-oxadiazolyl, 1-thiophene-3, 4-oxadiazolyl, furanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, furopyridinyl, benzothiazolyl (e.g. benzo [ d ] thiazol-6-yl), Indazolyl (e.g., 1H-indazol-5-yl) and 5,6,7, 8-tetrahydroisoquinoline.

The term "heterocycle" or "heterocyclyl" as used herein refers to a ring selected from the group consisting of 4-to 12-membered monocyclic, bicyclic, and tricyclic rings, as well as saturated and partially unsaturated rings. The saturated and partially unsaturated rings contain at least one carbon atom in addition to 1,2,3 or 4 heteroatoms selected from oxygen, sulfur and nitrogen. "heterocycle" also refers to a 5-to 7-membered heterocyclic ring containing at least 1 heteroatom selected from nitrogen, oxygen, and sulfur, said heteroatom being fused to a 5-, 6-, and/or 7-membered cycloalkyl, carbocyclic aromatic, or heteroaromatic ring, provided that when the heterocyclic ring is fused to a carbocyclic aromatic or heteroaromatic ring, the point of attachment is on the heterocyclic ring, and when the heterocyclic ring is fused to a cycloalkyl group, the point of attachment can be on the cycloalkyl or heterocyclic ring.

"heterocycle" also refers to an aliphatic spirocycle containing at least one heteroatom selected from nitrogen, oxygen, and sulfur, provided that the point of attachment is on the heterocycle. The ring may be saturated or have at least one double bond (i.e., partially unsaturated). The heterocyclic ring may be substituted by oxo. The attachment point in the heterocycle may be a carbon or a heteroatom. Heterocyclyl is not heteroaryl as defined in the specification.

Examples of heterocycles include, but are not limited to (numbering starting from the position of the linkage that assigns priority 1) 1-pyrrolidinyl, 2, 4-imidazolidinyl, 2, 3-pyrazolidinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2, 5-piperazinyl, pyranyl, 2-morpholinyl, 3-morpholinyl, oxiranyl, aziridinyl, thietanyl, azetidinyl, oxetanyl, thietanyl, 1, 2-dithianyl, 1, 3-dithianyl, dihydropyridinyl, tetrahydropyridinyl, thiomorpholinyl, thienyl, piperazinyl, homopiperazinyl, homopiperidinyl, azepanyl, oxepanyl, thienyl, 1, 4-oxathiolane, 1, 4-dioxepanyl, 1, 4-oxathiolane, 1, 4-oxaazepanyl, 1, 4-dithienyl, 1, 4-thiacycloheptane, 1, 4-diazepane, 1, 4-dithianyl, 1, 4-azepanyl, oxazepinyl, diazepinyl, thiazepinyl, dihydrothienyl, dihydropyranyl, dihydrofuranyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, dihydro-pyranyl, tetrahydro-pyranyl, 1, 4-dioxanyl, 1, 3-dioxolanyl, pyrazolinyl, pyrazolidinyl, dithiol, dithiolane, pyrazolylimidazolinyl, pyrazolylidinyl, pyrimidinone, 1-dioxo-thiomorpholinyl, 3-azabicyclo [3.1.0] hexyl, 3-azabicyclo [4.1.0] heptyl and azabicyclo [2.2.2] hexyl. Substituted heterocycles also include ring systems substituted with one or more moieties substituted with oxygen, such as piperidinyl N-oxide, morpholinyl N-oxide, 1-oxo-1-thiomorpholinyl, and 1, 1-dioxo-1-thiomorpholinyl.

The substituents are selected from: halogen, -R ', -OR', -O, -NR ', -N-OR', -NR 'R ", -SR', -SiR 'R'", -oc (O) R ', -c (O) R', -CO2R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR'-C(O)NR"R'"、-NR'SO2NR'"、-NR"CO2R'、-NH-C(NH2)=NH、-NR'C(NH2)=NH、-NH-C(NH2)=NR'、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R, -CN and-NO2、-N3、-CH(Ph)2Perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl in an amount ranging from 0 to 3, preferably a group having 0, 1 or 2 substituents. R ', R "and R'" are each independently hydrogen, unsubstituted (C1-C8) alkyl and heteroalkyl, unsubstituted aryl, aryl substituted with 1 to 3 halogens, unsubstituted alkyl, alkoxy or thioalkoxy, or aryl- (C1-C4) alkyl. When R' and R "are attached to the same nitrogen atom, they may combine with the nitrogen atom to form a 5-, 6-or 7-membered ring. Thus, -NR' R "includes 1-pyrrolidinyl and 4-morpholinyl and" alkyl "includes trihaloalkyl groups (e.g., -CF)3and-CH2CF3) And when the aryl group is 1,2,3, 4-tetralin, it may be substituted with a substituted or unsubstituted (C3-C7) spirocycloalkyl group. And the (C3-C7) spirocycloalkyl group may be substituted in the same manner as defined for "cycloalkyl" in the specification.

Preferred substituents are selected from: halogen, -R ', -OR', -O, -NR 'R ", -SR', -SiR 'R', -OC (O) R ', -C (O) R', -CO2R'、-CONR'R"、-OC(O)NR'R"、-NR"C(O)R'、-NR"CO2R'、-NR'-SO2NR"R'"、-S(O)R'、-SO2R'、-SO2NR'R"、-NR"SO2R, -CN and-NO2Perfluoro (C1-C4) alkoxy and perfluoro (C1-C4) alkyl, wherein R 'and R' are as defined above.

The compounds may contain asymmetric centers and may therefore exist as enantiomers. When a compound has two or more asymmetric centers, it may additionally exist as a non-enantiomeric isomer. Enantiomers and diastereomers belong to a broader class of stereoisomers. All possible stereoisomers include, for example, substantially pure, isolated enantiomers, racemic mixtures thereof, and mixtures of non-enantiomers. All stereoisomers of the compounds and/or pharmaceutically acceptable salts thereof are included where applicable. Unless otherwise indicated, references to isomers apply to any possible isomer. And when the isomeric composition is not specified, all possible isomers are included.

As used herein, "substantially pure" means that the target stereoisomer contains no more than 35%, such as no more than 30%, preferably no more than 25%, more preferably no more than 20% by weight of any other stereoisomer. In some embodiments, "substantially pure" means that the target stereoisomer contains no more than 10%, preferably no more than 5%, more preferably no more than 1% by weight of any other stereoisomer.

When a compound contains an olefinic double bond, unless otherwise indicated, a double bond indicates that the compound contains both E and Z geometric isomers.

Some compounds may exist at different points of attachment of hydrogen and are referred to as tautomers. For example, containing carbonyl-CH2Compounds with a c (o) -group (keto form) may undergo tautomerization to form a hydroxy-CH ═ c (oh) -group (enol form). Mixtures in which both the ketone and the enol are formed separately and contain both the ketone and the enol are also included where applicable.

It is advantageous to separate the reaction products from each other and/or from the starting materials. The desired product of each step or series of steps may be isolated and/or purified (hereinafter referred to as isolated) to a desired degree of homogeneity by techniques commonly used in the art. Typically the aforementioned separation involves heterogeneous extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography may include reverse phase chromatography, normal phase chromatography, size exclusion chromatography, ion exchange chromatography, high, medium, and low pressure liquid chromatography and apparatus, small scale analytical chromatography, simulated moving bed chromatography ("SMB"), and preparative thin layer chromatography or thick layer chromatography, as well as small scale thin layer and flash chromatography. The skilled person will apply the most suitable technique to achieve the desired separation.

Mixtures of non-enantiomers may be separated by methods well known to those skilled in the art, for example by chromatography and/or fractional crystallization based on physicochemical differences between the non-enantiomers. The mixture of enantiomers is converted to a mixture of non-enantiomers by reaction with a suitable optically active compound (e.g., a chiral auxiliary such as a chiral alcohol or Mosher acyl chloride), the non-enantiomers are separated and each non-enantiomer is converted (e.g., hydrolyzed) to the corresponding pure enantiomer. The enantiomers may also be separated using a chiral HPLC column.

Single stereoisomers, such as substantially pure enantiomers, can be obtained by using optically active separating agents (see Eliel, E.and Wilen, S.Stereochemistry of Organic Compounds.New York: John Wiley & Sons, Inc., 1994; Lochmuller, C.H., et al, "Chromatographic resolution of enantiomers: selected review," J.Chromatographer, 113(3) (1975) pp.283-302). The racemic mixture of the chiral isomers of the present invention can be isolated and purified by any suitable method, comprising: (1) forming the chiral isomer into an ionic non-enantiomer salt and separating by fractional crystallization or other methods; (2) forming a diastereoisomer by using a chiral derivatization reagent, separating the diastereoisomer, and converting the diastereoisomer into a pure stereoisomer; and (3) direct separation of substantially pure or enriched stereoisomers under chiral conditions. Reference may be made to: wainer, Irving w., ed. drug Stereochemistry: Analytical Methods and pharmacology.new York: marcel dekker, inc., 1993.

The term "pharmaceutically acceptable salt" as used herein includes, but is not limited to, the salts with inorganic acidsSalts thereof and salts with organic acids. The salts formed with inorganic acids can be selected, for example, from the hydrochloride, phosphate, diphosphate, hydrobromide, sulfate, sulfinate and nitrate salts. The salt with an organic acid can be selected, for example, from malic acid, maleic acid, fumaric acid, tartaric acid, succinic acid, citric acid, lactic acid, methanesulfonic acid, p-toluenesulfonic acid, 2-isethionic acid, benzoic acid, salicylic acid, stearic acid, alkanoic acid (e.g. acetic acid) and HOOC- (CH)2) n-COOH, wherein n is 0 to 4. Examples of pharmaceutically acceptable cations include, but are not limited to, sodium, potassium, calcium, aluminum, lithium, and ammonium.

Furthermore, if the compound is used as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt (e.g., a pharmaceutically acceptable addition salt) may be prepared by dissolving the free base in a suitable organic solvent and treating the solution with an acid, followed by conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art can prepare non-toxic pharmaceutically acceptable addition salts using a variety of synthetic methods without undue experimentation.

The term "treating" or "treatment" as used herein refers to administering at least one compound and/or at least one stereoisomer and/or at least one pharmaceutically acceptable salt thereof to a subject in need thereof, e.g., a cancer patient.

An "effective amount" as referred to herein, refers to an amount of at least one compound and/or at least one stereoisomer thereof and/or at least one pharmaceutically acceptable salt thereof that is effective to "treat" a disease or disorder in a subject. An effective amount will have some correlation with the biological or medical response of the tissue, system, animal or human to which it is administered, e.g., it will be sufficient to prevent the development of one or more diseases or conditions or to alleviate the symptoms of one or more of the conditions or conditions being treated to some extent when administered. The therapeutically effective amount will vary depending on the compound, the disease and its severity, and the age and weight of the mammal to be treated.

The term "at least one substituent" as used herein includes, but is not limited to, 1 to 4 substituents, 1 to 3 substituents, 1 or 2 substituents. For example, the "at least one substituent L-R4Contains 1 to 4, 1 to 3, 1 or 2L-R selected from the group described in the specification4Substituents of the list.

The compounds of the present invention, stereoisomers thereof and pharmaceutically acceptable salts thereof may be used alone or in combination with at least one other therapeutic agent for treatment. In some embodiments, the compounds, stereoisomers thereof, and pharmaceutically acceptable salts thereof, may be used in combination with at least one additional therapeutic agent. The at least one additional therapeutic agent may be selected from the group consisting of anti-hyperproliferative agents, anti-cancer agents, and chemotherapeutic agents. The compounds and/or a pharmaceutically acceptable salt disclosed herein may be administered together with at least one other therapeutic agent in a single dosage form, or as separate dosage forms. When administered as a separate dosage form, at least one other therapeutic agent can be administered prior to, concurrently with, or subsequent to the administration of a compound and/or a pharmaceutically acceptable salt disclosed herein.

The term "chemotherapeutic agent" as used herein refers to a chemical compound used to treat cancer regardless of its mechanism of action. Chemotherapeutic agents include compounds used in "targeted therapy" and conventional chemotherapy. Suitable chemotherapeutic agents, for example, may be selected from: apoptosis-inducing agents, polynucleotides (e.g., ribozymes), polypeptides (e.g., enzymes), drugs, biomimetics, alkaloids, alkylating agents, antitumor antibiotics, antimetabolites, hormones, platinum compounds, monoclonal antibodies that bind to anticancer drugs, toxins, and/or radionuclides, biological response modifiers (e.g., interferons such as IFN-alpha and interleukins such as IL-2), adoptive immunotherapeutic agents, hematopoietic growth factors, agents that induce tumor cell differentiation (e.g., all-trans retinoic acid), gene therapy agents, antisense therapeutic agents and nucleotides, tumor vaccines, and angiogenesis inhibitors.

Examples of chemotherapeutic agents include, but are not limited to, Erlotinib (Erlotinib) ((R))Genentech/OSI Pharm.); bortezomib (Bortezomib) (Bortezomib)

Figure BDA0002473031410000102

Millennium Pharm.); fulvestrant (Fulvestrant) ((ii))AstraZeneca); sunitinib (Sunitinib) ((Sunitinib))Pfeiri); letrozole (Letrozole) ((L))Noval); imatinib mesylate (imatinib mesylate) (ii) a salt thereof

Figure BDA0002473031410000106

Noval); PTK787/ZK 222584 (Nowa); oxaliplatin (Oxaliplatin) ((Oxaliptin))

Figure BDA0002473031410000107

Sanofi); 5-FU (5-fluorouracil); calcium folinate (Leucovorin); rapamycin (Rapamycin) (sirolimus,wyeth); lapatinib (Lapatinib) ((Lapatinib))GSK572016, Glaxo Smith Kline); lonafarnib (Lonafarnib) (SCH 66336); sorafenib (Sorafenib) ((Sorafenib))

Figure BDA00024730314100001010

Bayer); irinotecan (Irinotecan) (Irinotecan)Pyroxene and Gefitinib (Gefitinib) (Gefitinib)

Figure BDA00024730314100001012

Asitecavirenz); AG1478, AG1571(SU 5271, Sugen); alkylating agents, such as thiotepa and cyclophosphamide (C)(ii) a cyclosphosphamide); alkyl sulfonates such as busulfan, improsulfan and piposulfan; azepanes, such as benzodiazepine (benzodipa), carboquinoline (carboquone), meturedpa (meturedpa) and uredpa (uredpa); ethyleneimine and methylmelamine compounds such as altretamine (altretamine), triethylenemelamine (triethyleneamine), triethylenephosphoramide (triethylenephosphoramide), triethylenethiophosphoramide (triethylenethiophosphamide), and trimethylolmelamine (trimethylolmelamine); acetyl biosynthetic compounds (acetogenins) (e.g., bullatacin and bullatacin); camptothecin (camptothecin) (e.g., the synthetic analog topotecan); bryostatin; spongopus ketene (callystatin), CC-1065 and their adozelesin (adozelesin), carvelesin (carzelesin) and bizelesin (bizelesin) synthetic analogs; cryptophycins (e.g., cryptophycins 1 and 8); dolastatin (dolastatin); duocarmycins (duocarmycins) and their synthetic analogs, such as KW-2189 and CB1-TM 1; esophagin (eleutherobin); coprinus atrata base (pancratistatin); sarcodictyin; spongistatin (spongistatin); nitrogen mustards such as chlorambucil (chlorambucil), chlorambucil (chlorephazine), cholorophosphamide (chlorophosphamide), estramustine (estramustine), ifosfamide (ifosfamide), mechlorethamine (mechlorethamine), mechlorethamine hydrochloride (mechlorethamine oxydichloride), melphalan (melphalan), neomustard (novembichin), benzene mustard cholesterol (pherenesterene), prednimustine (prednimustine), triamcinolone (trofosfamide), uracil mustard (uracil mustard); nitrosoureas, e.g. carmustine (carmustine), chlorouretocin (chlorozotocin), fotemustine (fotemustine), lomustine (lomustine), nimustine (nimustine) and ramustine (ranirnustine)(ii) a Antibiotics, such as enediynes antibiotics (enediynes) (e.g., calicheamicins, especially calicheamicin γ 1I and calicheamicin ω I1 (see, e.g., AgnewChem. Intl. Ed. Engl. (1994) 33: 183-186), anthracyclines (kinemicins), including dynemicin A, bisphosphonates (e.g., clodronate), epothilones (esperamicin), and neocarzinostatin (neocarzinostatin) and related chromoproteins enediynes antibiotics chromophores, clarithromycin (aclomycins), actinomycins (actinomycin), anidamycins (auroramicins), azaserins (serzaines), blecins (eomycins), actinomycins (actinomycins), monocrotamycins (monocrotamycins) (e-5), monocrotamycins (monocrotamycins) (monocins) (monocrotamycins) (monocins), monocrotamycins (erythromycin (monocins) (monocrotamycins) (monocins) (monocrotamycins) (,

Figure BDA0002473031410000111

Doxorubicin (doxorubicin), morpholinodoxorubicin, cyanomorpholinodoxorubicin, 2-pyrrolodoxorubicin and deoxydoxorubicin), epirubicin (epirubicin), esorubicin (esorubicin), idarubicin (idarubicin), marijumycin (marcelomycin), mitomycins (mitomycins) such as mitomycin C, mycophenolic acid (mycophenolic acid), norramycin (nogalamycin), olivomycin (olivomycin), pelomycin (polyplomycin), pofiromycin (porfiromycin), puromycin (puromycin), griseofibrinomycin (quelamycin), rodobicin (rodorubicin), streptonigromycin (streptonigrin), streptozocin (streptazocin), tubercidin (tubercidin), amectin (zomycin), zorubicin (zosin), zosin (zostatin); antimetabolites such as methotrexate (methotrexate) and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs, such as fludarabine (fludarabine), 6-mercaptopurine (6-mercaptopurine), thiamiprine (thiamiprine), thioguanine (thioguanine); pyrimidine analogues, e.g. annDecitabine (ancitabine), azacitidine (azacitidine), 6-azauridine (6-azauridine), carmofur (carmofur), cytarabine (cytarabine), dideoxyuridine (dideoxyuridine), doxifluridine (doxifluridine), enocitabine (enocitabine), floxuridine (floxuridine); androgens such as carotinone (calusterone), dromostanolone propionate, epitioandrostanol (epitiostanol), mepiquitane (mepiquitane), testolactone (testolactone); anti-adrenal agents, such as aminoglutethimide (aminoglutethimide), mitotane (mitotane), trilostane (trilostane); folic acid supplements, such as folinic acid (frilic acid); acetoglucurolactone (acegultone); an aldophosphamide glycoside (aldophosphamideglycoside); aminolevulinic acid (aminol evulinic acid); eniluracil (eniluracil); amsacrine (amsacrine); estradiol-chlorambucil complex (bestrabucil); bisantrene; edatrexate (edatraxate); desphosphamide (defofamine); dimecorsine (demecolcine); diazaquinone (diaziqutone); erfurticin (elfortimihine); ammonium etitanium acetate; epothilone (epothilone); etoglut (etoglucid); gallium nitrate; hydroxyurea (hydroxyurea); lentinan (lentinan); lonidamine (lonidainine); maytansinoids (maytansinoids), such as maytansine (maytansine) and ansamitocins (ansamitocins); mitoguazone (mitoguzone); mitoxantrone (mitoxantrone); mogradol (mopidanmol); diamine nitracridine (nitrarine); pentostatin (pentostatin); methionine mustard (phenamett); pirarubicin (pirarubicin); losoxantrone (losoxantrone); podophyllinic acid (podophyllic acid); 2-ethyl hydrazide (2-ethyl hydrazide); procarbazine (procarbazine);polysaccharide complex (JHS natural products, Eugene, Oreg.); razoxane (rizoxane); rhizomycin (rhizoxin); azofurans (sizofurans); helical germanium (spirogermanium); tenuazonic acid (tenuazonic acid); triimine quinone (triaziquone); 2,2' -trichlorotriethylamine; trichothecenes, in particular T-2 toxin, verrucin A, rhabdocin A and rorodin ASnakeheads (anguidines)); urethane (urethan); vindesine (vindesine); dacarbazine (dacarbazine); mannomustine (mannomustine); dibromomannitol (mitobronitol); dibromodulcitol (mitolactol); piperobromane (pipobroman); a polycytidysine; cytarabine (arabine) ("Ara-C"); cyclophosphamide (cyclophosphamide); thiotepa (thiotepa); taxoids, e.g. taxol

Figure BDA0002473031410000122

Paclitaxel (paclitaxel, Bristol-Myers Squibb Oncology, Princeton, N.J.),without Cremophor-free, albumin engineered nanoparticle dosage forms of paclitaxel (American Pharmaceutical Partners, Schaumberg, Ill.) anddocetaxel (doxetaxel; Rhone-Poulenc Rorer, Antony, France); chlorambucil (chlorembucil);

Figure BDA0002473031410000131

gemcitabine (gemcitabine); 6-thioguanine (6-thioguanine); mercaptopurine (mercaptoprine); methotrexate (methotrexate); platinum analogs, such as cisplatin (cissplatin) and carboplatin (carboplatin); vinblastine (vinblastine); etoposide (VP-16); ifosfamide (ifosfamide); mitoxantrone (mitoxantrone); vincristine (vincristine);

Figure BDA0002473031410000132

vinorelbine (vinorelbine); oncostatin (novantrone); teniposide (teniposide); edatrexate (edatrexate); daunomycin (daunomycin); aminopterin (aminopterin); (xiloda); capecitabine (capecitabine)

Figure BDA0002473031410000133

Ibandronate (ibandronate); anticancer drug (CPT-11); topology differenceA brosidase inhibitor RFS 2000; difluoromethyl ornithine (DMFO); retinoids (e.g., tretinoin); and pharmaceutically acceptable salts, acids and derivatives of any of the foregoing.

The "chemotherapeutic agent" may also be selected from: (i) anti-hormonal agents used to modulate or inhibit the effects of hormones on tumors, such as anti-estrogens (anti-estrogens) and Selective Estrogen Receptor Modulators (SERMs), including tamoxifen (tamoxifen) (includingTamoxifen citrate (tamoxifen citrate)), raloxifene (raloxifene), droloxifene (droloxifene), 4-hydroxytamoxifen (4-hydroxytamoxifen), troloxifene (trioxifene), raloxifene (keoxifene), LY117018, onapristone (onapristone), and(toremifine citrate); (ii) aromatase inhibitors which inhibit aromatase which regulates estrogen production in the adrenal gland, e.g. 4(5) -imidazole, aminoglutethimide,

Figure BDA0002473031410000136

(megestrol acetate),

Figure BDA0002473031410000137

(exemestane; pyroxene), formestane (formestanie), fadrozole (fadrozole),

Figure BDA0002473031410000138

(vorozole),

Figure BDA0002473031410000139

(letrozole; noval) and(anastrozole; astemikang); (iii) anti-androgens, such as flutamide, nilutamide, bicalutamide, leuprolideRerelin (leuprolide) and goserelin (goserelin), and troxacitabine (troxacitabine) (1, 3-dioxolane nucleoside analogues), (iv) protein kinase inhibitors, (v) lipid kinase inhibitors, (vi) antisense oligonucleotides, in particular, that inhibit genes in signal transduction pathways associated with aberrant cell proliferation such as PKC- α, Ralf and H-Ras, (vii) ribozymes, such as VEGF expression inhibitors (e.g., such as) And inhibitors of HER2 expression; (viii) vaccines, e.g. gene therapy vaccines, e.g.And

Figure BDA00024730314100001314

rlL-2; topoisomerase 1 inhibitors, e.g.

Figure BDA00024730314100001315

Figure BDA00024730314100001316

rmRH; (ix) anti-angiogenic agents, e.g. bevacizumab (b)Genentech); and (x) pharmaceutically acceptable salts, acids and derivatives of any of the foregoing.

The "chemotherapeutic agent" may also be selected from therapeutic antibodies, such as the Allium monoclonal antibody (Campath), the Bevacizumab (R) antibody

Figure BDA0002473031410000141

Genentech); cetuximab monoclonal antibody (Imclone); monoclonal antibody against Palmaria (

Figure BDA0002473031410000143

Amgen), rituximab monoclonal antibody (

Figure BDA0002473031410000144

Genentech/Biogen Idec), Pertuzumab antibody (2C4, Genentech), and a Tremella monoclonal antibody (Genentech), tositumomum monoclonal antibody (Bexxar, Corixia), and antibody drug conjugate, gemtuzumab ozogamicin (a) monoclonal antibody

Figure BDA0002473031410000147

Wyeth)。

Humanized monoclonal antibodies having therapeutic potential as chemotherapeutic agents, together with their target compounds, stereoisomers thereof and pharmaceutically acceptable salts thereof, may be selected from the group consisting of: alemtuzumab (alemtuzumab), aliskirilowib monoclonal antibody (apiolizumab), asezuzumab monoclonal antibody (aselizumab), aliskirilowib monoclonal antibody (atlizumab), bapirozumab monoclonal antibody (bapineuzumab), bevacizumab antibody (bevacizumab), bivatuzumab antibody maytansine (bivatuzumab), memantine monoclonal antibody maytansine (cantuzumab mertansine), cetuzumab monoclonal antibody (cetuzumab), cetuzumab monoclonal antibody (daclizumab), eculizumab monoclonal antibody (escuzumab), eculizumab monoclonal antibody (efuzumab), efuzumab monoclonal antibody (efuzumab), ezetimibe monoclonal antibody (ezetimibe monoclonal antibody), ezetimibe monoclonal antibody (ezumab), ezetimibe monoclonal antibody (ezumab), zeuzumab monoclonal antibody (ezetimibe monoclonal antibody), zeuzumab monoclonal antibody (ezetimibe monoclonal antibody), zeuzumab), zetasizu monoclonal antibody (zeuzumab), zetasizu monoclonal antibody (zetasin monoclonal antibody (zetasi, Lintuzumab (lintuzumab), matuzumab (matuzumab), mepoppy monoclonal antibody (mepolizumab), motavizumab (motavizumab), natalizumab (motavizumab), nimustizumab (nimovizumab), norovizumab (nolovizumab), numazumab (numumab), aurilizumab (ocrelizumab), omalizumab (omalizumab), palivizumab (palivizumab), pascolizumab (cfpeutuzumab), pertuzumab (ctpezumab), pertuzumab (pertuzumab), pertuzumab (pervezumab), peglizumab (perlizumab), peglizumab (pexizumab), peglizumab (raplizumab), trastuzumab (rifugab), rituzumab (rituximab), rituximab (rituximab), rituzumab (rituximab), rituximab (rituximab), rituximab (renifob), rituximab (renlizumab), rituzumab), rituximab (rituximab), rituximab (rituximab) and (rituximab), rituximab (rituximab), and (rituximab), and (Rispe, rituximab), and (Rispe-, The antibody may be selected from the group consisting of a pintuzumab monoclonal antibody (soutuzumab), taclizumab monoclonal antibody (tesetant), taclizumab monoclonal antibody (taducizumab), talilizumab monoclonal antibody (talizumab), tefilzumab monoclonal antibody (tefibumab), tollizumab monoclonal antibody (tocilizumab), tollizumab monoclonal antibody (toralizumab), trastuzumab monoclonal antibody (trastuzumab), simon interleukin monoclonal antibody (tutuzumab celletin), tussizumab (tussizumab), umuzumab (umavuvivumab), umuzumab (urtuximab), and vesizumab (visilizumab).

The present invention further provides compositions comprising the subject compounds, stereoisomers thereof, and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier.

Compositions comprising the subject compounds, stereoisomers thereof, and pharmaceutically acceptable salts thereof may be administered in a variety of known ways, such as orally, topically, rectally, parenterally, by inhalation spray, or by implantable reservoirs. The most suitable route in any given case will depend on the particular host, and the nature and severity of the active ingredient being administered. The term "parenteral" as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques. The compositions disclosed herein may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art.

The compounds of the present invention and stereoisomers thereof and pharmaceutically acceptable salts thereof, may be administered orally in solid dosage forms such as capsules, tablets, lozenges, dragees, granules and powders; or orally in liquid dosage forms such as elixirs, syrups, emulsions, dispersions and suspensions. The subject compounds disclosed in this specification, stereoisomers thereof, and pharmaceutically acceptable salts thereof, may also be administered parenterally in sterile liquid dosage forms, such as dispersions, suspensions, or solutions. Other dosage forms may also be used to administer the subject compounds, stereoisomers thereof and pharmaceutically acceptable salts thereof, e.g. as ointments, creams, drops, transdermal patches or powders for topical administration; as ophthalmic solutions or suspensions, i.e., eye drops for ocular administration; aerosol spray or powder compositions for inhalation or intranasal administration; or a cream, ointment, spray or suppository for rectal or vaginal administration.

In addition, the compounds disclosed in the present specification and/or at least one pharmaceutically acceptable salt thereof and a powdered carrier, such as, but not limited to, lactose, starch, cellulose derivatives, magnesium stearate and stearic acid, may also be encapsulated in a gelatin capsule. Similar diluents can be used to prepare compressed tablets. Both tablets and capsules can be made into sustained release products to release the drug continuously over a period of time. Compressed tablets may be sugar-coated or film-coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric-coated to selectively disintegrate in the gastrointestinal tract.

Liquid dosage forms for oral administration may further comprise at least one pharmaceutical agent selected from coloring and flavoring agents to increase patient acceptance.

Generally, water, suitable oils, saline, aqueous dextrose (glucose) and related sugar solutions, and glycols, such as propylene glycol or polyethylene glycols, may be examples of suitable carriers for parenteral solutions. Solutions for parenteral administration may comprise at least one water-soluble salt of a compound described herein, at least one suitable stabilizer, and, if desired, at least one buffer substance. Antioxidants, such as sodium bisulfite, sodium sulfite, or ascorbic acid, alone or in combination, may be examples of suitable stabilizers. Citric acid and its salts and sodium edetate may also be used as examples of suitable stabilizers. Furthermore, the parenteral solution may further comprise at least one preservative, for example selected from benzalkonium chloride, methyl paraben, propyl paraben and chlorobutanol.

The "pharmaceutically acceptable carrier" described herein may be selected from carriers that are compatible with the active ingredients of the composition (and in some embodiments are capable of stabilizing the active ingredients) and not deleterious to the subject to be treated. For example, a solubilizing agent, such as cyclodextrin (which can form a specific more soluble complex with at least one compound and/or at least one pharmaceutically acceptable salt disclosed herein) can be used as a pharmaceutical excipient to deliver an active ingredient. Examples of other carriers include colloidal silicon dioxide, magnesium stearate, cellulose, sodium lauryl sulfate, and pigments such as D & C Yellow # 10. Suitable pharmaceutically acceptable carriers are referred to Remington's Pharmaceutical Sciences, a.osol, which is a standard reference in the art.

The compounds of interest, stereoisomers thereof and pharmaceutically acceptable salts thereof disclosed herein can be further validated for their efficacy in treating ADAM 9-associated diseases (conditions in which ADAM9 activity is overexpressed in one or more cell types and is pathogenic) by in vivo (in vivo) assays. For example, the compounds disclosed herein and/or at least one pharmaceutically acceptable salt thereof can be administered to an animal (e.g., a mouse model) having a disease associated with ADAM9, and a therapeutic effect can be obtained. A positive result in one or more tests is sufficient to demonstrate the actual utility of the tested compound and/or salt. And the appropriate dosage range and route of administration for an animal (e.g., a human) can be determined based on the results of the assay.

For administration by inhalation, the compounds of the present invention, stereoisomers thereof and pharmaceutically acceptable salts thereof may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer. The compounds of the present invention, stereoisomers thereof and pharmaceutically acceptable salts thereof may also be delivered in powder form, which may be formulated and the powder composition may be inhaled by means of an insufflation powder inhaler device. One exemplary delivery system for inhalation may be a Metered Dose Inhalation (MDI) aerosol, which may be formulated as a suspension or solution of the subject compounds disclosed herein, stereoisomers thereof, and pharmaceutically acceptable salts thereof in at least one suitable propellant, such as fluorocarbons and hydrocarbons.

For ophthalmic administration, ophthalmic formulations can be formulated with a suitable ophthalmic vehicle with a solution or suspension of the subject compound, its stereoisomers, and pharmaceutically acceptable salts thereof in an appropriate weight percentage such that the compound, its stereoisomers, and at least one pharmaceutically acceptable salt thereof, remains with the surface of the eye for a sufficient time to allow the compound to penetrate the cornea and interior regions of the eye.

Useful pharmaceutical dosage forms for administering the compounds of the present invention, stereoisomers thereof, and pharmaceutically acceptable salts thereof include, but are not limited to, hard and soft gelatin capsules, tablets, parenteral injections, and oral suspensions.

The dose administered will depend upon factors including the age of the subject, the health and weight of the subject, the extent of the disease, the type of concurrent treatment (if any), the frequency of treatment and the nature of the effect desired. In general the daily dose of the active ingredient may be non-quantitative, for example 0.1 to 2000mg per day. For example, 10-500mg once or more daily may be effective to achieve the desired result.

In some embodiments, a large number of unit capsules can be prepared by filling standard two-piece hard gelatin capsules, each having, for example, 100mg of a compound of the present invention, its stereoisomers and pharmaceutically acceptable salts thereof, and other pharmaceutically acceptable excipients, such as lactose, cellulose and magnesium stearate.

In some embodiments, a mixture containing a compound of the present invention, its stereoisomers and pharmaceutically acceptable salts thereof, is prepared into a digestible oil, such as soybean oil, cottonseed oil or olive oil, and injected by positive displacement pumping into gelatin to form soft gelatin capsules containing 100mg of the active ingredient, which are then washed and dried.

In some embodiments, a plurality of tablets may be prepared by conventional methods such that a dosage unit contains, for example, 100mg of a compound of the present invention, stereoisomers thereof and pharmaceutically acceptable salts thereof, as well as other pharmaceutically acceptable excipients, such as colloids, silicon dioxide, magnesium stearate, microcrystalline cellulose, starch and lactose. And may be provided with a suitable coating to increase palatability or delay absorption.

In some embodiments, parenteral compositions suitable for administration by injection may be prepared by stirring 1.5 weight percent of a compound of the invention and/or at least its enantiomers, diastereomers, or pharmaceutically acceptable salts in 10 volume percent propylene glycol. And the solution is brought to the desired volume with water for injection and sterilized.

In some embodiments, aqueous suspensions may be prepared for oral administration. For example, the aqueous suspension contains 100mg of finely divided compound, its stereoisomers and pharmaceutically acceptable salts, and other pharmaceutically acceptable excipients, such as sodium carboxymethyl cellulose, sodium benzoate, sorbitol solution, U.S. p. and vanillin, per 5 ml of aqueous suspension.

When the compounds of the present invention, stereoisomers thereof and pharmaceutically acceptable salts thereof are administered stepwise or in combination with at least one other therapeutic agent, the same dosage forms may generally be used. When the drugs are administered in physical combination, the dosage form and route of administration should be selected according to the compatibility of the combined drugs. Thus, co-administration as described herein is understood to encompass the concomitant or sequential administration of at least two agents, or as a fixed dose combination of at least two active ingredients.

The compounds disclosed in the present specification, stereoisomers and pharmaceutically acceptable salts thereof, may be administered as the sole active ingredient or in combination with at least one second active ingredient selected from other active ingredients known, for example, to be useful in the treatment of patients with ADAM 9-related diseases.

23页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:ASH1L抑制剂及用其进行治疗的方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!