ASH1L inhibitors and methods of treatment therewith

文档序号:1145037 发布日期:2020-09-11 浏览:20次 中文

阅读说明:本技术 Ash1l抑制剂及用其进行治疗的方法 (ASH1L inhibitors and methods of treatment therewith ) 是由 J·格雷姆贝卡 S·克洛索夫斯基 邓菁 T·塞皮基 李�昊 苗宏志 T·普罗希特 E· 于 2018-11-09 设计创作,主要内容包括:本文提供了与ASH1L结合并抑制ASH1L活性的小分子,及其用于治疗疾病(包括急性白血病、实体癌及依赖于ASH1L的活性的其它疾病)的使用方法。(Provided herein are small molecules that bind to ASH1L and inhibit the activity of ASH1L, and methods of use thereof for treating diseases including acute leukemia, solid cancers, and other diseases that depend on the activity of ASH 1L.)

1. A compound comprising the structure:

or a salt thereof,

wherein R is1、R2-5、R6、R7And X is selected from any combination of suitable substituents and moieties described and/or shown herein.

2. A compound comprising the structure:

or a salt thereof,

wherein R is1And R6Selected from any combination of suitable substituents and moieties described and/or shown herein.

3. The compound of one of claims 1-2, selected from compounds 21-150.

4. The compound of one of claims 1-2, wherein R1、R2-5、R6、R7And X is selected from said R of compounds 21-85 in any combination1、R2-5、R6、R7And an X group.

5. A pharmaceutical composition comprising a compound of any one of the preceding claims and a pharmaceutically acceptable carrier.

6. The pharmaceutical composition of claim 5, wherein the pharmaceutical composition is formulated for oral administration.

7. The pharmaceutical composition of claim 5, wherein the pharmaceutical composition is formulated for injection.

8. A method of inhibiting ASH1L, comprising contacting ASH1L with an effective amount of a compound of one of claims 1-4 or a pharmaceutical composition of one of claims 5-7.

9. The method of claim 8, wherein ASH1L activity is inhibited by binding of the compound or pharmaceutical composition to ASH 1L.

10. A method of treating a disease comprising administering to a subject a pharmaceutical composition of one of claims 5-7 in an amount effective to inhibit the activity of ASH 1L.

11. The method of claim 10, wherein the disease is cancer.

12. The method of claim 11, wherein the disease or condition comprises leukemia, hematologic malignancies, solid tumor cancer, breast cancer, prostate cancer, ovarian cancer, liver cancer, or thyroid cancer.

13. The method of claim 12, wherein the disease or condition comprises AML, ALL, mixed lineage leukemia, or leukemia with partial tandem repeats of MLL.

14. A method of treating a disorder mediated by a chromosomal rearrangement on chromosome 11q23 in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of the pharmaceutical composition of any one of claims 5-7.

15. The method of claim 14, wherein the pharmaceutical composition is co-administered with an additional therapeutic agent.

16. The method of claim 14, wherein the subject is a human.

17. Use of a composition of compounds according to one of claims 1 to 4 or a pharmaceutical composition according to one of claims 5 to 7 for the treatment of a disease.

Technical Field

Provided herein are small molecules that bind to ASH1L and inhibit the activity of ASH1L, and methods of use thereof for treating diseases including acute leukemia, solid cancers, and other diseases that depend on the activity of ASH 1L.

Background

ASH1L (deletion small and homeotic disc protein 1 homologue; EC: 2.1.1.43) is histone-lysine N-methyltransferase (KMTase) that methylates histone 3 lysine 36(H3K 36). ASH1L is essential for chromatin association of MLL fusion proteins and MLL fusion protein-mediated oncogenic transformation on key leukemia target genes, which means that ASH1L represents a therapeutic target in MLL leukemias and other leukemias with high HOX expression (ref.1; incorporated by reference in its entirety). ASH1L is also overexpressed in a variety of solid tumors, including thyroid and breast cancers (references 2, 3; incorporated by reference in its entirety). In thyroid cancer, ASH1L is overexpressed in a tumor-specific truncated form. The tumor suppressor microRNAmiR-142-3 p inhibits ASH1L protein expression by binding to the 3' UTR of ASH1L 3, a role associated with inhibition of colony formation and growth slowing of thyroid cancer cells (ref.2; incorporated by reference in its entirety). Furthermore, the ASH1L gene often undergoes copy number amplification in invasive basal-like breast cancers, and high expression of ASH1L mRNA correlates with shorter survival in breast cancer patients (reference 3; incorporated by reference in its entirety). Finally, in hepatocellular carcinoma (HCC), structural variations were found near the ASH1L gene, and the knockdown of ASH1L in HCC cells slowed proliferation (reference 4; incorporated by reference in its entirety).

In a variety of developmental and oncogenic settings, ASH1L activates the HOXA-B, -C, and-D genes and MEIS1 (references 5-8; incorporated by reference in their entirety). The KMTase activity of ASH1L is required for at least some of its gene activation functions, as deletion of the ASH1L SET domain in differentiated mouse embryonic stem cells results in loss of expression of 152 genes, including members of the Hox and Wnt families (ref.8; incorporated by reference in its entirety). These findings are highly relevant, as the HOX gene is an oncogenic driver in many different blood and solid tumors (reference 9; incorporated by reference in its entirety). For example, overexpression of HOXA9 was highly correlated with poor prognosis in AML (reference 10; incorporated by reference in its entirety), and HOXA9 and its collaborator MEIS1 were necessary for MLL-rearranged leukemia cell survival (references 11, 12; incorporated by reference in its entirety). ASH1L deficiency resulted in a massive reduction of long-term Hematopoietic Stem Cells (HSCs) in mouse bone marrow, but had very limited effect on peripheral blood cell counts due to increased proliferation of progenitor cells downstream of HSCs (reference 5; incorporated by reference in its entirety). ASH1L also plays an important role in diseases other than cancer. For example, in facioscapulohumeral muscular dystrophy, ASH1L is recruited by non-coding RNA to the chromosomal region 4q35, where it causes H3K36 dimethylation, chromatin remodeling, and aberrant transcription of the 4q35 gene (reference 13; incorporated by reference in its entirety). In liver fibrosis, ASH1L is up-regulated during the differentiation transformation of hepatic stellate cells into fibrogenic myofibroblasts, and binds to and activates profibrogenic genes (ref.14; incorporated by reference in its entirety).

Disclosure of Invention

Provided herein are small molecules that bind to ASH1L and inhibit the activity of ASH1L, and methods of use thereof for treating diseases including acute leukemia, solid cancers, and other diseases that depend on the activity of ASH 1L.

In some embodiments, provided herein are compounds that bind to and inhibit the activity of ASH 1L. In some embodiments, the compounds herein comprise an ASH1L binding moiety. In some embodiments, binding of a compound herein to ASH1L inhibits ASH1L activity.

In some embodiments, provided herein are ASH1L inhibitory compounds comprising the structure of formula (IVa):

wherein R is1Selected from the group consisting of H, alkyl, substituted alkyl (e.g., halo-substituted alkyl), branched alkyl, substituted branched alkyl (e.g., halo-substituted branched alkyl), alkoxy, amine, substituted amine, thioalkyl, ketone, amide, substituted amide, cyano, sulfonyl, carboxyl, dialkylphosphine oxide, carbocyclic ring, substituted carbocyclic ring, aromatic ring, substituted aromatic ring, heterocyclic aromatic ring, substituted or unsubstituted heterocyclic non-aromatic ring (e.g., piperidine, methylpiperidine, bridged piperidine, tetrahydropyran, alkylsulfonyl-substituted piperidine, sulfonamide-substituted piperidine, 1- ((trifluoromethyl) sulfonyl) piperidine, difluorocyclohexane, monofluorocyclohexane, cyclohexane, substituted difluorocyclohexane, bicyclooctane, cycloheptane), carbocyclic or heterocyclic aromatic ring fused to another aromatic ring, hydrogen bond donor, hydrogen bond, hydroxyl, or hydroxyl, Hydrogen bond receptors and combinations thereof;

wherein R is2、R3、R4、R5And R7Independently selected from H, halogen (e.g., Cl, F, Br, I), CH3、OH、SH、NH2、CN、CF3、CCl3、-CH2-CH3、-CH2-OH、-CH2NH2、CH3SH、CH2Cl、CH2Br、CH2F、CHF2、CH2CN、CH2CF3、CH2Cl3Alkyl groupHaloalkyl and alcohol; and is

Wherein R is6Selected from the group consisting of H, alkyl, substituted alkyl, hydroxyl, alkoxy, amine, substituted amine, alkylamine, substituted alkylamine, thioalkyl, halogen, ketone, amide, substituted amide, alkylamide, substituted alkylamide, cyano, sulfonyl, carboxyl, dialkylphosphine oxide, carbocycle, substituted carbocycle, aromatic ring, substituted aromatic ring, heterocycloaromatic ring, substituted or unsubstituted heterocycloaromatic ring (e.g., azetidine), a carbocycle or heterocycloaromatic ring fused to another aromatic ring, hydrogen bond donor, hydrogen bond acceptor, and combinations thereof.

In some embodiments, R of formula (IVa)1And/or R6The substituents are independently one of the formulae (a-q):

formula (a):

formula (b):

formula (c):

Figure BDA0002573038890000043

formula (d):

Figure BDA0002573038890000044

formula (e):

Figure BDA0002573038890000051

formula (f):

Figure BDA0002573038890000052

formula (g):

Figure BDA0002573038890000053

formula (h):

formula (i):

Figure BDA0002573038890000055

formula (j):

and

formula (k):

Figure BDA0002573038890000057

formula (l):

formula (m):

formula (n):

formula (o):

Figure BDA0002573038890000064

formula (p):

and

formula (q):

j, Q therein1Or J1One of which, when present, is connected to the primary support;

each of which J, J1、J2、J3And J4When present, is independently selected from the group consisting of: covalent bond, H, alkyl1-15Alkenyl radical1-6Alkynyl group1-6、(CH2)0-6C(S)NH2、(CH2)0-6C(O)NH2、O、S、NH、(CH2)0-6C(O)NH(CH2)1-6、(CH2)0-6NHC(O)(CH2)1-6Alkylsulfonyl, sulfonamide, alkylsulfonamide, (CH)2)0-6C(S)NH(CH2)1-6、(CH2)0-6O(CH2)1-6、(CH2)0-6OH、(CH2)0-6S(CH2)1-6、(CH2)0-6SH、(CH2)0-6NH(CH2)1-6、(CH2)0-6N(CH2)1-6(CH2)1-6、(CH2)0-6NH2、(CH2)0-6SO2(CH2)1-6、(CH2)0-6NHSO2(CH2)1-6、(CH2)0-6SO2NH2Halogen (e.g., F, Cl, Br or I), haloalkyl (e.g., (CH)2)0-6CH2F、(CH2)0-3CHF(CH2)0-2CH3Or analogous groups to Br, Cl or I), dihaloalkyl (e.g., (CH)2)0-6CF2H、(CH2)0-3CF2(CH2)0-2CH3Or with Br, Cl or ILike groups of (i), trihaloalkyl (e.g., (CH))2)0-6CF3Or analogous groups to Br, Cl or I), alkyl having 1-3 halogens at two or more positions along its length, (CH)2)1-4SP(Ph)-2=S、(CH2)0-6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1- 5NH2、(CH2)0-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-5SH、(CH2)0-6O(CH2)1-6NH(CH2)1- 5OH、(CH2)0-6O(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6O(CH2)1-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-6O(CH2)1-5SH、(CH2)0-6O(CH2)1-6S(CH2)1-5OH、(CH2)0-6O(CH2)1-6S(CH2)1-5NH2、(CH2)0-6O(CH2)1-6S(CH2)1-5SH、(CH2)0-6S(CH2)1-6NH(CH2)1-5OH、(CH2)0-6S(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6S(CH2)1-6NH(CH2)1-5SH、(CH2)0-6S(CH2)1- 6O(CH2)1-5OH、(CH2)0-6S(CH2)1-6O(CH2)1-5NH2、(CH2)0-6S(CH2)1-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-6S(CH2)1-5SH、(CH2)0-6NH(CH2)1- 6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6NH(CH2)1-5SH、(CH2)0-6NH(CH2)1-6O(CH2)1-5OH、(CH2)0-6NH(CH2)1-6O(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6O(CH2)1-5SH、(CH2)0- 6NH(CH2)1-6S(CH2)1-5OH、(CH2)0-6NH(CH2)1-6S(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6S(CH2)1-5SH、(CH2)0-3C(O)O(CH2)0-3、(CH2)0-3C(S)O(CH2)0-3、(CH2)0-3C(O)S(CH2)0-3、(CH2)0-3C(S)S(CH2)0-3、(CH2)0-3C(O)NH(CH2)0-3、(CH2)0-3C(S)NH(CH2)0-3、(CH2)0-3NHC(O)(CH2)0-3、(CH2)0-3NHC(S)(CH2)0-3、(CH2)0-3OC(O)(CH2)0-3、(CH2)0-3OC(S)(CH2)0-3、(CH2)0-3SC(O)(CH2)0-3、(CH2)0-3SC(S)(CH2)0-3、(CH2)0-3NHC(O)NH(CH2)0-3、(CH2)0-3NHC(S)NH(CH2)0-3、(CH2)0-3OC(O)NH(CH2)0-3、(CH2)0-3OC(S)NH(CH2)0-3、(CH2)0-3SC(O)NH(CH2)0-3、(CH2)0-3SC(S)NH(CH2)0-3、(CH2)0-3NHC(O)O(CH2)0-3、(CH2)0-3NHC(S)O(CH2)0-3、(CH2)0-3OC(O)O(CH2)0-3、(CH2)0-3OC(S)O(CH2)0-3、(CH2)0- 3SC(O)O(CH2)0-3、(CH2)0-3SC(S)O(CH2)0-3、(CH2)0-3NHC(O)S(CH2)0-3、(CH2)0-3NHC(S)S(CH2)0-3、(CH2)0-3OC(O)S(CH2)0-3、(CH2)0-3OC(S)S(CH2)0-3、(CH2)0-3SC(O)S(CH2)0-3、(CH2)0-3SC(S)S(CH2)0-3、(CH2O)1-6And trimethyl methane;

each of which Q, Q1And Q2When present, is independently selected from the group consisting of: furan, benzofuran, isobenzofuran, pyrrole, indole, isoindole, thiophene, benzothiophene, benzo [ c]Thiophene, imidazole, benzimidazole, purine, pyrazole, indazole, oxazole, benzoxazoleOxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, benzene, naphthalene, pyridine, quinolone, isoquinoline, pyrazine, quinoxaline, pyrimidine, quinazoline, pyridazine, cinnoline, phthalazine, thalidomide, triazine (e.g., 1,2, 3-triazine; 1,2, 4-triazine; 1,3, 5-triazine), thiadiazole, aziridine, thiirane (episulfide), oxirane (ethylene oxide, epoxide), oxaziridine, dioxirane, azetidine, oxetane, thietane, diazetidine, dioxetane, dithiolane, pyrrolidine, tetrahydrofuran, thiacyclopentane, imidazolidine, pyrazolidine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, piperidine, azetidine, dioxane, thiane, piperazine, pyridine, and pyridine, Morpholine, thiomorpholine, dioxane, dithiane, trioxane, trithiane (thithithaane), azepane, oxepane, thiepane, homopiperazine, azocane, tetrahydropyran, cyclobutene, cyclopentene, cyclohexene, cycloheptene, 1, 3-cyclohexadiene, 1, 4-cyclohexadiene, 1, 5-cyclooctadiene, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, any suitable C3-C7Cycloalkyl groups and any ring structures shown in table 4;

each of which Q, Q1And Q2When present, may exhibit one or more additional J groups at any position on the Q ring;

wherein any of the above alkyl groups or (CH)2)x-yThe groups may be linear or branched;

wherein any of the above alkyl groups or (CH)2)x-yThe radicals may additionally contain OH on one or more carbons, ═ O, NH2CN, dihaloalkyl (e.g. CF)2H) Trihaloalkyl (e.g., CF)3) Or a halogen (e.g., F) substituent; and is

Wherein the number of hydrogens at the terminal position of the above group can be adjusted in the case where the group is linked to another group (e.g., CH)3Is adjusted to CH2OH to O, etc.) or the group is terminally adjusted (e.g., CH)2Is adjusted to CH3O is adjusted to OH, etc.).

TABLE 4 non-limiting examples of ring structures.

Figure BDA0002573038890000091

Figure BDA0002573038890000111

In some embodiments, provided herein are ASH1L inhibitory compounds comprising the structure of formula (IVb):

Figure BDA0002573038890000122

wherein R is1Selected from the group consisting of H, alkyl, substituted alkyl (e.g., halo-substituted alkyl), branched alkyl, substituted branched alkyl (e.g., halo-substituted branched alkyl), alkoxy, amine, substituted amine, thioalkyl, ketone, amide, substituted amide, cyano, sulfonyl, carboxyl, dialkylphosphine oxide, carbocyclic ring, substituted carbocyclic ring, aromatic ring, substituted aromatic ring, heterocyclic aromatic ring, substituted or unsubstituted heterocyclic non-aromatic ring (e.g., piperidine, methylpiperidine, bridged piperidine, tetrahydropyran, alkylsulfonyl-substituted piperidine, sulfonamide-substituted piperidine, 1- ((trifluoromethyl) sulfonyl) piperidine, difluorocyclohexane, monofluorocyclohexane, cyclohexane, substituted difluorocyclohexane, bicyclooctane, cycloheptane), carbocyclic or heterocyclic aromatic ring fused to another aromatic ring, hydrogen bond donor, hydrogen bond, hydroxyl, or hydroxyl, Hydrogen bond receptors and combinations thereof; and is

Wherein R is6Selected from H, alkyl, substituted alkyl, hydroxyl, alkoxy, amine, substituted amine, alkylamineSubstituted alkylamines, thioalkyl groups, halogens, ketones, amides, substituted amides, alkylamides, substituted alkylamides, cyano groups, sulfonyl groups, carboxyl groups, dialkylphosphine oxides, carbocycles, substituted carbocycles, aromatic rings, substituted aromatic rings, heterocyclic aromatic rings, substituted or unsubstituted heterocyclic non-aromatic rings (e.g., azetidines), carbocyclic or heterocyclic aromatic rings fused to another aromatic ring, hydrogen bond donors, hydrogen bond acceptors, and combinations thereof.

In some embodiments, R1And/or R6The substituents are independently one of formula (a-q);

formula (a):

formula (b):

formula (c):

formula (d):

formula (e):

formula (f):

Figure BDA0002573038890000136

formula (g):

Figure BDA0002573038890000141

formula (h):

Figure BDA0002573038890000142

formula (i):

formula (j):

and

formula (k):

Figure BDA0002573038890000145

formula (l):

formula (m):

Figure BDA0002573038890000151

formula (n):

Figure BDA0002573038890000152

formula (o):

formula (p):

and

formula (q):

Figure BDA0002573038890000155

j, Q therein1Or J1One of which, when present, is connected to the primary support;

each of which J, J1、J2、J3And J4When present, is independently selected from the group consisting of: covalent bond, H, alkyl1-15Alkenyl radical1-6Alkynyl group1-6、(CH2)0-6C(S)NH2、(CH2)0-6C(O)NH2、O、S、NH、(CH2)0-6C(O)NH(CH2)1-6、(CH2)0-6NHC(O)(CH2)1-6Alkylsulfonyl, sulfonamide, alkylsulfonamide, (CH)2)0-6C(S)NH(CH2)1-6、(CH2)0-6O(CH2)1-6、(CH2)0-6OH、(CH2)0-6S(CH2)1-6、(CH2)0-6SH、(CH2)0-6NH(CH2)1-6、(CH2)0-6N(CH2)1-6(CH2)1-6、(CH2)0-6NH2、(CH2)0-6SO2(CH2)1-6、(CH2)0-6NHSO2(CH2)1-6、(CH2)0-6SO2NH2Halogen (e.g., F, Cl, Br or I), haloalkyl (e.g., (CH)2)0-6CH2F、(CH2)0-3CHF(CH2)0-2CH3Or analogous groups to Br, Cl or I), dihaloalkyl (e.g., (CH)2)0-6CF2H、(CH2)0-3CF2(CH2)0-2CH3Or analogous groups to Br, Cl or I), trihaloalkyl (e.g., (CH)2)0-6CF3Or like Br, Cl or I), having 1-3 halogens at two or more positions along its lengthAlkyl group, (CH)2)1-4SP(Ph)-2=S、(CH2)0-6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1- 5NH2、(CH2)0-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-5SH、(CH2)0-6O(CH2)1-6NH(CH2)1- 5OH、(CH2)0-6O(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6O(CH2)1-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-6O(CH2)1-5SH、(CH2)0-6O(CH2)1-6S(CH2)1-5OH、(CH2)0-6O(CH2)1-6S(CH2)1-5NH2、(CH2)0-6O(CH2)1-6S(CH2)1-5SH、(CH2)0-6S(CH2)1-6NH(CH2)1-5OH、(CH2)0-6S(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6S(CH2)1-6NH(CH2)1-5SH、(CH2)0-6S(CH2)1- 6O(CH2)1-5OH、(CH2)0-6S(CH2)1-6O(CH2)1-5NH2、(CH2)0-6S(CH2)1-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-6S(CH2)1-5SH、(CH2)0-6NH(CH2)1- 6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6NH(CH2)1-5SH、(CH2)0-6NH(CH2)1-6O(CH2)1-5OH、(CH2)0-6NH(CH2)1-6O(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6O(CH2)1-5SH、(CH2)0- 6NH(CH2)1-6S(CH2)1-5OH、(CH2)0-6NH(CH2)1-6S(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6S(CH2)1-5SH、(CH2)0-3C(O)O(CH2)0-3、(CH2)0-3C(S)O(CH2)0-3、(CH2)0-3C(O)S(CH2)0-3、(CH2)0-3C(S)S(CH2)0-3、(CH2)0-3C(O)NH(CH2)0-3、(CH2)0-3C(S)NH(CH2)0-3、(CH2)0-3NHC(O)(CH2)0-3、(CH2)0-3NHC(S)(CH2)0-3、(CH2)0-3OC(O)(CH2)0-3、(CH2)0-3OC(S)(CH2)0-3、(CH2)0-3SC(O)(CH2)0-3、(CH2)0-3SC(S)(CH2)0-3、(CH2)0-3NHC(O)NH(CH2)0-3、(CH2)0-3NHC(S)NH(CH2)0-3、(CH2)0-3OC(O)NH(CH2)0-3、(CH2)0-3OC(S)NH(CH2)0-3、(CH2)0-3SC(O)NH(CH2)0-3、(CH2)0-3SC(S)NH(CH2)0-3、(CH2)0-3NHC(O)O(CH2)0-3、(CH2)0-3NHC(S)O(CH2)0-3、(CH2)0-3OC(O)O(CH2)0-3、(CH2)0-3OC(S)O(CH2)0-3、(CH2)0- 3SC(O)O(CH2)0-3、(CH2)0-3SC(S)O(CH2)0-3、(CH2)0-3NHC(O)S(CH2)0-3、(CH2)0-3NHC(S)S(CH2)0-3、(CH2)0-3OC(O)S(CH2)0-3、(CH2)0-3OC(S)S(CH2)0-3、(CH2)0-3SC(O)S(CH2)0-3、(CH2)0-3SC(S)S(CH2)0-3、(CH2O)1-6And trimethyl methane;

each of which Q, Q1And Q2When present, is independently selected from the group consisting of: furan, benzofuran, isobenzofuran, pyrrole, indole, isoindole, thiophene, benzothiophene, benzo [ c]Thiophene, imidazole, benzimidazole, purine, pyrazole, indazole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, benzene, naphthalene, pyridine, quinolone, isoquinoline, pyrazine, quinoxaline, pyrimidine, quinazoline, pyridazine, cinnoline, phthalazine, thalidomide, triazine(e.g., 1,2, 3-triazine; 1,2, 4-triazine; 1,3, 5-triazine), thiadiazole, aziridine, epithioethane (episulfide), oxirane (ethylene oxide, epoxide), oxaziridine, diepoxide, azetidine, oxetane, thietane, diazetidine, dioxetane, dithiolane, pyrrolidine, tetrahydrofuran, thietane, imidazolidine, pyrazolidine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, piperidine, azetidine, dioxane, thiane, piperazine, morpholine, thiomorpholine, dioxane, dithiane, trioxane, trithiane, azepane, oxepane, thiepane, homopiperazine, azocane, tetrahydropyran, cyclobutene, cyclopentene, Cyclohexene, cycloheptene, 1, 3-cyclohexadiene, 1, 4-cyclohexadiene, 1, 5-cyclooctadiene, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, any suitable C3-C7Cycloalkyl groups and any ring structures shown in table 4;

each of which Q, Q1And Q2When present, may exhibit one or more additional J groups at any position on the Q ring;

wherein any of the above alkyl groups or (CH)2)x-yThe groups may be linear or branched;

wherein any of the above alkyl groups or (CH)2)x-yThe radicals may additionally contain OH on one or more carbons, ═ O, NH2CN, dihaloalkyl (e.g. CF)2H) Trihaloalkyl (e.g., CF)3) Or a halogen (e.g., F) substituent; and is

Wherein the number of hydrogens at the terminal position of the above group can be adjusted in the case where the group is linked to another group (e.g., CH)3Is adjusted to CH2OH to O, etc.) or the group is terminally adjusted (e.g., CH)2Is adjusted to CH3O is adjusted to OH, etc.).

In some embodiments, R of formula (IVa) and/or (IVb)1And R6The substituents are independently any substituent present on compounds 21-85Groups, as shown in Table 9, but not limited to, the positions of substituents on compounds 21-85.

Table 9 exemplary compounds of formula (IVb).

Figure BDA0002573038890000181

Figure BDA0002573038890000201

Figure BDA0002573038890000241

Figure BDA0002573038890000271

Figure BDA0002573038890000281

Figure BDA0002573038890000311

Figure BDA0002573038890000321

Figure BDA0002573038890000351

In some embodiments, the compound is selected from the compounds shown in table 9 (e.g., compounds 21-85).

In some embodiments, provided herein are compounds defined by one of formulas (IVa) and (IVb), but wherein the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety (as shown in formulas (IVa) and (IVb)). For example, for any formula (e.g., formulae (IVa) and (IVb)) or compound (e.g., compounds 21-85) described herein having benzothioamide-pyrrole or benzamido-pyrrole ring connectivity, such as:

provided herein are corresponding formulae (e.g., formulae (IVc) and (IVd)) and compounds (e.g., compounds 86-150) having a benzothioamide-benzene or benzamido-benzene ring linkage, such as:

and are within the scope of the embodiments herein. For example, any of the embodiments, substituents, compounds, etc., described herein in connection with formula (IVa) may also be provided in embodiments in connection with formula (IVc):

and is

Any of the embodiments, substituents, compounds, etc., described herein in connection with formula (IVb) may also be provided in embodiments herein in connection with formula (IVd):

compounds 86-150 correspond to compounds 21-85, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety. For example, compound 86 corresponds to compound 21, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; compound 87 corresponds to compound 22, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; compound 88 corresponds to compound 23, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; and so on.

In some embodiments, provided herein are pharmaceutical compositions comprising a compound described herein and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated for oral administration, injection, or any other suitable route of administration.

In some embodiments, provided herein are methods of inhibiting the activity of ASH1L, comprising contacting ASH1L with an effective amount of a compound described herein.

In some embodiments, provided herein are methods of treating a disease comprising administering to a subject a pharmaceutical composition described herein in an amount effective to inhibit the activity of ASH 1L. In some embodiments, the disease is cancer. In some embodiments, the disease is a proliferative disorder. In some embodiments, the pharmaceutical composition is co-administered with an additional cancer therapeutic. In some embodiments, the subject is a human.

In some embodiments, provided herein is the use of a compound described herein. In some embodiments, provided herein is the use of a compound described herein for inhibiting activity of ASH 1L. In some embodiments, provided herein is the use of a compound described herein for the treatment of a disease (e.g., cancer).

Drawings

Fig. 1. Compound 37 inhibits the enzymatic activity of ASH 1L. IC measured in Histone Methyltransferase (HMT) assay50Value (IC)50=0.75μM)

FIG. 2, FIGS. A-C. FIG. A is a drawing. The MTT assay showed that compound 22 was effective on MLL leukemia cells 14 days after treatment: growth inhibition by MOLM13 and KOPN 8. K562(CML cell line) represents a negative control cell line. And (B). Compound 34 inhibited the proliferation of the MLL leukemia cell line (MV 4; 11, SEM, KOPN8, MOLM13), but not the control cell line K562. GI measurement in MTT cell viability assay after 14 days of treatment with Compound 3450The value is obtained. And (C). After 7 days of treatment, compound 22 was purified at MV 4; 11 inducing the differentiation in the cells of the species,

fig. 3. Compound 22 down-regulated the expression of MLL fusion target genes (HOXA gene, MEIS1, MEF2C) and increased the expression level of MNDA differentiation markers in a dose-dependent manner.

Fig. 4. Compound 22 is non-toxic to normal human hematopoietic hCD34+ cells.

FIG. 5, FIGS. A-B. Compound 59 MV4 in MLL leukemia; 11 slow the progression of leukemia in vivo in a xenografted mouse model. FIG. A is a drawing. Quantification of bioluminescence levels in mice after 7 days treatment with compound 59(40mg/kg, i.p., q.d.) or vehicle (6-7 mice per group). And (B). MV4 after 21 days (n-6-7) treatment with compound 59(25mg/kg, i.p., b.i.d.) or vehicle; flow cytometric analysis of hCD45+ cells in the spleen of 11 mice showed that compound 59 inhibited leukemia progression in mice.

Definition of

Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the embodiments described herein, some preferred methods, compositions, devices, and materials are described herein. However, before the present materials and methods are described, it is to be understood that this invention is not limited to the particular molecules, compositions, methods or protocols described herein as these may vary according to routine experimentation and optimization. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only, and is not intended to limit the scope of the embodiments described herein.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In case of conflict, the present specification, including definitions, will control. Thus, the following definitions apply in the context of the embodiments described herein.

As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an ASH1L inhibitor" is a reference to one or more ASH1L inhibitors and equivalents thereof known to those skilled in the art, and so forth.

As used herein, the term "comprising" and its grammatical variations mean the presence of the recited features, elements, method steps, etc., but do not preclude the presence of additional features, elements, method steps, etc. Conversely, the term "consisting of …" and its language of variation indicates the presence of the recited features, elements, method steps, etc., but excludes any non-recited features, elements, method steps, etc., except for normally associated impurities. The phrase "consisting essentially of …" means that the recited features, elements, method steps, etc., as well as any additional features, elements, method steps that do not materially affect the basic properties of the composition, system, or method. Many of the embodiments herein are described in the open "inclusive" language. Such embodiments encompass embodiments having a closure "consisting of …" and/or "consisting essentially of …," which may alternatively be claimed or described in such language.

The chemical names of all substituents should be interpreted according to IUPAC and/or modified forms, where the functional groups within the substituents are read in the order in which they branch from the scaffold or main structure. For example, in a modified nomenclature, methyl-sulfonyl-propanol refers to CH2SO2CH2CH2CH2OH or:

as another example, according to the modified nomenclature, the methyl-amine substituents are:

Figure BDA0002573038890000392

and the amino-methyl substituents are:

Figure BDA0002573038890000401

the chemical names of all substituents should be interpreted according to IUPAC and/or modified nomenclature and with reference to the chemical structures shown and/or described herein.

As used herein, the term "subject" broadly refers to any animal, including, but not limited to, humans and non-human animals (e.g., dogs, cats, cows, horses, sheep, poultry, fish, crustaceans, etc.). As used herein, the term "patient" generally refers to a subject being treated for a disease or condition.

As used herein, the term "subject at risk for a disease" (e.g., "subject at risk for cancer") refers to a subject who has one or more risk factors for developing a disease (e.g., cancer). Depending on the particular disease, risk factors may include, but are not limited to, gender, age, genetic predisposition, environmental exposure, infection and past medical history, lifestyle, and the like.

As used herein, the term "effective amount" refers to an amount of a composition sufficient to achieve a beneficial or desired result. An effective amount may be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or route of administration.

As used herein, the term "administration" refers to the act of administering a drug, prodrug, or other agent or therapeutic treatment to a subject or to cells, tissues and organs in vivo, in vitro or ex vivo. Exemplary routes of administration to the human body can be through the subarachnoid space of the brain or spinal cord (intrathecal), ocular (ocular), oral (oral), dermal (topical or transdermal), nasal (nasal), pulmonary (inhalation), oral mucosal (buccal), ear, rectal, vaginal, by injection (e.g., intravenous, subcutaneous, intratumoral, intraperitoneal, etc.), and the like.

As used herein, the term "co-administration" refers to the administration of at least two agents (e.g., an ASH1L inhibitor and one or more additional therapeutic agents) or therapies to a subject. In some embodiments, the co-administration of two or more agents or therapies is simultaneous. In other embodiments, the first dose/therapy is administered prior to the second dose/therapy. It will be appreciated by those skilled in the art that the formulation and/or route of administration of the various agents or therapies used may vary. The appropriate dosage for co-administration can be readily determined by one skilled in the art. In some embodiments, when agents or therapies are co-administered, the respective agents or therapies are administered at a lower dose than is suitable for administration alone. Thus, co-administration is particularly desirable in embodiments where co-administration of an agent or therapy reduces the necessary dose of a potentially harmful (e.g., toxic) agent or when co-administration of two or more agents results in a subject being susceptible to the beneficial effects of one agent via co-administration of another agent.

As used herein, the term "pharmaceutical composition" refers to a combination of an active agent and an inert or active carrier, which makes the composition particularly suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.

The term "pharmaceutically acceptable" or "pharmacologically acceptable" as used herein refers to a composition that produces substantially no adverse reactions, such as toxicity, allergy, or immune reactions, when administered to a subject.

As used herein, the term "pharmaceutically acceptable carrier" refers to any standard pharmaceutical carrier, including, but not limited to, phosphate buffered saline solutions, water, emulsions (e.g., oil/water or water/oil emulsions) and various types of wetting agents, any and all solvents, dispersion media, coatings, sodium lauryl sulfate, isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like. The composition may also include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, e.g., Martin, Remington's Pharmaceutical Sciences, 15 th edition, Mack pub.

As used herein, the term "pharmaceutically acceptable salt" refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention that, when administered to a subject, is capable of providing a compound of the present invention or an active metabolite or residue thereof. As known to those skilled in the art, "salts" of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, p-toluenesulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic, and the like. Other acids, such as oxalic, while not per se pharmaceutically acceptable, may be useful in the preparation of intermediates useful in obtaining the compounds of the present invention and their pharmaceutically acceptable acid addition salts.

Examples of bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metals (e.g., magnesium), hydroxides, ammonia, and NW4 +Wherein W is C1-4Alkyl groups, and the like.

Example bag of saltIncluding but not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, fluoroheptanoate (flucoheptanoate), glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmitate (palmoate), pectate (pectate), persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds of the invention with suitable cations such as Na+、NH4 +And NW4 +Composite (wherein W is C1-4Alkyl), and the like.

For therapeutic use, salts of the compounds herein that are pharmaceutically acceptable are contemplated. However, salts of non-pharmaceutically acceptable acids and bases may also find use, for example, in the preparation or purification of pharmaceutically acceptable compounds.

As used herein, the term "instructions for administering the compound to a subject" and grammatical equivalents thereof include instructions for treating a condition using the compositions contained in the kit (e.g., providing a dosage, a route of administration, a decision tree for a treating physician to correlate patient-specific characteristics with a course of therapeutic action).

"amino" means-NH2And (4) partial.

"carbonyl" refers to a moiety of the formula-C (═ O) -.

"Carboxy/carboxyl" means-CO2And (4) a H part.

"cyano" refers to the moiety-CN.

"Hydroxy/hydroxyl" refers to the-OH moiety.

"imino" refers to an ═ NH moiety. Unless otherwise specified specifically in the specification, imino is optionally substituted.

"nitro" means-NO2And (4) partial.

"oxo" refers to an ═ O moiety.

"thio (Thioxo)" refers to the moiety S.

"acyl" refers to the group-C (═ O) RaWherein R isaSelected from the group consisting of: alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon), heteroalkyl, and heterocycloalkyl. Unless stated otherwise specifically in the specification, acyl is optionally substituted.

"alkyl" means a straight or branched hydrocarbon chain moiety consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C)1-C12Alkyl), preferably one to eight carbon atoms (C)1-C8Alkyl) or one to six carbon atoms (C)1-C6Alkyl) and is linked to the rest of the molecule by a single bond, for example methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1-dimethylethyl (tert-butyl), 3-methylhexyl, 2-methylhexyl, vinyl, prop-1-enyl, but-1-enyl, pent-1, 4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl and the like. Alkyl groups include alkenyl (one or more carbon-carbon double bonds) and alkynyl (one or more carbon-carbon triple bonds). Unless stated otherwise specifically in the specification, alkyl is optionally substituted.

"alkoxy" means a group of the formula-ORaWherein R isaIs an alkyl group as defined herein containing from one to twelve carbon atoms. Unless stated otherwise specifically in the specification, alkoxy is optionally substituted.

"alkylamino" refers to the formula-NHRaor-NRaRbWherein R isaAnd RbEach independently an alkyl group as defined herein containing from one to twelve carbon atoms. Unless stated otherwise specifically in the specification, alkylamino is optionally substituted.

"alkylaminoalkyl" refers to an alkyl moiety that contains at least one alkylamino substituent. The alkylamino substituent may be on a tertiary, secondary or primary carbon. Unless stated otherwise specifically in the specification, alkylaminoalkyl is optionally substituted.

"amide" or "amido" refers to a compound having the formula-C (═ O) NRaRbor-NRaC(=O)RbWherein R isaAnd RbEach independently selected from the group consisting of: hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon), heteroalkyl, and heterocycloalkyl, each of which moieties may itself be optionally substituted. In some embodiments, it is C1-C4An amido or amide group which includes an amide carbonyl group in the total carbon number of the group. Of amides-NRaRbR of (A) to (B)aRbOptionally together with the nitrogen to which they are attached to form a 4,5, 6 or 7 membered ring. Unless otherwise specifically stated in the specification, the amido group is optionally substituted.

"aminoalkyl" refers to an alkyl moiety comprising at least one amino substituent. The amino substituent may be on a tertiary, secondary or primary carbon. Unless otherwise specifically stated in the specification, aminoalkyl is optionally substituted.

"aminocarbonyl" refers to the formula-C (═ O) NRaRbWherein R isaAnd RbEach independently is H or alkyl. Unless otherwise specifically stated in the specification, aminocarbonyl is optionally substituted.

"aryl" means a hydrocarbon ring system moiety containing from 6 to 18 carbon atoms and at least one aromatic ring. For the purposes of the present invention, aryl moieties are monocyclic, bicyclic, tricyclic or tetracyclic ring systems, which may include fused or bridged ring systems. Aryl moieties include, but are not limited to, aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, phenanthrylene, and mixtures thereof,Fluoranthene, fluorene, asymmetric indacene, symmetric indacene, indane, indene, naphthalene, phenalene, phenanthrene, obsidian, pyrene and triphenylene. Unless otherwise specifically stated in the specification, the term "aryl" or the prefix "aryl-" (as in "aralkyl-")) Is meant to include optionally substituted aryl groups.

"aralkyl" means a group of the formula-Rb-RcWherein R isbIs an alkylene chain as defined herein, and RcIs one or more aryl moieties as defined herein, e.g., benzyl, diphenylmethyl, and the like. Unless otherwise specified specifically in the specification, aralkyl is optionally substituted.

"aralkylamino" refers to aralkyl-NRa-a moiety wherein RaIs H or alkyl. Unless otherwise specifically stated in the specification, an aralkylamino group is optionally substituted.

"aralkyloxy" refers to an aralkyl-O-moiety. Unless otherwise specifically stated in the specification, aralkyloxy is optionally substituted.

"arylamino" refers to-NRa-an aryl moiety, wherein RaIs H or alkyl. Unless stated otherwise specifically in the specification, arylamino is optionally substituted.

"aryloxy" refers to the-O-aryl moiety. Unless stated otherwise specifically in the specification, an aryloxy group is optionally substituted.

"Bicycloalkyl" refers to a moiety having two cycloalkyl moieties having two or more atoms in common. Cycloalkyl moieties are said to be "fused" if they have exactly two adjacent common atoms. Examples include, but are not limited to, bicyclo [3.1.0] hexyl, perhydronaphthyl, and the like. Cycloalkyl moieties are said to be "bridged" if they have more than two atoms in common. Examples include, but are not limited to, adamantyl, bicyclo [3.2.1] heptyl ("norbornyl"), bicyclo [2.2.2] octyl, and the like. Unless stated otherwise specifically in the specification, bicycloalkyl is optionally substituted.

"carboxyalkyl" means a radical of the formula-Rb-RcWherein R isbIs an alkylene chain as defined herein, and RcIs a carboxyl group as defined herein. Unless otherwise specifically stated in the specification, carboxyalkyl is optionally substituted.

"cyanoalkyl" means a group of formula-Rb-RcWherein R isbIs an alkylene chain as defined herein, and RcIs cyano as defined herein. Unless stated otherwise specifically in the specification, cyanoalkyl is optionally substituted.

"carbocycle" or "carbocyclic ring" refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic hydrocarbon moiety which may include fused or bridged ring systems having from three to fifteen carbon atoms, preferably from three to ten carbon atoms, including cycloalkyls, cycloalkenyls, and the like. "cycloalkyl" means a saturated, non-aromatic, monocyclic or polycyclic hydrocarbon moiety, which can include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably from three to ten carbon atoms. Monocyclic cycloalkyl moieties include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyl moieties include, for example, adamantyl, norbornyl, decahydronaphthyl, 7-dimethyl-bicyclo [2.2.1] heptanyl, and the like. "cycloalkenyl" is cycloalkyl groups that include one or more carbon-carbon double bonds within the ring, such as cyclopentenyl and cyclohexenyl. Unless stated otherwise specifically in the specification, cycloalkyl is optionally substituted.

"cycloalkylalkyl" means a compound of the formula-RbRdWherein R isbIs an alkylene chain as defined herein, and RdIs a cycloalkyl moiety as defined herein. Unless stated otherwise specifically in the specification, cycloalkylalkyl is optionally substituted.

"cycloalkylalkylamino" refers to cycloalkylalkyl-NRa-a moiety wherein RaIs H or alkyl, and wherein the cycloalkylalkyl moiety is attached to the nitrogen via a carbon atom, wherein the nitrogen acts as a linker connecting the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, cycloalkylalkylamino is optionally substituted.

"cycloalkylalkyloxy" refers to an-O-cycloalkylalkyl moiety in which the cycloalkylalkyl moiety is attached to oxygen via a carbon atom, with the oxygen serving as a linker to attach the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, cycloalkylalkyloxy is optionally substituted.

"Cycloalkylamino" refers to-NRa-a cycloalkyl moiety, wherein RaIs H or alkyl. Unless stated otherwise specifically in the specification, cycloalkylamino is optionally substituted.

"cycloalkyloxy" means an-O-cycloalkyl moiety. Unless stated otherwise specifically in the specification, cycloalkyloxy is optionally substituted.

"halo" or "halogen" refers to fluoro, chloro, bromo, or iodo.

"haloalkyl" refers to an alkyl group as defined herein substituted with one or more halogen atoms as defined herein, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trichloromethyl, -CH2CF3、-CH2CHF2、-CH2CH2F、-CHFCF3、-CHFCHF2、-CHFCH2F、-CHFCH3、-CF2CF3、-CF2CHF2、-CF2CH2F、-CF2CH3、-CH2CF2CH3、-CH2CHFCH33-bromo-2-fluoropropyl, 1, 2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, haloalkyl is optionally substituted.

As used herein, the term "heteroatom" or "ring heteroatom" is meant to include any element other than carbon or hydrogen. Preferred heteroatoms are oxygen (O), nitrogen (N), sulfur (S) and phosphorus (P).

Unless otherwise specified, "heteroalkyl," by itself or in combination with another term, means a straight or branched chain containing at least one carbon atom and at least one heteroatom such as O, N, P, Si and S; a monocyclic or polycyclic moiety, which may comprise a fused or bridged ring system; or any combination thereof, wherein one or more of the heteroatoms may be oxidized. The heteroatom may be in the alkyl moiety, e.g. -CH2-O-CH2-; at the point of attachment to the rest of the molecule, e.g. -SO2CH(CH3)CH2-; or combinations thereof, e.g. -NH2CH2CH2SO2CH2-. Unless otherwise stated specifically in the specification, heteroalkyl is optionalAnd (3) substituted.

"heteroaryl" means a 5 to 14 membered ring system moiety containing one to thirteen carbon atoms; one to six heteroatoms, such as nitrogen, oxygen and sulfur; and one or more rings, wherein at least one ring is aromatic. For the purposes of the present invention, heteroaryl groups may be monocyclic, bicyclic, tricyclic or tetracyclic ring systems, which may include fused or bridged ring systems, and one or more heteroatoms may be oxidized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl (benzodioxolyl), benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzo [ b ] [1,4] dioxepinyl, 1, 4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranonyl, benzothiophenyl (benzothiophenyl), benzotriazolyl, benzo [4,6] imidazo [1,2-a ] pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, etc, Indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridyl, 1-oxidopyrimidinyl, 1-oxidopyridyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalyl, quinolyl, quinuclidinyl, isoquinolyl, tetrahydroquinolyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e., thienyl). Unless stated otherwise specifically in the specification, heteroaryl is optionally substituted.

"Heteroarylalkyl" means a compound of the formula-RbRfWherein R isbIs an alkylene chain as defined herein, and RfIs heteroaryl as defined herein. Unless otherwiseAs further specified in the specification, heteroarylalkyl is optionally substituted.

"Heteroarylalkylamino" refers to heteroarylalkyl-NRa-a moiety wherein RaIs H or alkyl. Unless stated otherwise specifically in the specification, heteroarylalkylamino is optionally substituted.

"Heteroarylalkyloxy" means a heteroarylalkyl-O-moiety. Unless stated otherwise specifically in the specification, heteroarylalkyloxy is optionally substituted.

"Heteroarylamino" means-NRa-a heteroaryl moiety, wherein RaIs H or alkyl. Unless stated otherwise specifically in the specification, heteroarylamino is optionally substituted.

"heteroaryloxy" means an-O-heteroaryl moiety. Unless stated otherwise specifically in the specification, heteroaryloxy is optionally substituted.

"heterobicycloalkyl" refers to a bicycloalkyl structure in which at least one carbon ring atom is replaced with a heteroatom such as oxygen, nitrogen and sulfur. Unless stated otherwise specifically in the specification, heterobicycloalkyl is optionally substituted.

"heterocyclyl" or "heterocyclic ring" refers to a 3 to 18 membered non-aromatic ring consisting of two to twelve carbon atoms and one to six heteroatoms, such as nitrogen, oxygen, and sulfur. Unless stated otherwise specifically in the specification, heterocyclyl is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; the heteroatom may optionally be oxidized; and the heterocyclic group may be unsaturated or saturated. Examples of such heterocyclyl moieties include, but are not limited to, dioxolanyl, thienyl [1,3] dithianyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidinonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuranyl, trithianyl, tetrahydropyranyl, thiomorpholinyl (thiomorpholinyl), 1-oxo-thiomorpholinyl, and 1, 1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, heterocyclyl is optionally substituted.

"Heterocyclylalkyl" or "heterocycloalkyl" refers to the formula-RbReWherein R isbIs an alkylene chain as defined herein, and ReIs a heterocyclyl moiety as defined herein, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to an alkyl moiety at the nitrogen atom. Unless stated otherwise specifically in the specification, heterocyclylalkyl groups are optionally substituted.

"Heterocyclylalkylamino" refers to heterocyclylalkyl-NRa-a moiety wherein RaIs H or alkyl, and wherein the heterocyclylalkyl moiety is attached to the nitrogen via a carbon atom, wherein the nitrogen serves as a linker connecting the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, heterocyclylalkylamino groups are optionally substituted.

"Heterocyclylalkyloxy" means an-O-heterocycloalkyl moiety in which the heterocyclylalkyl moiety is attached to oxygen via a carbon atom, where the oxygen serves as a linker to attach the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, heterocyclylalkyloxy is optionally substituted.

"Heterocyclylamino" means-NRa-a heterocyclyl moiety wherein RaIs H or alkyl, and wherein the heterocyclyl moiety is attached to the nitrogen via a carbon atom, wherein the nitrogen acts as a linker connecting the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, heterocyclylamino is optionally substituted.

"heterocyclyloxy" refers to an-O-heterocyclyl moiety, wherein the heterocyclyl moiety is attached to oxygen via a carbon atom, wherein the oxygen serves as a linker to attach the moiety to the rest of the molecule. Unless stated otherwise specifically in the specification, heterocyclyloxy is optionally substituted.

"hydroxyalkyl" refers to an alkyl group that contains at least one hydroxyl substituent. the-OH substituent may be on a primary, secondary or tertiary carbon. Unless stated otherwise specifically in the specification, hydroxyalkyl is optionally substituted.

"N-heteroaryl" refers to a heteroaryl moiety, as defined herein, which contains at least one nitrogen, and wherein the point of attachment of the heteroaryl moiety to the rest of the molecule is through a nitrogen atom in the heteroaryl ring. Unless stated otherwise specifically in the specification, N-heteroaryl is optionally substituted.

"N-heterocyclyl" refers to a heterocyclyl moiety, as defined herein, that contains at least one nitrogen and wherein the point of attachment of the heterocyclyl moiety to the remainder of the molecule is through a nitrogen atom in the heterocyclyl ring. Unless stated otherwise specifically in the specification, the N-heterocyclyl group is optionally substituted.

"Thioalkyl" means a compound of the formula-SRaWherein R isaIs an alkyl moiety as defined herein containing from one to twelve carbon atoms. Unless stated otherwise specifically in the specification, thioalkyl is optionally substituted.

"alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking two groups in a molecule, which may be saturated or unsaturated (i.e., containing one or more double and/or triple bonds), and has from one to twelve carbon atoms, preferably from one to eight carbon atoms (C)1-C8Alkylene) or one to six carbon atoms (C)1-C6Alkylene) such as methylene, ethylene, propylene, n-butylene, vinylene, propenylene, n-butylene, propynylene, n-butylene, and the like. The alkylene chain is connected to the rest of the molecule by a single or double bond. The point of attachment of the alkylene chain to the remainder of the molecule may be through one carbon in the chain, for example methylene, or any two carbons in the chain, for example-CH2CH(CH3)CH2CH2-. Unless otherwise specifically stated in the specification, the alkylene chain is optionally substituted.

"Alkylenecarbonyl" refers to a compound of the formula-C (═ O) RaA moiety of (a) wherein RaIs an alkylene chain as defined herein. Unless otherwise specifically stated in the specification, an alkylene carbonyl group is optionally substituted.

An "alkenylene group" is an unsaturated alkylene group, as defined herein, which contains one or more carbon-carbon double bonds. Unless stated otherwise specifically in the specification, alkenylene is optionally substituted.

"Alkenylenecarbonyl" refers to an unsaturated alkylenecarbonyl group, as defined herein, that contains one or more carbon-carbon double bonds. Unless otherwise specifically stated in the specification, alkenylenecarbonyl is optionally substituted.

"arylene" refers to a divalent aromatic radical that connects one part of a molecule to another part of the molecule. Unless otherwise specifically stated, arylene is optionally substituted.

"Heteroalkylene" refers to an alkylene group that contains at least one heteroatom (e.g., N, O or S). In some embodiments, the heteroatom is within the alkylene chain (i.e., the heteroalkylene comprises at least one carbon-heteroatom-carbon bond). In other embodiments, the heteroatom is terminal to the alkylene group and connects the alkylene group to the rest of the molecule (e.g., M1-H-a-M2, where M1 and M2 are part of the molecule, H is a heteroatom, and a is an alkylene group). Heteroalkylidene groups may have both internal and terminal heteroatoms, e.g. -OCH2CH2OCH2CH2O-is formed. Unless stated otherwise specifically in the specification, heteroalkylene groups are optionally substituted.

"Heteroalkylcarbonylene" refers to the formula-C (═ O) RaA moiety of (a) wherein RaIs a heteroalkylene chain as defined herein. Unless otherwise specified in the specification, a heteroalkylenecarbonyl group is optionally substituted.

"heteroarylene" refers to a divalent heteroaryl group that connects one part of a molecule to another part of the molecule. Unless otherwise specifically stated, heteroarylene is optionally substituted.

"heteroarylenecarbonyl" refers to the formula-C (═ O) RaA moiety of (a) wherein RaIs a heteroarylene group as defined herein. Heteroarylenecarbonyl is optionally substituted, unless specifically stated otherwise.

"Heterocyclylalkyl" refers to a divalent heterocyclic group that connects one part of a molecule to another part of the molecule. Unless otherwise specifically stated, heterocycloalkylene is optionally substituted.

"Heterocyclylalkylcarbonyl" refers to the formula-C (═ O) RaA moiety of (a) wherein RaIs heterocycloalkylene as defined herein. Unless otherwise specifically stated, heterocycloalkylene carbonyl is optionally substituted.

The term "substituted" as used herein means substituted with any of the above groups (e.g., amino, carboxyl, hydroxyl, imino, acyl, alkyl, alkoxy, alkylamino, alkylaminoalkyl, amide, aminoalkyl, aminocarbonyl, aryl, aralkyl, aralkylamino, aralkyloxy, arylamino, aryloxy, bicycloalkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkyloxy, cycloalkylamino, cycloalkyloxy, halo, haloalkyl, heteroatom, heteroalkyl, heteroaryl, heteroarylalkyl, heteroarylalkylamino, heteroarylalkyloxy, heteroarylamino, heteroaryloxy, heterobicycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclylalkylamino, heterocyclylalkyloxy, heterocyclylamino, heterocyclyloxy, hydroxyalkyl, N-heteroaryl, N-heteroarylalkyl, N-substituted with any of the above groups, N-heterocyclyl, thioalkyl, alkylene, alkylenecarbonyl, alkenylene, alkenylenecarbonyl, arylene, heteroalkylene, heteroalkylenecarbonyl, heteroarylene, heteroarylenecarbonyl, heterocyclylalkyl, and/or heterocyclylalkylcarbonyl) replacing at least one hydrogen atom, wherein the at least one hydrogen atom is replaced with a bond to a non-hydrogen atom such as, but not limited to: halogen atoms such as F, Cl, Br and I; oxygen atoms in groups such as hydroxyl, alkoxy, and ester groups; a group such as a thiol group, a thioalkyl group, a sulfone group such as an alkylsulfone group, a sulfonyl group such as a sulfonamide group, and a sulfonylalkyl group such as a sulfonylmethane, and a sulfoxide group such as a sulfur atom in an alkylsulfoxide group; nitrogen atoms in groups such as amino, amine, amide, alkylamine, dialkylamine, arylamine, alkylarylamine, diarylamine, N-oxide, imide, and enamine; silicon atoms in groups such as trialkylsilyl, dialkylarylsilyl, alkyldiarylsilyl and triarylsilyl; groups such as the phosphorus atom in a dialkylphosphine oxide group; and other heteroatoms in various other groups. "substituted" also means any of the above groups in which one or more hydrogens are presentAtoms are replaced with higher bonds (e.g., double or triple bonds) to carbon atoms or heteroatoms such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced by-NRgRh、-NRgC(=O)Rh、-NRgC(=O)NRgRh、-NRgC(=O)ORh、-NRgSO2Rh、-OC(=O)NRgRh、-ORg、-SRg、-SORg、-SO2Rg、-OSO2Rg、-SO2ORg、=NSO2Rg、-SO2NRgRh、-C(=O)Rg、-C(=O)ORg、-C(=O)NRgRh、-CH2SO2Rgor-CH2SO2NRgRhReplacement of wherein RgAnd RhIndependently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heteroalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl, and/or heteroarylalkyl. "substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to: amino, carbonyl, carboxyl, cyano, hydroxyl, imino, nitro, oxo, thio, acyl, alkyl, alkoxy, alkylamino, alkylaminoalkyl, amide, aminoalkyl, aminocarbonyl, aryl, aralkyl, aralkylamino, aralkyloxy, arylamino, aryloxy, bicycloalkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkyloxy, cycloalkylamino, cycloalkyloxy, halo, haloalkyl, heteroatom, heteroalkyl, heteroaryl, heteroarylalkyl, heteroarylalkylamino, heteroarylalkyloxy, heteroarylamino, heteroarylalkyloxy, heterobicycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclylalkylamino, heterocyclylalkyloxy, heterocyclylamino, heterocyclylalkyloxy, heterocyclylalkyl, and heterocyclylalkyloxyOxy, hydroxyalkyl, N-heteroaryl, N-heterocyclyl, thioalkyl, alkylene, alkylenecarbonyl, alkenylene, alkenylenecarbonyl, arylene, heteroalkylene, heteroalkylenecarbonyl, heteroarylene, heteroarylenecarbonyl, heterocyclylalkyl, heterocyclylalkylene carbonyl, trimethylsilyl, dialkylphosphine oxide, -ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)S(O)tRa(wherein t is1 or 2), -S (O)tORa(wherein t is1 or 2), -S (O)tN(Ra)2(wherein t is1 or 2), -PO (R)a)2OR-PO (OR)a)2Group, wherein each RaIndependently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, or heteroarylalkyl. Further, each of the foregoing substituents is optionally substituted with one or more of the substituents described above.

The term "optionally substituted" as used herein means that the group referred to (e.g., alkyl, cycloalkyl, etc.) may or may not be substituted with one or more additional groups.

As used herein, the term "absent" when used in reference to a functional group or substituent, particularly a chemical structure of a compound, means that the specified functional group or substituent is not present in the compound being described. When used in reference to a substituent (e.g., pendant group, not a linking group), the absence of a substituent generally means that the bond to the substituent is absent and that the absence of the bond is compensated for by an H atom. When used to refer to a position within a chain or ring (e.g., a linking group, not a pendant group), deletion of a position generally means that two positions that would otherwise be connected by the deleted position are either (1) directly connected by a covalent bond, or (2) unconnected, as is apparent or explicitly indicated by the structure.

As used herein, the terms "ring system" and "polycyclic ring system" refer to a chemical structure or moiety comprising two or more rings that share at least one bond (and two or more atomic positions). For example, polycyclic ring systems comprising cyclohexane and cyclopentane are:

if an aryl or heteroaryl ring is included in the polycyclic ring system, the aromaticity of the ring is maintained unless otherwise described, for example, a polycyclic ring system comprising benzene and cyclohexane is:

Detailed Description

Provided herein are small molecules that bind to ASH1L and inhibit the activity of ASH1L, and methods of use thereof for treating diseases including acute leukemia, solid cancers, and other diseases that depend on the activity of ASH 1L.

In some embodiments, provided herein are small molecules that directly target the SET domain of ASH 1L. In some embodiments, the compounds described herein block the catalytic activity of ASH1L once bound to the SET domain. In experiments performed during the development of the embodiments herein, small molecule inhibitors of ASH1L exhibited anti-proliferation and down-regulation of target gene expression.

In some embodiments, the compounds described herein are useful for treating or preventing a disease (e.g., cancer (e.g., leukemia, breast cancer, ovarian cancer, melanoma, prostate cancer, thyroid cancer or metastases thereof, muscular dystrophy, liver fibrosis, etc.) and/or alleviating a symptom associated therewith.

In some embodiments, provided herein are compounds that inhibit ASH1L activity, the compounds comprising a structure of one or both of formulas (IVa) and/or (IVb):

Figure BDA0002573038890000541

[ formula (IVa)](ii) a And

Figure BDA0002573038890000551

[ formula (IVb)]。

In some embodiments, the X group, when present (e.g., in formula (IVa)), is C (e.g., CH) or N.

In some embodiments, R2、R3、R4、R5And R7When present (e.g., when not H) is independently selected from halogen (e.g., Cl, F, Br, I), CH3、OH、SH、NH2、CN、CF3、CCl3、-CH2-CH3、-CH2-OH、-CH2NH2、CH3SH、CH2Cl、CH2Br、CH2F、CHF2、CH2CN、CH2CF3、CH2Cl3Alkyl, haloalkyl and alcohol.

In some embodiments, R1When present (e.g., in formulas (IVa) and (IVb)) are selected from H, alkyl, substituted alkyl (e.g., halo-substituted alkyl), branched alkyl, substituted branched alkyl (e.g., halo-substituted branched alkyl), alkoxy, amine, substituted amine, thioalkyl, ketone, amide, substituted amide, cyano, sulfonyl, carboxyl, dialkylphosphine oxide, carbocyclic ring, substituted carbocyclic ring, aromatic ring, substituted aromatic ring, heterocyclic aromatic ring, substituted or unsubstituted heterocyclic non-aromatic ring (e.g., piperidine, methylpiperidine, bridged piperidine, tetrahydropyran, alkylsulfonyl-substituted piperidine, sulfonamide-substituted piperidine, 1- ((trifluoromethyl) sulfonyl) piperidine, difluorocyclohexane, monofluorocyclohexane, cyclohexane, substituted difluorocyclohexane, bicyclooctane, cycloheptane),A carbocyclic or heterocyclic aromatic ring fused to another aromatic ring, a hydrogen bond donor, a hydrogen bond acceptor, and combinations thereof. In particular embodiments, R1Selected from the substituents shown in table 10 or 13:

TABLE 10 exemplary R1And (4) a substituent.

Figure BDA0002573038890000561

Wherein R is8When present in R of Table 101When in the substituents, are selected from the groups shown in table 11 or table 14:

TABLE 11 exemplary R8And (4) a substituent.

Wherein R is11Selected from the groups shown in table 12:

TABLE 12R of TABLE 118Exemplary R of substituents11And (4) a substituent.

TABLE 13 exemplary R1And (4) a substituent.

Wherein R is8When present in R of tables 10 or 131When in the substituents, are selected from the groups shown in table 14:

TABLE 14 exemplary R8And (4) a substituent.

Figure BDA0002573038890000574

In some embodiments, any R is1The substituent is one of the formulae (a-q); j, Q therein1Or J1One of which, when present, is connected to the primary support; each of which J, J1、J2、J3And J4When present, is independently selected from the group consisting of: covalent bond, H, alkyl1-15Alkenyl radical1-6Alkynyl group1-6、(CH2)0-6C(S)NH2、(CH2)0-6C(O)NH2、O、S、NH、(CH2)0-6C(O)NH(CH2)1-6、(CH2)0-6NHC(O)(CH2)1-6Alkylsulfonyl, sulfonamide, alkylsulfonamide, (CH)2)0-6C(S)NH(CH2)1-6、(CH2)0-6O(CH2)1-6、(CH2)0-6OH、(CH2)0-6S(CH2)1-6、(CH2)0-6SH、(CH2)0-6NH(CH2)1-6、(CH2)0-6N(CH2)1-6(CH2)1-6、(CH2)0-6NH2、(CH2)0-6SO2(CH2)1-6、(CH2)0-6NHSO2(CH2)1-6、(CH2)0-6SO2NH2Halogen (e.g., F, Cl, Br or I), haloalkyl (e.g., (CH)2)0-6CH2F、(CH2)0-3CHF(CH2)0-2CH3Or analogous groups to Br, Cl or I), dihaloalkyl (e.g., (CH)2)0-6CF2H、(CH2)0-3CF2(CH2)0-2CH3Or analogous groups to Br, Cl or I), trihaloalkyl (e.g., (CH)2)0-6CF3Or like groups with Br, Cl or I), at two or more along its lengthAlkyl having 1-3 halogens at one position, (CH)2)1-4SP(Ph)-2=S、(CH2)0-6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1- 5NH2、(CH2)0-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-5SH、(CH2)0-6O(CH2)1-6NH(CH2)1- 5OH、(CH2)0-6O(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6O(CH2)1-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-6O(CH2)1-5SH、(CH2)0-6O(CH2)1-6S(CH2)1-5OH、(CH2)0-6O(CH2)1-6S(CH2)1-5NH2、(CH2)0-6O(CH2)1-6S(CH2)1-5SH、(CH2)0-6S(CH2)1-6NH(CH2)1-5OH、(CH2)0-6S(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6S(CH2)1-6NH(CH2)1-5SH、(CH2)0-6S(CH2)1- 6O(CH2)1-5OH、(CH2)0-6S(CH2)1-6O(CH2)1-5NH2、(CH2)0-6S(CH2)1-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-6S(CH2)1-5SH、(CH2)0-6NH(CH2)1- 6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6NH(CH2)1-5SH、(CH2)0-6NH(CH2)1-6O(CH2)1-5OH、(CH2)0-6NH(CH2)1-6O(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6O(CH2)1-5SH、(CH2)0- 6NH(CH2)1-6S(CH2)1-5OH、(CH2)0-6NH(CH2)1-6S(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6S(CH2)1-5SH、(CH2)0-3C(O)O(CH2)0-3、(CH2)0-3C(S)O(CH2)0-3、(CH2)0-3C(O)S(CH2)0-3、(CH2)0-3C(S)S(CH2)0-3、(CH2)0-3C(O)NH(CH2)0-3、(CH2)0-3C(S)NH(CH2)0-3、(CH2)0-3NHC(O)(CH2)0-3、(CH2)0-3NHC(S)(CH2)0-3、(CH2)0-3OC(O)(CH2)0-3、(CH2)0-3OC(S)(CH2)0-3、(CH2)0-3SC(O)(CH2)0-3、(CH2)0-3SC(S)(CH2)0-3、(CH2)0-3NHC(O)NH(CH2)0-3、(CH2)0-3NHC(S)NH(CH2)0-3、(CH2)0-3OC(O)NH(CH2)0-3、(CH2)0-3OC(S)NH(CH2)0-3、(CH2)0-3SC(O)NH(CH2)0-3、(CH2)0-3SC(S)NH(CH2)0-3、(CH2)0-3NHC(O)O(CH2)0-3、(CH2)0-3NHC(S)O(CH2)0-3、(CH2)0-3OC(O)O(CH2)0-3、(CH2)0-3OC(S)O(CH2)0-3、(CH2)0- 3SC(O)O(CH2)0-3、(CH2)0-3SC(S)O(CH2)0-3、(CH2)0-3NHC(O)S(CH2)0-3、(CH2)0-3NHC(S)S(CH2)0-3、(CH2)0-3OC(O)S(CH2)0-3、(CH2)0-3OC(S)S(CH2)0-3、(CH2)0-3SC(O)S(CH2)0-3、(CH2)0-3SC(S)S(CH2)0-3、(CH2O)1-6And trimethyl methane; each of which Q, Q1And Q2When present, is independently selected from the group consisting of: furan, benzofuran, isobenzofuran, pyrrole, indole, isoindole, thiophene, benzothiophene, benzo [ c]Thiophene, imidazole, benzimidazole, purine, pyrazole, indazole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, benzene, naphthalene, pyridine, quinolone, isoquinoline, pyrazine, quinoxaline, pyrimidine, quinazoline, pyridazine, cinnoline, phthalazine, thalidomide, triazine (examples areSuch as, 1,2, 3-triazine; 1,2, 4-triazine; 1,3,5 triazine), thiadiazole, aziridine, epithiirane (episulfide), oxirane (ethylene oxide, epoxide), oxaaziridine, diepoxyethane, azetidine, oxetane, thietane, diazetidine, dioxetane, dithiolane, pyrrolidine, tetrahydrofuran, thiacyclopentane, imidazolidine, pyrazolidine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, piperidine, dioxane, thiane, piperazine, azetidine morpholine, thiomorpholine, dioxane, dithiane, trioxane, trithiane, azepane, oxepane, thiezepane, homopiperazine, azocane, tetrahydropyran, cyclopentene, cyclohexene, cycloheptene, 1, 3-cyclohexadiene, 1, 4-cyclohexadiene, thiane, cycloheptane, thietane, homopiperazine, azocane, tetrahydropyran, cyclobutene, cyclopentene, cyclohexene, cycloheptene, 1, 3-cyclohexadiene, cyclohexane, and mixtures, 1, 5-cyclooctadiene, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, any suitable C3-C7Cycloalkyl groups and any ring structures shown in table 4; each of which Q, Q1And Q2When present, may exhibit one or more additional J groups at any position on the Q ring; wherein any of the above alkyl groups or (CH)2)x-yThe groups may be linear or branched; wherein any of the above alkyl groups or (CH)2)x-yThe radicals may additionally contain OH on one or more carbons, ═ O, NH2CN, dihaloalkyl (e.g. CF)2H) Trihaloalkyl (e.g., CF)3) Or a halogen (e.g., F) substituent; and wherein the number of hydrogens in the terminal position of the above group can be adjusted in the case where the group is linked to another group (e.g., CH)3Is adjusted to CH2OH to O, etc.) or the group is terminally adjusted (e.g., CH)2Is adjusted to CH3O is adjusted to OH, etc.).

In some embodiments, R6When present (e.g. in formulae (IVa) and (IVb)) is selected from H, alkyl, substituted alkyl (e.g. halogen substituted alkyl), branched alkyl, substituted branched alkyl (e.g. halogen substituted branched alkyl) hydroxy, alkoxy, amine, substituted amine, thioalkyl, halogen, ketoneAn amide, a substituted amide, a cyano, a sulfonyl, a carboxyl, a dialkylphosphine oxide, a carbocycle, a substituted carbocycle, an aromatic ring, a substituted aromatic ring, a heterocyclic aromatic ring, a substituted or unsubstituted heterocyclic non-aromatic ring (e.g., piperidine, methylpiperidine, bridged piperidine, tetrahydropyran, alkylsulfonyl substituted piperidine, sulfonamide substituted piperidine), a carbocyclic or heterocyclic aromatic ring fused to another aromatic ring, a hydrogen bond donor, a hydrogen bond acceptor, and combinations thereof. In particular embodiments, R6Selected from the substituents shown in table 15:

TABLE 15 exemplary R6Substituent group

Figure BDA0002573038890000601

In some embodiments, R6The substituent is one of the formulae (a-q); j, Q therein1Or J1One of which, when present, is connected to the primary support; each of which J, J1、J2、J3And J4When present, is independently selected from the group consisting of: covalent bond, H, alkyl1-15Alkenyl radical1-6Alkynyl group1-6、(CH2)0-6C(S)NH2、(CH2)0-6C(O)NH2、O、S、NH、(CH2)0-6C(O)NH(CH2)1-6、(CH2)0- 6NHC(O)(CH2)1-6Alkylsulfonyl, sulfonamide, alkylsulfonamide, (CH)2)0-6C(S)NH(CH2)1-6、(CH2)0-6O(CH2)1-6、(CH2)0-6OH、(CH2)0-6S(CH2)1-6、(CH2)0-6SH、(CH2)0-6NH(CH2)1-6、(CH2)0-6N(CH2)1-6(CH2)1-6、(CH2)0-6NH2、(CH2)0-6SO2(CH2)1-6、(CH2)0-6NHSO2(CH2)1-6、(CH2)0-6SO2NH2Halogen (e.g., F, Cl, Br or I), haloalkyl (e.g., (CH)2)0-6CH2F、(CH2)0-3CHF(CH2)0-2CH3Or analogous groups to Br, Cl or I), dihaloalkyl (e.g., (CH)2)0-6CF2H、(CH2)0-3CF2(CH2)0-2CH3Or analogous groups to Br, Cl or I), trihaloalkyl (e.g., (CH)2)0-6CF3Or analogous groups to Br, Cl or I), alkyl having 1-3 halogens at two or more positions along its length, (CH)2)1-4SP(Ph)-2=S、(CH2)0-6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1- 5NH2、(CH2)0-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-5SH、(CH2)0-6O(CH2)1-6NH(CH2)1- 5OH、(CH2)0-6O(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6O(CH2)1-6NH(CH2)1-5SH、(CH2)0-6O(CH2)1-6O(CH2)1-5OH、(CH2)0-6O(CH2)1-6O(CH2)1-5NH2、(CH2)0-6O(CH2)1-6O(CH2)1-5SH、(CH2)0-6O(CH2)1-6S(CH2)1-5OH、(CH2)0-6O(CH2)1-6S(CH2)1-5NH2、(CH2)0-6O(CH2)1-6S(CH2)1-5SH、(CH2)0-6S(CH2)1-6NH(CH2)1-5OH、(CH2)0-6S(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6S(CH2)1-6NH(CH2)1-5SH、(CH2)0-6S(CH2)1- 6O(CH2)1-5OH、(CH2)0-6S(CH2)1-6O(CH2)1-5NH2、(CH2)0-6S(CH2)1-6O(CH2)1-5SH、(CH2)0-6S(CH2)1-6S(CH2)1-5OH、(CH2)0-6S(CH2)1-6S(CH2)1-5NH2、(CH2)0-6S(CH2)1-6S(CH2)1-5SH、(CH2)0-6NH(CH2)1- 6NH(CH2)1-5OH、(CH2)0-6NH(CH2)1-6NH(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6NH(CH2)1-5SH、(CH2)0-6NH(CH2)1-6O(CH2)1-5OH、(CH2)0-6NH(CH2)1-6O(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6O(CH2)1-5SH、(CH2)0- 6NH(CH2)1-6S(CH2)1-5OH、(CH2)0-6NH(CH2)1-6S(CH2)1-5NH2、(CH2)0-6NH(CH2)1-6S(CH2)1-5SH、(CH2)0-3C(O)O(CH2)0-3、(CH2)0-3C(S)O(CH2)0-3、(CH2)0-3C(O)S(CH2)0-3、(CH2)0-3C(S)S(CH2)0-3、(CH2)0-3C(O)NH(CH2)0-3、(CH2)0-3C(S)NH(CH2)0-3、(CH2)0-3NHC(O)(CH2)0-3、(CH2)0-3NHC(S)(CH2)0-3、(CH2)0-3OC(O)(CH2)0-3、(CH2)0-3OC(S)(CH2)0-3、(CH2)0-3SC(O)(CH2)0-3、(CH2)0-3SC(S)(CH2)0-3、(CH2)0-3NHC(O)NH(CH2)0-3、(CH2)0-3NHC(S)NH(CH2)0-3、(CH2)0-3OC(O)NH(CH2)0-3、(CH2)0-3OC(S)NH(CH2)0-3、(CH2)0-3SC(O)NH(CH2)0-3、(CH2)0-3SC(S)NH(CH2)0-3、(CH2)0-3NHC(O)O(CH2)0-3、(CH2)0-3NHC(S)O(CH2)0-3、(CH2)0-3OC(O)O(CH2)0-3、(CH2)0-3OC(S)O(CH2)0-3、(CH2)0- 3SC(O)O(CH2)0-3、(CH2)0-3SC(S)O(CH2)0-3、(CH2)0-3NHC(O)S(CH2)0-3、(CH2)0-3NHC(S)S(CH2)0-3、(CH2)0-3OC(O)S(CH2)0-3、(CH2)0-3OC(S)S(CH2)0-3、(CH2)0-3SC(O)S(CH2)0-3、(CH2)0-3SC(S)S(CH2)0-3、(CH2O)1-6And trimethyl methane; each of which Q, Q1And Q2When present, is independently selected from the group consisting of: furan, benzofuran, isobenzofuran, pyrrole, indole, isoindole, thiophene, benzothiophene, benzo [ c]Thiophene, imidazole, benzimidazole, purine, pyrazole, indazole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, benzene, naphthalene, pyridine, quinolone, isoquinoline, pyrazine, quinoxaline, pyrimidine, quinazoline, pyridazine, cinnoline, phthalazine, thalidomide, triazine (e.g., 1,2, 3-triazine; 1,2, 4-triazine; 1,3, 5-triazine), thiadiazole, aziridine, thiirane (episulfide), oxirane (ethylene oxide, epoxide), oxaaziridine, dioxirane, azetidine, oxetane, thietane, diazetidine, dioxetane, dithiocyclobutane, pyrrolidine, tetrahydrofuran, thiacyclopentane, imidazolidine, pyrazolidine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, Dithiolane, piperidine, dioxane, thiane, piperazine, azetidine, morpholine, thiomorpholine, dioxane, dithiane, trioxane, trithiane, azepane, oxepane, thiepane, homopiperazine, azocane, tetrahydropyran, cyclobutene, cyclopentene, cyclohexene, cycloheptene, 1, 3-cyclohexadiene, 1, 4-cyclohexadiene, 1, 5-cyclooctadiene, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, any suitable C3-C7Cycloalkyl groups and any ring structures shown in table 4; each of which Q, Q1And Q2When present, may show one at any position on the Q-ringOr a plurality of additional J groups; wherein any of the above alkyl groups or (CH)2)x-yThe groups may be linear or branched; wherein any of the above alkyl groups or (CH)2)x-yThe radicals may additionally contain OH on one or more carbons, ═ O, NH2CN, dihaloalkyl (e.g. CF)2H) Trihaloalkyl (e.g., CF)3) Or a halogen (e.g., F) substituent; and wherein the number of hydrogens in the terminal position of the above group can be adjusted in the case where the group is linked to another group (e.g., CH)3Is adjusted to CH2OH to O, etc.) or the group is terminally adjusted (e.g., CH)2Is adjusted to CH3O is adjusted to OH, etc.).

In some embodiments, the compounds comprise any suitable group or substituent in any combination present in the compounds listed in table 9.

In some embodiments, the compound is selected from the compounds shown in table 9 (e.g., compounds 21-85).

In some embodiments, provided herein are compounds defined by one of formulas (IVa) and (IVb), but wherein the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety (as shown in formulas (IVa) and (IVb)). For example, for any formula (e.g., formulae (IVa) and (IVb)) or compound (e.g., compounds 21-85) described herein having benzothioamide-pyrrole or benzamido-pyrrole ring connectivity, such as:

provided herein are corresponding formulae (e.g., formulae (IVc) and (IVd)) and compounds (e.g., compounds 86-150) having benzothioamide-benzene or benzamido-benzene ring connectivity, such as:

Figure BDA0002573038890000661

and are within the scope of the embodiments herein. For example, any of the embodiments, substituents, compounds, etc., described herein in connection with formula (IVa) may also be provided in embodiments in connection with formula (IVc):

and is

Any of the embodiments, substituents, compounds, etc., described herein in connection with formula (IVb) may also be provided in embodiments herein in connection with formula (IVd):

Figure BDA0002573038890000663

compounds 86-150 correspond to compounds 21-85, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety. For example, compound 86 corresponds to compound 21, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; compound 87 corresponds to compound 22, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; compound 88 corresponds to compound 23, except that the benzothioamide (or benzamide) is attached to the benzene moiety of the indole bicyclic structure instead of the pyrrole moiety; and so on.

The compounds described herein may exist in some cases as diastereomers, enantiomers, or other stereoisomeric forms. The compounds given herein include all diastereomeric, enantiomeric and epimeric forms and appropriate mixtures thereof. The separation of stereoisomers may be carried out by chromatography, or by formation of diastereomers and separation by recrystallization or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H.Wilen, "Enantiomers, racemes And solutions," John Wiley And Sons, Inc., 1981, the disclosure of which is incorporated herein by reference). Stereoisomers may also be obtained by stereoselective synthesis.

In some embodiments, the compounds may exist in tautomeric forms. All tautomers are encompassed within the chemical formulas described herein.

Unless otherwise specified, divalent variables or groups described herein may be attached in the orientation they are shown, or they may be attached in the opposite orientation.

The methods and compositions described herein include the use of amorphous as well as crystalline forms (also referred to as polymorphs). The compounds described herein may be in the form of pharmaceutically acceptable salts. Likewise, active metabolites of these compounds having the same type of activity are also included within the scope of the present disclosure. In addition, the compounds described herein may exist in unsolvated forms as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds set forth herein are also considered disclosed herein.

In some embodiments, a compound or salt described herein can be a prodrug. "prodrug" refers to an agent that is converted in vivo to the parent drug. Prodrugs are often useful because, in some cases, they may be easier to administer than the parent drug. For example, they may be bioavailable by oral administration, but not the parent. The prodrug may also have improved solubility in pharmaceutical compositions compared to the parent drug. One example, but not limited to, of a prodrug is a compound described herein that is administered in the form of an ester ("prodrug") to facilitate delivery across the cell membrane where water solubility is detrimental to mobility, but which is metabolically hydrolyzed to the active entity carboxylic acid once inside the cell where water solubility is beneficial. A further example of a prodrug may be a short peptide (polyamino acid) bonded to an acid group, where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, the prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In certain embodiments, the prodrug is enzymatically metabolized to the biologically, pharmaceutically, or therapeutically active form of the compound by one or more steps or processes.

To produce a prodrug, the pharmaceutically active compound is modified so that the active compound will regenerate after in vivo administration. Prodrugs can be designed to alter the metabolic stability or transport properties of the drug to mask side effects or toxicity, to improve the taste of the drug, or to alter other properties or characteristics of the drug. In some embodiments, prodrugs of a pharmaceutically active compound are designed once the compound is determined, with the aid of knowledge of the pharmacodynamic processes and drug metabolism in vivo. (see, for example, Nogrady (1985) Medicinal Chemistry A Biochemical apparatus, Oxford University Press, New York, pp. 388-.

The compounds described herein may be isotopically (e.g., with a radioisotope) or otherwise labeled, including but not limited to the use of chromophores or fluorescent moieties, bioluminescent labels, light activated or chemiluminescent labels, affinity labels (e.g., biotin), and the like.

The compounds and salts described herein include isotopically labeled compounds. Isotopically-labeled compounds are generally identical to those recited in the various formulae and structures presented herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example each2H、3H、13C、14C、15N、18O、17O、35S、18F、36And (4) Cl. Certain isotopically-labeled compounds described herein, for example, those into which a radioactive isotope such as3H and14c, useful in drug and/or substrate tissue distribution assays. Further, with isotopes such as deuterium (i.e., deuterium)2H) The substitution may be due to metabolic stabilityHigher potency may result in certain therapeutic advantages, such as increased in vivo half-life or reduced dosage requirements, for example.

In additional or further embodiments, the compounds described herein are metabolized upon administration to an organism in need thereof to produce a metabolite, which is then used to produce a desired effect, including a desired therapeutic effect.

The compounds described herein may be formed into and/or used as pharmaceutically acceptable salts. Types of pharmaceutically acceptable salts include, but are not limited to: (1) an acid addition salt formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid and the like; or an organic acid such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo- [2.2.2] oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4' -methylenebis- (3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, laurylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid, valproic acid, and the like; (2) salts are formed when an acidic proton present in the parent compound is replaced by a metal ion, such as an alkali metal ion (e.g., lithium, sodium, potassium), an alkaline earth ion (e.g., magnesium or calcium), or an aluminum ion. In some cases, the compounds described herein can be complexed with an organic base such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris (hydroxymethyl) methylamine. In other instances, the compounds described herein may form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases for forming salts with compounds that include acidic protons include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.

It will be understood that reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs. Solvates contain either stoichiometric or non-stoichiometric amounts of solvent and may be formed during the crystallization process with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of the compounds described herein may be conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to unsolvated forms for the purposes of the compounds and methods provided herein.

In some embodiments, the compounds described herein having any suitable substituents and functional groups disclosed herein are in various forms, including but not limited to amorphous, milled, and nanoparticle forms. In addition, the compounds described herein include crystalline forms, also referred to as polymorphs. Polymorphs include different crystal packing arrangements of the same elemental composition of a compound. Polymorphs typically have different X-ray diffraction patterns, melting points, densities, hardness, crystal shape, optical properties, stability and solubility. Various factors such as recrystallization solvent, crystallization rate, and storage temperature can cause the single crystal form to dominate.

Screening and characterization of pharmaceutically acceptable salts, polymorphs, and/or solvates can be accomplished using a variety of techniques, including but not limited to thermal analysis, x-ray diffraction, spectroscopy, vapor adsorption, and microscopy. Thermal analysis methods address thermochemical degradation or thermophysical processes, including but not limited to polymorphic transformations, and such methods are used to analyze relationships between polymorphic forms, determine weight loss, determine glass transition temperature, or for excipient compatibility studies. Such methods include, but are not limited to, Differential Scanning Calorimetry (DSC), modulated differential scanning calorimetry (MDCS), thermogravimetric analysis (TGA), and thermogravimetric and infrared analysis (TG/IR). X-ray diffraction methods include, but are not limited to, single crystal and powder diffractometers and synchrotron radiation sources. Various spectroscopic techniques employed include, but are not limited to, Raman, FTIR, UV-VIS, and NMR (liquid and solid). Various microscopy techniques include, but are not limited to, polarized light microscopy, Scanning Electron Microscopy (SEM) with energy dispersive X-ray analysis (EDX), ambient scanning electron microscopy (in a gas or water vapor atmosphere) with EDX, IR microscopy, and raman microscopy.

Throughout the specification, groups and substituents thereof may be selected to give stable moieties and compounds.

Pharmaceutical composition

In certain embodiments, a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituent and functional group disclosed herein is combined with one or more additional agents to form a pharmaceutical composition. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The appropriate formulation will depend on the route of administration chosen. Additional details regarding suitable excipients for the pharmaceutical compositions described herein can be found, for example, in Remington: the science and Practice of Pharmacy, nineteenth edition (Easton, Pa.: Mack Publishing Company, 1995); hoover, John e., Remington's Pharmaceutical Sciences, Mack Publishing co, Easton, Pennsylvania 1975; liberman, h.a. and Lachman, l. eds, Pharmaceutical DosageForms, Marcel Decker, New York, n.y., 1980; and Pharmaceutical document Forms and drug delivery Systems, seventh edition (Lippincott Williams & Wilkins1999), the disclosures of which are incorporated herein by reference.

Pharmaceutical composition as used herein refers to a mixture of a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein, and other chemical components such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents and/or excipients. The pharmaceutical composition facilitates administration of the compound to an organism. In practicing the treatment or methods of use provided herein, a therapeutically effective amount of a compound described herein is administered in the form of a pharmaceutical composition to a mammal suffering from the disease, disorder, or condition to be treated. In some embodiments, the mammal is a human. The therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used, and other factors. The compounds or salts of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein may be used alone or in combination with one or more therapeutic agents as a component of a mixture (e.g. in a combination therapy).

The pharmaceutical formulations described herein can be administered to a subject by a variety of routes of administration including, but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal routes of administration. In addition, the pharmaceutical compositions described herein comprising a compound of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein can be formulated into any suitable dosage form, including, but not limited to, aqueous oral dispersions, liquids, gels, syrups, elixirs, slurries, suspensions, aerosols, fast-melt formulations, effervescent formulations, lyophilized formulations, tablets, powders, pills, dragees, and capsules.

The compounds and/or compositions may be administered locally rather than systemically, for example via direct injection of the compound into an organ or tissue, typically in the form of a depot or sustained release formulation. Such long acting formulations may be administered by implantation (e.g. subcutaneously or intramuscularly) or by intramuscular injection. Furthermore, the drug may be administered in a targeted drug delivery system, for example in a liposome coated with an organ-specific antibody. Liposomes will be selectively targeted and absorbed by the organ. In addition, the drug may be provided in a fast release formulation, an extended release formulation, or an intermediate release formulation.

Pharmaceutical compositions comprising the compounds described herein may be manufactured in a conventional manner, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compressing processes, as merely examples.

The pharmaceutical composition will comprise at least one compound of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein as the active ingredient in free acid or free base form or in pharmaceutically acceptable salt form.

In certain embodiments, the compositions provided herein can further include one or more preservatives to inhibit microbial activity. Suitable preservatives include quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.

Pharmaceutical formulations for oral use may be obtained by mixing one or more solid excipients with one or more compounds or salts of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein, optionally grinding the resulting mixture, and, if desired, treating the mixture of granules, after addition of suitable auxiliaries, to obtain tablets, pills or capsules. Suitable excipients include, for example, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, such as, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, microcrystalline cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose; or other substances, such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents can be added, such as cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate.

Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbomer gel, polyethylene glycol and/or titanium dioxide, varnish solutions and suitable organic solvents or solvent mixtures. Dyes or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.

Pharmaceutical preparations for oral use include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Push-fit capsules can contain the active ingredient in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added.

In some embodiments, the solid dosage forms disclosed herein can be in the form of a tablet (including a suspension tablet, an immediate-melt tablet, a bite-block tablet, an rapidly disintegrating tablet, an effervescent tablet or caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder or an effervescent powder), a capsule (including a soft or hard capsule, such as a capsule made from gelatin of animal origin or HPMC of vegetable origin, or a "sprinkle capsule"), a solid dispersion, a solid solution, a bioerodible dosage form, a multiparticulate dosage form, a pellet, a granule, or an aerosol. In other embodiments, the pharmaceutical formulation is in powder form. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including but not limited to an immediate-melt tablet. In addition, pharmaceutical formulations of the compounds described herein can be administered as a single capsule or in a multi-capsule dosage form. In some embodiments, the pharmaceutical formulation is administered in two, three, or four capsules or tablets.

In some embodiments, solid dosage forms, such as tablets, effervescent tablets, and capsules, are prepared by mixing particles of a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein with one or more pharmaceutical excipients to form a bulk blend composition. When referring to these bulk blend compositions as homogeneous, it is meant that the particles of the compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein are uniformly dispersed throughout the composition such that the composition can be subdivided into equally effective unit dosage forms, such as tablets, pills and capsules. Individual unit doses may also include film coatings that disintegrate upon oral ingestion or contact with diluents. These formulations may be prepared by conventional pharmacological techniques.

The pharmaceutical solid dosage forms described herein can include a compound of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein and one or more pharmaceutically acceptable additives, such as compatible carriers, binders, fillers, suspending agents, flavoring agents, sweeteners, disintegrants, dispersants, surfactants, lubricants, colorants, diluents, solubilizers, wetting agents, plasticizers, stabilizers, permeation enhancers, wetting agents, antifoaming agents, antioxidants, preservatives, or one or more combinations thereof. In still other aspects, a film coating is provided around the formulation of the compounds described herein using standard coating procedures, such as those described in Remington's pharmaceutical sciences, 20 th edition (2000). In one embodiment, some or all of the particles of the compounds described herein are coated. In another embodiment, some or all of the particles of the compounds described herein are microencapsulated. In yet another embodiment, the particles of the compounds described herein are not microencapsulated and uncoated.

Suitable carriers for use in the solid dosage forms described herein include, but are not limited to, gum arabic, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerol, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglycerides, diglycerides, pregelatinized starch, hydroxypropyl methylcellulose acetate stearate, sucrose, microcrystalline cellulose, lactose, mannitol, and the like.

Suitable fillers for use in the solid dosage forms described herein include, but are not limited to, lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates (dextrates), dextran, starch, pregelatinized starch, hydroxypropyl methylcellulose (HPMC), hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate stearate (HPMCAS), sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.

In order to release the compounds of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein from the solid dosage form matrix as efficiently as possible(IVb) a compound or salt of any one of (IVb), often a disintegrant is used in the formulation, especially when the dosage form is compressed with a binder. Disintegrants help to break the matrix of the dosage form by swelling or capillary action as water is absorbed into the dosage form. Suitable disintegrants for use in the solid dosage forms described herein include, but are not limited to, native starches (e.g., corn or potato starch), pregelatinized starches (e.g., National 1551 or) Or sodium starch glycolate (e.g. sodium starch glycolate)Or) Cellulose (e.g. wood products), methyl crystalline cellulose (e.g. wood products)

Figure BDA0002573038890000754

PH101、

Figure BDA0002573038890000755

PH102、

Figure BDA0002573038890000756

PH105、P100、Ming

Figure BDA0002573038890000759

And

Figure BDA00025730388900007510

) Methylcellulose, croscarmellose or cross-linked cellulose such as croscarmellose sodium

Figure BDA00025730388900007511

Croscarmellose or crosslinked croscarmellose, crosslinked starch (such as sodium starch glycolate), crosslinked polymer (such as crospovidone), crosslinked polyvinylpyrrolidone, alginate such as alginic acid or a salt of alginic acid (such as sodium alginate), clay such as sodium alginateHV (magnesium aluminum silicate), gums (such as agar, guar gum, locust bean gum, carrageenan, pectin, or tragacanth gum), sodium starch glycolate, bentonite, natural sponge, surfactants, resins (such as cation exchange resins), citrus pulp, sodium lauryl sulfate, combinations of sodium lauryl sulfate and starch, and the like.

Binders impart cohesion to the solid oral dosage formulation: for powder filled capsule formulations they help to form a plug that can be filled into soft or hard shell capsules, and for tablet formulations they ensure that the tablet remains intact after compression and help to ensure blending homogeneity prior to the compression or filling step. Materials suitable for use as binders in the solid dosage forms described herein include, but are not limited to, carboxymethylcellulose, methylcellulose (e.g.,) Hydroxypropyl methylcellulose (e.g., hypromellose USP Pharmacoat-603, hydroxypropyl methylcellulose acetate stearate (aqoate HS-LF and HS), hydroxyethyl cellulose, hydroxypropyl cellulose (e.g.,) Ethyl cellulose (e.g.,) And microcrystalline cellulose (e.g.,) Microcrystalline dextrose, amylose, magnesium aluminum silicate, gluconic acid, bentonite, gelatin, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, starchFlour, pregelatinized starch, gum tragacanth, dextrin, sugars such as sucrose (e.g.,) Glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g.,) Lactose, natural or synthetic gums such as acacia, tragacanth, ghatti, mucilage of isapol husks, starch, polyvinylpyrrolidone (e.g.,

Figure BDA0002573038890000767

CL、

Figure BDA0002573038890000768

CL、

Figure BDA0002573038890000769

XL-10 and

Figure BDA00025730388900007610

k-12), larch arabinogalactan,Polyethylene glycol, wax, sodium alginate, etc.

Generally, binder levels of 20-70% are used in powder filled gelatin capsule formulations. The level of binder used in tablet formulations varies depending on whether direct compression, wet granulation, roller compaction or the use of other excipients such as fillers which themselves act as moderate binders. In some embodiments, the formulator determines the binder level of the formulation, but binder use levels as high as 70% are common in tablet formulations.

Suitable lubricants or glidants for the solid dosage forms described herein include, but are not limited to, stearic acid, calcium hydroxide, talc, corn starch, sodium stearyl fumarate, alkali and alkaline earth metal salts (e.g., aluminum, calcium, magnesium, zinc, stearic acid, sodium stearate, magnesium stearate, sodium stearyl fumarate, and sodium stearyl fumarate,Zinc stearate), wax,

Figure BDA00025730388900007612

Boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, polyethylene glycol or methoxypolyethylene glycol (e.g. Carbowax)TMPEG 4000, PEG 5000, PEG 6000), propylene glycol, sodium oleate, glyceryl behenate, glyceryl palmitostearate, glyceryl benzoate, magnesium lauryl sulfate, sodium lauryl sulfate, or the like.

Suitable diluents for the solid dosage forms described herein include, but are not limited to, sugars (including lactose, sucrose, and dextrose), polysaccharides (including dextrates and maltodextrins), polyols (including mannitol, xylitol, and sorbitol), cyclodextrins, and the like.

Suitable wetting agents for use in the solid dosage forms described herein include, for example, oleic acid, glycerol monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, quaternary ammonium compounds (e.g., Polyquat)) Sodium oleate, sodium lauryl sulfate, magnesium stearate, docusate sodium, triacetin, vitamin E TPGS, and the like.

Suitable surfactants for use in the solid dosage forms described herein include, for example, sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, poloxamers, bile salts, glycerol monostearate, copolymers of ethylene oxide and propylene oxide such as(BASF) and the like.

Suitable suspending agents for use in the solid dosage forms described herein include, but are not limited to, polyvinylpyrrolidone (e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30), polyethylene glycol (e.g., the polyethylene glycol may have a molecular weight of from about 300 to about 6000 or from about 3350 to about 4000 or from about 5400 to about 7000), vinylpyrrolidone/vinyl acetate copolymer (S630), sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums (such as, for example, tragacanth and acacia), guar gum, xanthans (including xanthan gum), sugars, celluloses (such as, for example, sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose), polysorbate-80, guar gum-1, xanthan gum, sugars, celluloses (such as, sodium carboxymethylcellulose, methylcellulose, carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose), sorbitol-80, and mixtures thereof, Sodium alginate, polyethoxylated sorbitan monolaurate, povidone, and the like.

Suitable antioxidants for use in the solid dosage forms described herein include, for example, Butylated Hydroxytoluene (BHT), sodium ascorbate, and tocopherol.

There is considerable overlap between the additives used in the solid dosage forms described herein. Thus, the additives listed above should be considered as merely illustrative and non-limiting types of additives that may be included in the solid dosage forms of the pharmaceutical compositions described herein.

In other embodiments, one or more layers of the pharmaceutical formulation are plasticized. Illustratively, plasticizers are typically high boiling point solids or liquids. Suitable plasticizers may be added in an amount of about 0.01% to about 50% by weight (w/w) of the coating composition. Plasticizers include, but are not limited to, diethyl phthalate, citric acid esters, polyethylene glycol, glycerol, acetylated glycerides, triacetin, polypropylene glycol, polyethylene glycol, triethyl citrate, dibutyl sebacate, stearic acid, stearyl alcohol, stearates, and castor oil.

Compressed tablets are solid dosage forms prepared by compacting a bulk blend of the above formulations. In various embodiments, a compressed tablet designed to dissolve in the oral cavity will include one or more flavoring agents. In other embodiments, the compressed tablet will comprise a film surrounding the final compressed tablet. In some embodiments, the film coating aids in patient compliance (e.g.,

Figure BDA0002573038890000781

a coating or a sugar coating). Comprises thatThe film coating of (a) typically comprises from about 1% to about 3% by weight of the tablet. In other embodiments, the compressed tablet comprises one or more excipients.

Capsules can be prepared, for example, by placing a bulk blend of a formulation of the above-described compounds within a capsule. In some embodiments, the formulations (non-aqueous suspensions and solutions) are placed in soft gelatin capsules. In other embodiments, the formulation is placed in a standard gelatin or non-gelatin capsule (e.g., a capsule comprising HPMC). In other embodiments, the formulation is placed in a sprinkle capsule, wherein the capsule can be swallowed whole, or the capsule can be opened and the contents sprinkled on food before eating. In some embodiments, the therapeutic dose is divided among multiple (e.g., two, three, or four) capsules. In some embodiments, the entire dose of the formulation is delivered in capsule form.

In various embodiments, the granules of the compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein and one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 30 minutes, less than about 35 minutes, less than about 40 minutes, less than about 45 minutes, less than about 50 minutes, less than about 55 minutes, or less than about 60 minutes after oral administration, thereby releasing the formulation into the gastrointestinal fluid.

In another aspect, the dosage form may comprise a microencapsulated formulation. In some embodiments, one or more other compatible materials are present in the microencapsulated material. Exemplary materials include, but are not limited to, pH adjusters, corrosion promoters, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegrants, fillers, surfactants, solubilizing agents, stabilizers, lubricants, wetting agents, and diluents.

Materials useful for microencapsulation as described herein include materials compatible with the compounds described herein, which substantially isolate the compounds from other incompatible excipients.

In still other embodiments, effervescent powders are also prepared according to the present disclosure. Effervescent salts have been used to disperse drugs in water for oral administration. Effervescent salts are granules or meals containing the medicament in a dry mixture, usually consisting of sodium bicarbonate, citric acid and/or tartaric acid. When such salts are added to water, the acid and base react to release carbon dioxide gas, causing "effervescence". Examples of effervescent salts include, for example, the following: sodium bicarbonate or a mixture of sodium bicarbonate and sodium carbonate, citric acid and/or tartaric acid. Any acid-base combination that results in the release of carbon dioxide may be used in place of the combination of sodium bicarbonate and citric and tartaric acids, so long as the ingredients are suitable for pharmaceutical use and result in a pH of about 6.0 or higher.

In other embodiments, the formulations described herein comprising a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituent and functional group disclosed herein are solid dispersions. Methods of preparing such solid dispersions include, but are not limited to, for example, U.S. patent nos. 4,343,789, 5,340,591, 5,456,923, 5,700,485, 5,723,269, and U.S. patent publication No. 2004/0013734. In still other embodiments, the formulations described herein are solid solutions. Solid solutions combine the substance with the active agent and other excipients such that heating the mixture causes the drug to dissolve, and then cooling the resulting composition to give a solid blend, which can be further formulated, either added directly to capsules or compressed into tablets. Methods of preparing such solid solutions include, but are not limited to, for example, U.S. patent nos. 4,151,273, 5,281,420, and 6,083,518.

In some embodiments, there is provided a pharmaceutical formulation comprising particles of a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein and at least one dispersing or suspending agent for oral administration to a subject. The formulation may be a powder and/or granules for suspension and when mixed with water gives a substantially homogeneous suspension.

The liquid formulation dosage form for oral administration may be an aqueous suspension selected from the group including, but not limited to: pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels and syrups. See, for example, Singh et al, Encyclopedia of Pharmaceutical Technology, 2 nd edition, pages 754-757 (2002).

The aqueous suspensions and dispersions described herein can be maintained in a homogeneous state for at least 4 hours as defined in The USP pharmaceuticals' Pharmacopeia (2005 edition, chapter 905). Homogeneity should be determined by a sampling method consistent with the determination of homogeneity for the bulk composition. In one embodiment, the aqueous suspension may be resuspended to a homogenous suspension by physical agitation for less than 1 minute. In another embodiment, the aqueous suspension may be resuspended to a homogenous suspension by physical agitation for less than 45 seconds. In yet another embodiment, the aqueous suspension may be resuspended to a homogenous suspension by physical agitation for less than 30 seconds. In yet another embodiment, no agitation is required to maintain a homogeneous aqueous dispersion.

The pharmaceutical compositions described herein may include sweeteners such as, but not limited to, gum arabic syrup, acesulfame potassium, alitame, anise, apple, aspartame, banana, bavaria cream, berry, blackcurrant, butterscotch, calcium citrate, camphor, caramel, cherry cream, chocolate, cinnamon, bubble gum, citrus panne (citrus punch), citrus cream, marshmallow, cocoa, cola, cold cherry, cold orange, cyclamate, dextrose, eucalyptus, eugenol, fructose, fruit panne, ginger, glycyrrhetinate, licorice (licorice juice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glycyrrhetinate

Figure BDA0002573038890000801

Maltitol, mannitol, maple leaf, marshmallow, menthol, peppermint cream, mixed berries, neohesperidin DC, neotame, orange, pear, peach, mint, peppermint cream, peppermint, corn, and corn,

Figure BDA0002573038890000802

Raspberry, leja, rum, saccharin, safrole, sorbitol, spearmint cream, strawberry cream, stevia, sucralose, sucrose, saccharin sodium, saccharin, aspartame, acesulfame, mannitol, talin, sucralose, sorbitol, swiss cream, tagatose, orange, thaumatin, lactulose, vanilla, walnut, watermelon, malpighian, wintergreen, xylitol, or any combination of these flavoring ingredients, such as anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof.

In some embodiments, the pharmaceutical formulation described herein may be a self-emulsifying drug delivery system (SEDDS). An emulsion is a dispersion of one immiscible phase in another phase, usually in the form of droplets. Generally, emulsions are produced by vigorous mechanical dispersion. In contrast to emulsions or microemulsions, SEDDS spontaneously forms an emulsion when added to excess water without any external mechanical dispersion or agitation. One advantage of SEDDS is that only gentle mixing is required to distribute the droplets throughout the solution. In addition, water or an aqueous phase may be added immediately before application, which ensures the stability of the unstable or hydrophobic active ingredient. Thus, the SEDDS provides an effective delivery system for oral and parenteral delivery of hydrophobic active ingredients. The SEDDS can improve the bioavailability of the hydrophobic active ingredient. Methods of making self-emulsifying dosage forms include, but are not limited to, for example, U.S. patent nos. 5,858,401, 6,667,048, and 6,960,563.

There is an overlap between the additives listed above for use in the aqueous dispersions or suspensions described herein, as a given additive is often classified in different ways by different practitioners in the art, or is generally used for any of several different functions. Thus, the additives listed above should be considered as merely illustrative and not limiting types of additives that may be included in the formulations described herein.

Potential excipients for intranasal formulation include, for example, U.S. patent nos. 4,476,116, 5,116,817, and 6,391,452. Solutions of the formulation in saline are used with benzyl alcohol or other suitable preservatives, fluorocarbons, and/or other solubilizing or dispersing agents. See, e.g., Ansel, H.C. et al, Pharmaceutical document Forms and Drug delivery systems, sixth edition (1995). These compositions and formulations are preferably prepared using suitable non-toxic pharmaceutically acceptable ingredients. The choice of a suitable carrier is highly dependent on the exact nature of the nasal dosage form desired, e.g., a solution, suspension, ointment, or gel. Nasal formulations typically contain a large amount of water in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusting agents, emulsifying or dispersing agents, preservatives, surfactants, gelling or buffering agents, and other stabilizing and solubilizing agents may also be present. Preferably, the nasal dosage form should be isotonic with nasal secretions.

For administration by inhalation, the compounds described herein may be in the form of an aerosol, mist, or powder. The pharmaceutical compositions described herein are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of a compound as described herein and a suitable powder base such as lactose or starch, as merely examples.

Various formulations can be used to administer the buccal formulations comprising the compounds described herein, including, but not limited to, U.S. Pat. nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136. In addition, the buccal dosage forms described herein may further comprise a bioerodible (hydrolyzable) polymeric carrier, which also serves to adhere the dosage form to the buccal mucosa. Buccal dosage forms are formulated to gradually erode over a predetermined period of time, wherein delivery of the compound is provided substantially throughout. Buccal drug delivery avoids the disadvantages encountered with oral drug administrationSuch as slow absorption, degradation of the active agent by fluids present in the gastrointestinal tract, and/or first-pass inactivation in the liver. With respect to the bioerodible (hydrolyzable) polymeric carrier, virtually any such carrier can be used so long as the desired drug release profile is not adversely affected and the carrier is compatible with the compounds described herein and any other components that may be present in the buccal dosage unit. Typically, the polymeric carrier comprises hydrophilic (water-soluble and water-swellable) polymers that adhere to the wet surface of the buccal mucosa. Examples of polymeric carriers useful herein include acrylic polymers and copolymers, such as those known as "carbomers" (available from b.f. goodrich)

Figure BDA0002573038890000821

Is one such polymer). Other components may also be incorporated into the buccal dosage forms described herein, including, but not limited to, disintegrants, diluents, binders, lubricants, flavoring agents, coloring agents, preservatives, and the like. For buccal or sublingual administration, the compositions may take the form of tablets, lozenges or gels formulated in conventional manner.

A variety of devices can be used to administer the transdermal formulations described herein, including, but not limited to, U.S. patent nos. 3,598,122, 3,598,123, 3,710,795, 3,731,683, 3,742,951, 3,814,097, 3,921,636, 3,972,995, 3,993,072, 3,993,073, 3,996,934, 4,031,894, 4,060,084, 4,069,307, 4,077,407, 4,201,211, 4,230,105, 4,292,299, 4,292,303, 5,336,168, 5,665,378, 5,837,280, 5,869,090, 6,923,983, 6,929,801, and 6,946,144.

The transdermal dosage forms described herein may incorporate certain pharmaceutically acceptable excipients that are conventional in the art. In one embodiment, the transdermal formulations described herein include at least three components: (1) formulations of compounds or salts of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein; (2) a penetration enhancer; and (3) an aqueous adjuvant. In addition, the transdermal formulations may include additional components such as, but not limited to, gelling agents, creams, ointment bases, and the like. In some embodiments, the transdermal formulation may further include a woven or nonwoven backing material to enhance absorption and prevent removal of the transdermal formulation from the skin. In other embodiments, the transdermal formulations described herein may maintain a saturated or supersaturated state to facilitate diffusion into the skin.

Formulations suitable for transdermal administration of the compounds described herein may employ transdermal delivery devices and transdermal delivery patches, and may be lipophilic emulsions or buffered aqueous solutions, dissolved and/or dispersed in polymers or adhesives. Such patches may be configured for continuous, pulsatile, or on-demand delivery of the medicament. Further, transdermal delivery of the compounds described herein may be accomplished with the aid of iontophoresis patches and the like. In addition, transdermal patches can provide controlled delivery of the compounds described herein. The rate of absorption can be slowed by the use of a rate controlling membrane or by entrapping the compound within a polymer matrix or gel. Conversely, absorption enhancers may be used to increase absorption. The absorption enhancer or carrier may include an absorbable pharmaceutically acceptable solvent to aid passage through the skin. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound, optionally together with a carrier, a rate controlling barrier optionally to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and a mechanism to secure the device to the skin.

Formulations suitable for intramuscular, subcutaneous or intravenous injection may include physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, cremophor, etc.), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. Formulations suitable for subcutaneous injection may also contain additives such as preservatives, wetting agents, emulsifying agents and partitioning agents. Prevention of the growth of microorganisms can be ensured by various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.

For intravenous injection, the compounds described herein can be formulated in aqueous solution, preferably in a physiologically compatible buffer, such as hank's solution, ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally recognized in the art. For other parenteral injections, suitable formulations may include aqueous or non-aqueous solutions, preferably with physiologically compatible buffers or excipients. Such excipients are generally recognized in the art.

Parenteral injection may involve bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The pharmaceutical compositions described herein may be in a form suitable for parenteral injection as a sterile suspension, solution or emulsion in an oily or aqueous vehicle, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compound to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

In certain embodiments, delivery systems for pharmaceutical compounds may be employed, such as, for example, liposomes and emulsions. In certain embodiments, the compositions provided herein further comprise a mucoadhesive polymer selected from, for example, carboxymethylcellulose, carbomer (acrylic acid polymer), poly (methyl methacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate, and dextran.

In some embodiments, the compounds described herein can be administered topically and are formulated into various compositions that can be administered topically, such as solutions, suspensions, lotions, gels, pastes, sticks, balms, creams, or ointments. Such pharmaceutical compounds may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.

The compounds described herein may also be formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, gel suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, and synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In suppository forms of the composition, the first to melt is a low melting point wax, such as but not limited to a mixture of fatty acid glycerides, optionally in combination with cocoa butter.

Generally, an agent, such as a compound of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein, is administered in an amount effective to ameliorate the symptoms of a disease or disorder or to prevent the development of the symptoms of a disease or disorder (i.e., a therapeutically effective amount). Thus, a therapeutically effective amount may be an amount capable of at least partially preventing or reversing a disease or condition. The dosage required to obtain an effective amount may vary depending on the agent, formulation, disease or condition, and the condition of the individual to whom the agent is administered.

Determination of an effective amount may also involve an in vitro assay in which different doses of an agent are administered to cells in culture and the concentration of the agent effective to ameliorate some or all of the symptoms is determined in order to calculate the desired concentration in vivo. An effective amount may also be based on in vivo animal studies.

The agent may be administered before, concurrently with, and after the onset of symptoms of the disease or disorder. In some embodiments, the agent is administered to a subject with a family history of a disease or disorder or with a phenotype that may indicate a predisposition to a disease or disorder or with a genotype that predisposes the subject to a disease or disorder.

In some embodiments, the compositions described herein are provided in the form of pharmaceutical and/or therapeutic compositions. The pharmaceutical and/or therapeutic compositions of the present invention may be administered in a variety of ways depending on whether local or systemic treatment is desired and the area to be treated. Administration can be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, transdermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intracerebroventricular, administration. Compositions and formulations for topical administration include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. A conventional carrier; an aqueous powder or an oily base; a thickener; etc. may be necessary or desirable. Compositions and formulations for oral administration include powders or granules, suspensions or solutions in aqueous or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders are desirable. Compositions and formulations for parenteral, intrathecal or intracerebroventricular administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients. The pharmaceutical and/or therapeutic compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions can be produced from a variety of components, including but not limited to, preformed liquids, self-emulsifying solids, and self-emulsifying semisolids.

Pharmaceutical and/or therapeutic formulations, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical/nutraceutical arts. Such techniques include the step of bringing into association the active ingredient with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. The compositions of the present invention may be formulated into any of a number of possible dosage forms, such as, but not limited to, tablets, capsules, liquid syrups, soft capsules, suppositories, and enemas. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous, oil-based or mixed media. The suspension may further contain substances which increase the viscosity of the suspension, including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. The suspension may also contain a stabilizer. In one embodiment of the present invention, the pharmaceutical composition may be formulated and used in the form of a foam. Pharmaceutical foams include formulations such as, but not limited to, emulsions, microemulsions, creams, jellies, and liposomes. Although these formulations are substantially similar in nature, they differ in the composition and consistency of the final product.

The pharmaceutical compositions described herein may be in unit dosage forms suitable for single administration of precise dosages. In unit dosage form, the preparation is divided into unit doses containing appropriate quantities of one or more compounds. The unit dose can be in the form of a package containing discrete quantities of the formulation. Non-limiting examples are packeted tablets or capsules and powders in vials or ampoules. The aqueous suspension composition may be packaged in a single-dose non-reclosable container. Alternatively, multi-dose reclosable containers may be used, in which case they typically include a preservative in the composition. By way of example only, formulations for parenteral injection may include, but are not limited to, unit dosage forms in ampoules or in multi-dose containers with an added preservative.

The clinician or other skilled person in the pharmacological arts will formulate dosing and administration regimens based on well-known pharmacological and therapeutic considerations, including, but not limited to, the level of therapeutic effect desired and the actual level of therapeutic effect that can be achieved. Generally, it is recommended that chemotherapeutic agents be administered following well-known pharmacological principles (e.g., it is generally recommended that the dosage be changed by no more than 50% at that time, and no more than every 3-4 drugs, agent half-lives). For compositions with relatively little or no dose-related toxicity considerations, and where maximum efficacy is desired, it is not uncommon for the dose to exceed the average required dose. This mode of administration is commonly referred to as a "maximum dose" strategy. In certain embodiments, the compound is administered to the subject at a dose of about 0.01mg/kg to about 200mg/kg, more preferably about 0.1mg/kg to about 100mg/kg, even more preferably about 0.5mg/kg to about 50 mg/kg. When a compound described herein is co-administered with another agent (e.g., as a sensitizer), the effective amount may be lower than when the agent is used alone. Administration may be once daily or multiple times daily for one or more consecutive days.

Method of treatment

The present disclosure provides compounds and methods for inhibiting the activity of ASH 1L. In certain embodiments, the present disclosure provides compounds that bind to and/or inhibit the activity of ASH 1L.

Inhibition of ASH1L activity can be assessed and demonstrated by a variety of means widely known in the art. Non-limiting examples include measuring (a) a direct decrease in ASH1L activity; (b) a decrease in cell proliferation and/or cell viability; (c) an increase in cell differentiation; (d) a decreased level of a downstream target of ASH1L activity; and (e) a reduction in tumor volume and/or tumor volume growth rate. One or more of the above can be determined using kits and commercial assays.

The present disclosure provides compounds and methods for treating a subject afflicted with a disease, comprising administering to the subject a compound or salt described herein, e.g., a compound or salt of any one of formulas (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein. In certain embodiments, the disease is selected from a disease (e.g., cancer) associated with expression (e.g., aberrant expression, overexpression, etc.) and/or activity of ASH 1L. In certain embodiments, the disease is mediated by ASH1L activity and/or expression (e.g., aberrant expression, overexpression, etc.). In certain embodiments, the disease is leukemia, hematologic malignancies, solid tumor cancer, glioma, other cancers, muscular dystrophy, liver fibrosis, and the like.

In some embodiments, the present disclosure provides a method for treating cancer in a subject, comprising administering to the subject a compound or salt described herein, e.g., a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituent and functional group disclosed herein. In some embodiments, the cancer is mediated by ASH1L expression (e.g., aberrant expression, overexpression, etc.) and/or activity. In certain embodiments, the cancer is leukemia, breast cancer, prostate cancer, pancreatic cancer, lung cancer, thyroid cancer, liver cancer, skin cancer, or brain tumor.

In certain embodiments, the present disclosure provides a method of treating a disease in a subject, wherein the method comprises determining whether the subject has an ASH1 l-mediated condition (e.g., cancer), and administering to the subject a therapeutically effective dose of a compound or salt described herein, e.g., a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein.

In some embodiments, ASH1L expression (e.g., aberrant expression, overexpression, etc.) and/or activity has been identified in hematological malignancies, such as cancers affecting the blood, bone marrow, and/or lymph nodes. Thus, certain embodiments relate to administering to a subject having a hematological malignancy a compound or salt described herein, e.g., a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituent and functional group disclosed herein. Such malignancies include, but are not limited to, leukemia and lymphoma. For example, the presently disclosed compounds may be used to treat diseases such as ALL, AML, Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Lymphoma (SLL), Chronic Myelogenous Leukemia (CML), acute monocytic leukemia (AMoL), hairy cell leukemia, and/or other leukemias. In certain embodiments, the compounds or salts of the present disclosure are useful for treating lymphomas, such as hodgkin's lymphoma or all subtypes of non-hodgkin's lymphoma.

Determining whether a tumor or cancer expresses (e.g., over-expresses, aberrantly expresses, etc.) ASH1L can be performed by assessing the nucleotide sequence encoding ASH1L or by assessing the amino acid sequence of ASH 1L. Methods for detecting the ASH1L nucleotide sequence are known to those skilled in the art. These methods include, but are not limited to, polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assays, polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) assays, real-time PCR assays, PCR sequencing, mutant allele specific PCR amplification (MASA) assays, direct sequencing, primer extension reactions, electrophoresis, oligonucleotide ligation assays, hybridization assays, TaqMan assays, SNP genotyping assays, high resolution melting assays, and microarray analysis. Methods for detecting ASH1L protein are known to those skilled in the art. These methods include, but are not limited to, detection using binding agents (e.g., antibodies) specific for ASH1L, protein electrophoresis and western blotting (western blotting), and direct peptide sequencing.

Methods for determining whether a tumor or cancer expresses (e.g., overexpresses, aberrantly expresses, etc.) ASH1L or is mediated by ASH1L activity may use a variety of samples. In some embodiments, the sample is taken from a subject having a tumor or cancer. In some embodiments, the sample is taken from a subject having a cancer or tumor. In some embodiments, the sample is a fresh tumor/cancer sample. In some embodiments, the sample is a frozen tumor/cancer sample. In some embodiments, the sample is a formalin fixed paraffin embedded sample. In some embodiments, the sample is processed into a cell lysate. In some embodiments, the sample is processed into DNA or RNA.

In certain embodiments, the present disclosure provides methods of inhibiting ASH1L activity in a sample comprising administering to the sample comprising ASH1L a compound or salt described herein. The present disclosure provides methods of treating a disease by administering to a subject suffering from the disease a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein, wherein the compound binds ASH1L and/or inhibits ASH1L activity. In certain embodiments, the compound is covalently bound to ASH 1L. In certain embodiments, the compound is non-covalently bound to ASH 1L.

The present disclosure also relates to methods of treating a hyperproliferative disorder in a mammal comprising administering to the mammal a therapeutically effective amount of a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein. In some embodiments, the methods involve treating a cancer, such as acute myelogenous leukemia, juvenile cancer, childhood adrenocortical carcinoma, cancers associated with AIDS such as lymphoma and kaposi's sarcoma, anal cancer, appendiceal cancer, astrocytoma, atypical teratomas, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumor, burkitt's lymphoma, carcinoid tumor, atypical teratomas, embryonic tumor, germ cell tumor, primary lymphoma, cervical cancer, childhood cancer, chordoma, cardiac tumor, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), chronic myeloproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic Ductal Carcinoma In Situ (DCIS), embryonic tumor, leukemia, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, olfactory neuroblastoma, Ewing's sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, ocular cancer, osteocyte cytoma, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, cardiac cancer, liver cancer, Hodgkin's lymphoma, hypopharynx cancer, intraocular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, Lobular Carcinoma In Situ (LCIS), lung cancer, lymphoma, occult primary metastatic squamous neck cancer, mid-line cancer, oral cancer multiple endocrine tumor syndrome, multiple myeloma/plasmacytoid tumor, mycosis fungoides, myelodysplastic syndrome, myelodysplastic/myeloproliferative neoplasm, Multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, cancer of nasal and paranasal sinuses, nasopharyngeal carcinoma, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer (NSCLC), oral cancer, cancer of lips and oral cavity, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paragangliomas, paranasal sinuses and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary Central Nervous System (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, gastric cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-cell lymphoma, testicular cancer, laryngeal cancer, thymoma and thymus cancer, thyroid cancer, transitional cell cancer of renal pelvis and ureter, trophoblastic tumors, childhood cancer, rare cancers, cancer of the kidney and ureter, Urinary tract cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or virus-induced cancer. In some embodiments, the methods relate to treating a non-cancerous hyperproliferative disorder, such as benign hyperplasia of the skin, e.g., psoriasis, restenosis, or prostate, e.g., Benign Prostatic Hypertrophy (BPH). In some cases, the methods involve treating leukemia, hematologic malignancies, solid tumor cancer, prostate cancer (e.g., castration resistant prostate cancer), breast cancer, ewing's sarcoma, osteosarcoma, primary osteosarcoma, T-cell prolymphocytic leukemia, glioma, glioblastoma, liver cancer (e.g., hepatocellular carcinoma), or diabetes.

Subjects that can be treated according to the methods of the present invention with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, stereoisomer, isotopologue, hydrate, or derivative of said compound, include, for example, subjects who have been diagnosed with: acute myelogenous leukemia, juvenile cancer, childhood adrenocortical carcinoma, cancers associated with AIDS such as lymphoma and Kaposi's sarcoma, anal cancer, appendiceal cancer, astrocytoma, atypical teratoma, basal cell carcinoma, cholangiocarcinoma, bladder cancer, bone cancer, brain Stem glioma, brain tumor, breast cancer, bronchial tumor, Burkitt's lymphoma, carcinoid tumor, atypical teratoma, embryonic tumor, germ cell tumor, primary lymphoma, cervical cancer, childhood cancer, chordoma, heart tumor, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), chronic myeloproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic Ductal Carcinoma In Situ (DCIS), embryonic tumor, CNS cancer, endometrial cancer, ependymoma, meningioma, and meningioma, Esophageal cancer, olfactory neuroblastoma, ewing's sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, osteocyte cytoma, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hodgkin's lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, kidney cancer, larynx cancer, lip and oral cancer, liver cancer, Lobular Carcinoma In Situ (LCIS), lung cancer, lymphoma, squamous neck cancer with occult primary metastasis, mid-line cancer, oral cancer multiple endocrine tumor syndrome, multiple myeloma/plasma cell tumor, mycosis fungoides, myelodysplasia syndrome, myelodysplasia/myeloproliferative tumor, multiple myeloma, Merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, cancer of nasal cavity and paranasal sinuses, nasopharyngeal carcinoma, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer (NSCLC), cancer of oral cavity, cancer of lips and oral cavity, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paragangliomas, paranasal sinuses and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary Central Nervous System (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, gastric cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-cell lymphoma, testicular cancer, rare cancer, thymoma and thymus cancer, thyroid cancer, transitional cell cancer of kidney and ureter, trophoblastic cell tumor, childhood cancer, urinary tract cancer, cervical, Uterine sarcoma, vaginal cancer, vulvar cancer, virus-induced cancer, leukemia, hematologic malignancies, solid tumor cancer, prostate cancer, castration-resistant prostate cancer, breast cancer, ewing's sarcoma, osteosarcoma, primary osteosarcoma, T-cell prolymphocytic leukemia, glioma, glioblastoma, hepatocellular carcinoma, liver cancer, or diabetes. In some embodiments, subjects treated with a compound of the invention include subjects who have been diagnosed with: a non-cancerous hyperproliferative disorder, such as benign hyperplasia of the skin, e.g. psoriasis, restenosis or prostate, e.g. Benign Prostatic Hypertrophy (BPH).

The invention further provides methods of inhibiting ASH1L activity by contacting ASH1L with an effective amount of a compound or salt of any one of formulas (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein (e.g., by contacting a cell, tissue or organ that expresses ASH 1L). In some embodiments, the present invention provides methods of inhibiting ASH1L activity in a subject, including but not limited to rodents and mammals (e.g., humans), by administering to the subject an effective amount of a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein. In some embodiments, the percentage of inhibition is greater than 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.

In some embodiments, the present disclosure provides methods of inhibiting ASH1L activity in a cell by contacting the cell with a compound of the invention in an amount sufficient to inhibit the activity. In some embodiments, the present invention provides methods of inhibiting ASH1L activity in a tissue by contacting the tissue with a compound or salt of any one of formulas (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein in an amount sufficient to inhibit ASH1L activity in the tissue. In some embodiments, the present invention provides methods of inhibiting ASH1L activity in an organism (e.g., a mammal, a human, etc.) by contacting the organism with a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein in an amount sufficient to inhibit i ASH1L activity in the organism.

Compositions containing the compounds described herein or salts thereof can be administered for prophylactic and/or therapeutic treatment. In therapeutic applications, the composition is administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease. The amount effective for this use will depend on the severity and course of the disease, previous treatments, the patient's health, weight and response to the drug, and the judgment of the treating clinician.

In prophylactic applications, compositions containing a compound described herein or a salt thereof are administered to a patient susceptible to or otherwise at risk of developing a particular disease, disorder, or condition. Such an amount is defined as a "prophylactically effective amount or dose". In such use, the exact amount will also depend on the health, weight, etc. of the patient. When used in a patient, an effective amount for such use will depend on the severity and course of the disease, condition or disorder, previous treatments, the patient's health and response to the drug, and the judgment of the treating clinician.

In cases where the condition of the patient is not improved, administration of the compound may be carried out chronically, i.e., for an extended period of time, including throughout the life of the patient, as determined by the clinician, in order to alleviate or otherwise control or limit symptoms of the patient's disease.

In cases where the patient's condition does improve, administration of the compound may be given continuously, as determined by the clinician; alternatively, the dose of drug administered may be temporarily reduced or suspended for a period of time (i.e., a "drug holiday"). The length of the drug holiday can vary between 2 days and 1 year, including, by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday can be from about 10% to about 100%, including by way of example only about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.

Once the patient's condition has improved, a maintenance dose is administered as necessary. Subsequently, depending on the symptoms, the dose or frequency of administration, or both, can be reduced to a level at which improvement in the disease, disorder or condition is maintained. However, if any symptoms recur, the patient may require intermittent treatment for a long period of time.

The amount of a given agent that corresponds to such an amount will vary depending on a variety of factors, such as the particular compound, the disease and its severity, the nature (e.g., body weight) of the subject or host in need of treatment, but can nevertheless be determined in a manner well-recognized in the art depending on the particular circumstances of the case, including, for example, the particular agent being administered, the route of administration, the condition being treated, and the subject or host being treated. In general, however, the dosage employed for adult human treatment will generally range from about 0.02 to about 5000mg per day, and in some embodiments from about 1 to about 1500mg per day. The required dose may conveniently be provided in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.

Toxicity and therapeutic efficacy of such treatment regimens can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, determining LD50(dose lethal to 50% of the population) and ED50(a therapeutically effective dose in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as LD50With ED50The ratio of (a) to (b). Compounds exhibiting high therapeutic indices are preferred. Data obtained from cell culture assays and animal studies can be used to formulate a range of dosage for use in humans. The dosage of such compounds is preferably at an ED which includes minimal toxicity50In the circulating concentration range of (c). The dosage may vary within this range depending upon the dosage form employed and the route of administration employed.

Combination therapy

Provided herein are methods for combination therapy, wherein agents known to modulate other pathways or other components of the same pathway or even overlapping groups of target enzymes are used in combination with compounds or salts of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein. In one aspect, such therapies include, but are not limited to, the combination of one or more compounds of the present invention with chemotherapeutic agents, targeting agents, therapeutic antibodies, and radiation therapy to provide synergistic or additive therapeutic effects.

In general, the compositions described herein and other agents in embodiments employing combination therapy need not be administered in the same pharmaceutical composition and may have to be administered by different routes due to different physical and chemical properties. The determination of the mode of administration and, where possible, the feasibility of administration in the same pharmaceutical composition is well known to the clinician. Initial administration can be carried out according to established protocols recognized in the art, and then the dosage, mode of administration, and time of administration can be modified by the clinician based on the observed effect.

In certain instances, it may be appropriate to administer at least one compound described herein in combination with another therapeutic agent. Merely by way of example, if a patient receives one of the compounds herein, such as a compound or salt of any one of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein, and one of the side effects experienced is nausea, it may be appropriate to administer an anti-nausea agent in combination with the initial therapeutic agent. Or by way of example only, the therapeutic effectiveness of one of the compounds described herein can be improved by administering an adjuvant (i.e., the therapeutic benefit of the adjuvant by itself may be minimal, but the overall therapeutic benefit to the patient is improved when combined with another therapeutic agent). Or, by way of example only, the benefit experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (including treatment regimens) that also has therapeutic benefit. In any case, regardless of the disease, disorder, or condition being treated, the overall benefit experienced by the patient may simply be an additive of the two therapeutic agents, or the patient may experience a synergistic benefit.

The particular choice of compound used will depend upon the diagnosis and judgment of the patient's condition and the appropriate treatment regimen. Administration of the compounds can be simultaneous (e.g., simultaneous, substantially simultaneous, or within the same treatment regimen) or sequential, depending on the nature of the disease, disorder, or condition, the condition of the patient, and the actual choice of compounds used. After assessing the disease being treated and the condition of the patient, the clinician is fully able to determine the order of administration of each therapeutic agent and the number of repeated administrations during the treatment regimen.

When the drugs are used in therapeutic combinations, the therapeutically effective dose may vary. Methods for experimentally determining therapeutically effective doses of drugs and other agents for combination treatment regimens are described in the literature. For example, it is widely described in the literature to use a rhythmic administration, i.e. to provide more frequent, lower doses, in order to minimize toxic side effects. Combination therapy further includes periodic treatments that are started and stopped at different times to assist in clinical management of the patient.

For the combination therapies described herein, the dosage of the co-administered compounds will, of course, vary depending on the type of co-drug used, the particular drug used, the disease being treated, and the like. Further, when co-administered with one or more bioactive agents, the compounds provided herein can be administered simultaneously or sequentially with the bioactive agent. If administered sequentially, the attending physician will decide the appropriate order in which to administer the combination of protein and biologically active agent.

In any event, the multiple therapeutic agents (one of which is a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituents and functional groups disclosed herein) can be administered in any order or even simultaneously.

It is to be understood that the dosage regimen for treating, preventing or ameliorating the condition for which relief is sought may be modified depending on a variety of factors. These factors include the disorder or condition from which the subject is suffering and the age, weight, sex, diet and medical condition of the subject. Thus, the actual dosage regimen employed may vary widely and may therefore deviate from the dosage regimen set forth herein.

The agents making up the combination therapies disclosed herein may be in a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The agents that make up the combination therapy may also be administered sequentially, with either therapeutic compound being administered by a regimen that requires two-step administration. A two-step administration regimen may require sequential administration of the active agents or intermittent administration of separate active agents. The time period between the multiple administration steps may range from a few minutes to several hours, depending on the properties of each agent, such as the potency, solubility, bioavailability, plasma half-life and kinetic profile of the agent. Circadian variation in target molecule concentration may also determine the optimal dosage interval.

In addition, the compounds described herein may also be used in combination with procedures that may provide additional or synergistic benefits to the patient. By way of example only, it is contemplated that a patient may receive therapeutic and/or prophylactic benefit according to the methods described herein, wherein a pharmaceutical composition of a compound disclosed herein and/or a combination with other therapeutic agents is combined with genetic testing to determine whether the individual is a carrier of a mutant gene known to be associated with a certain disease or disorder.

The compounds and combination therapies described herein can be administered before, during, or after the onset of disease, and the timing of administration of the composition containing the compound can vary. Thus, for example, the compounds can be used as prophylactic compounds, and can be administered continuously to a subject predisposed to developing a condition or disease, in order to prevent the onset of the disease. The compounds and compositions can be administered during or as soon as possible after the onset of symptoms. Administration of the compound may begin within the first 48 hours of symptom onset, preferably within the first 48 hours of symptom onset, more preferably within the first 6 hours of symptom onset, and most preferably within 3 hours of symptom onset. Initial administration can be via any feasible route, such as intravenous injection, bolus injection, infusion, pill, capsule, transdermal patch, buccal delivery, and the like, or a combination thereof, for example, over about 5 minutes to about 5 hours. It is preferred to administer the compound as soon as possible after the onset of disease is detected or suspected, and for a period of time required to treat the disease, such as, for example, 1 day to about 3 months. The length of treatment may vary for each subject, and the length may be determined using known criteria. For example, the compound or formulation containing the compound may be administered for at least 2 weeks, preferably from about 1 month to about 5 years.

In particular, when the compounds and pharmaceutical compositions herein are used to treat cancer, they may be co-administered with one or more chemotherapeutic agents. Many chemotherapeutic drugs are currently known in the art and may be used in combination with the compounds herein. In some embodiments, the chemotherapeutic agent is selected from the group consisting of: mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, protein-protein interaction inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.

Non-limiting examples are chemotherapeutic agents, cytotoxic agents and non-peptide small molecules, e.g.

Figure BDA0002573038890000971

(imatinib mesylate),

Figure BDA0002573038890000972

(bortezomib), Casodex (bicalutamide),(gefitinib) and doxorubicin, as well as a number of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and Cyclophosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines, such as benzodidopa, carboquone, medopa, and urodopa; ethyleneimine and methyl melamine, including hexamethylmelamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, and trimethylolmelamine; nitrogen mustards, such as chlorambucil, chlorophosphamide, estramustine, ifosfamide, mechlorethamine oxide hydrochloride, melphalan, neonebixin, benzene mustarol, prednissium, trofosfamide, uramustine; aNitroureas, such as carmustine, chlorouramicin, fotemustine, lomustine, nimustine, ranimustine; antibiotics, such as aclacinomycin, actinomycin D (actinomycin), oxytetracycline, azaserine, bleomycin, actinomycin C (cactinomycin), calicheamicin (calicheamicin), carabicin (carabicin), carminomycin, carcinophin (carzinophilin), casodextM, tryptomycin, dactinomycin, daunomycin, ditocin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, isorubicin, idarubicin, milbemycin, mitomycin, mycophenolic acid, nogenin, olivomycin, pelomycin, porphyrinomycin (potfiromycin), puromycin, griseofulvin (quelemin), roxidomicin, pronuclavine, streptozotocin, tubercidin, ubenicillin, stastatin, zoxamine; antimetabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs, such as fludarabine, 6-mercaptopurine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as carpoterone, drostandrosterone propionate, epitioandrostanol, meiandrane, testolactone; anti-adrenaline, such as aminoglutethimide, mitotane, trostane; folic acid supplements, such as folinic acid (frilic acid); acetyl glucurolactone; an aldehydic phosphoramide glycoside; (ii) aminolevulinic acid; amfenadine; bestrabuucil; a bisantrene group; edatrexae; desphosphamide (defofamine); colchicine; mitoquinone (diaziquone); elfosmithine; ammonium etiolate; etoglut; gallium nitrate; a hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidanol; the nitro group can be moistened; pentostatin; methionine mustard (phenamett); pirarubicin; podophyllinic acid; 2-ethyl hydrazide; procarbazine; PSK.RTM.; lezoxan; a texaphyrin; a germanium spiroamine; alternarionic acid; a tri-imine quinone; 2,2',2 "-trichlorotriethylamine; a urethane; vindesine; dacarbazine; mannomustine; dibromomannitol; dibromodulcitol; pipobroman; a polycytidysine; cytarabine("Ara-C"); cyclophosphamide; thiotepa; taxanes such as paclitaxel (TAXOLTM, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERETM, Rhone-Poulenc Rorer, Antonyx, France); retinoic acid; preparing larmycin; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the foregoing. Also included as suitable chemotherapeutic cell modulators are anti-hormonal agents which act to modulate or inhibit the action of hormones on tumors, such as anti-estrogens, including for example tamoxifen (nolvadex), raloxifene, aromatase inhibiting 4(5) -imidazole, 4-hydroxyttamoxifen, trooxifene, keoxifene, LY 117018, onapristone and toremifene (Fareston); and antiandrogens, such as flutamide, nilutamide, bicalutamide, leuprorelin and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs, such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; novier; the tumor can be eliminated; (ii) teniposide; daunomycin; aminopterin; (ii) Hirodad; ibandronate; camptothecin-11 (CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO). Where desired, the compounds or pharmaceutical compositions of the present invention may be used in combination with conventionally prescribed anti-cancer drugs, e.g. ABVD, AVICINE, Abamectin, acridinecarboxamide, Admumab, 17-N-allylamino-17-demethoxygeldanamycin, Alpharadin, Avastin, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, aminonaphthalene unspecific, anthracenedione, anti-CD 22 immunotoxins, antineoplastic agents, apizone, atenolone, atimod, azathioprine, belotecan, bendamustine, BIBW 2992, biricotid, Brostallicin, bryostatin, buthionine sulfoximine, CBV (chemotherapy), calyculin, cell cycle unspecific antineoplastic agents, dichloroacetic acid, beta-glucosidase, and beta-glucosidase,Discodermolide, elsamitrucin, enocitabine, epothilone, eribulin, everolimus, irinotecan, esisullin, minoxidil, forodesine, fosfestrol, an ICE chemotherapy regimen, IT-101, pimecronone, imiquimod, indolocarbazole, ilorffene, Laniquidar, larotaxel, lenalidomide, methidadone, lurtotecan, macsfamide, mitozolamide, nafoxidil, nedaplatin, olaparib, otaxel, PAC-1, bapopo, picraslotron, proteasome inhibitors, fringen, resiquicin, rubitecan, SN-38, Salinosporamide a, sapapatabine, stanford V, swainsonine, talaporfine, tariquiudar, gafur-uracil, tizoxamine, tizotocystin, tixofenamiprid, tritripine, tricitabine, tetritamin, triquetiamide, tretazidovudine, trovamin (2-trovagliptin), trovagliptin, Vadixan, vinflunine, ZD6126, or azaquinad.

Embodiments herein further relate to methods of inhibiting abnormal cell growth or treating a hyperproliferative disorder in a mammal using a compound or salt or pharmaceutical composition of any of formulae (IVa) and (IVb) provided herein having any suitable substituents and functional groups disclosed herein in combination with radiation therapy. Techniques for administering radiation therapy are known in the art, and these techniques may be used for the combination therapies described herein. Administration of a compound of the invention in such combination therapy can be determined as described herein.

Radiation therapy may be administered by one or a combination of several methods, including, but not limited to, external beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, total body radiotherapy, and permanent or temporary interstitial brachytherapy. The term "brachytherapy" as used herein refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near the site of a tumor or other proliferative tissue disease. The term is intended to include, but is not limited to, exposure to radioisotopes (e.g., At-211, I-131, I-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as cell modulators of the invention include solids and liquids. By way of non-limiting example, the radiation source may be a radionuclide, such as I-125, I-131, Yb-169, Ir-192, as a solid source, I-125 as a solid source, or other radionuclide that emits photons, beta particles, gamma radiation, or other therapeutic radiation. The radioactive material may also be a fluid made from any solution of a radionuclide, such as a solution of I-125 or I-131, or a slurry of a suitable fluid containing small particles of a solid radionuclide (e.g., Au-198, Y-90) may be used to produce the radioactive fluid. Furthermore, the radionuclide may be embodied as a gel or radioactive microsphere.

The compounds or pharmaceutical compositions herein are also used in combination with an amount of one or more substances selected from the group consisting of an anti-angiogenic agent, a signal transduction inhibitor, an antiproliferative agent, a glycolytic inhibitor, or an autophagy inhibitor.

Anti-angiogenic agents such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors may be used in combination with the compounds of the present invention and the pharmaceutical compositions described herein. Anti-angiogenic agents include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab. Examples of useful COX-II inhibitors include CELEBREXTM (alecoxib), valdecoxib, and rofecoxib. WO 96/33172 (published 24.10.1996), WO 96/27583 (published 7.3.1996), European patent application No. 97304971.1 (filed 8.7.1997), European patent application No. 99308617.2 (filed 29.10.29.1999), WO 98/07697 (published 26.2.1998), WO 98/03516 (published 29.1.1998), WO98/34918 (published 13.8.1998), WO 98/34915 (published 13.8.1998), WO 98/33768 (published 6.8.6.1998), WO 98/30566 (published 16.7.1998), European patent publication 606,046 (published 13.7.1994), European patent publication 931,788 (published 28.7.1999), WO 90/05719 (published 31.5.31.1990), WO 99/52910 (published 21.10.21.1999), WO 99/52889 (published 21.21.10.1999), Examples of useful matrix metalloproteinase inhibitors are described in WO 99/29667 (published 17.6.1999), PCT International application No. PCT/IB98/01113 (filed 21.7.1998), European patent application No. 99302232.1 (filed 25.3.1999), British patent application No. 9912961.1 (filed 3.3.1999), U.S. provisional application No. 60/148,464 (filed 12.8.1999), U.S. Pat. No. 5,863, 949 (granted 26.26.1.1999), U.S. Pat. No. 5,861,510 (granted 19.1.1999), and European patent publication No. 780,386 (published 25.6.1997), all of which are incorporated herein by reference in their entirety. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity for inhibiting MMP-1. More preferred are those that selectively inhibit MMP-2 and/or AMP-9 relative to other matrix-metalloproteases (e.g., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-ll, MMP-12, and MMP-13). Some specific examples of MMP inhibitors that may be used in the present invention are AG-3340, RO 32-3555, and RS 13-0830.

Autophagy inhibitors include, but are not limited to, chloroquine, 3-methyladenine, hydroxychloroquine (Plaquenil)TM) Bafilomycin a1, 5-amino-4-imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-inhibiting algal toxins that inhibit type 2A or type 1 protein phosphatases, analogs of cAMP, and agents that elevate cAMP levels, such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine. In addition, antisense or siRNA that inhibit the expression of proteins, including but not limited to ATG5 (which is associated with autophagy), can also be used.

In some embodiments, the compounds described herein are formulated or administered in conjunction with a liquid or solid tissue barrier (also referred to as a lubricant). Examples of tissue barriers include, but are not limited to, polysaccharides, seprafilm, interceded, and hyaluronic acid.

In some embodiments, the drug administered in conjunction with the compounds described herein includes any suitable drug that is effectively delivered by inhalation, such as analgesics, e.g., codeine, dihydromorphine, ergotamine, fentanyl, or morphine; angina formulations such as diltiazem; antiallergic agents such as cromoglycate, ketotifen or nedocromil; anti-infectives, such as cephalosporins, penicillins, streptomycins, sulfonamides, tetracyclines or pentamidine; antihistamines, such as methamphetamine; anti-inflammatory agents, such as beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone; antitussives, such as noscapine; bronchodilators, for example ephedrine, epinephrine, fenoterol, formoterol, isoproterenol, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutaline, isotallin, tulobuterol, metaproterenol or (-) -4-amino-3, 5-dichloro-alpha- [ [ [6- [2- (2-pyridyl) ethoxy ] hexyl ] -amino ] methyl ] benzyl alcohol; diuretics, such as amiloride; anticholinergics, such as ipratropium, atropine, or oxitropine; hormones, such as cortisone, hydrocortisone, or prednisolone; xanthines, such as aminophylline, choline theophyllinate, lysine theophyllinate or theophylline; and therapeutic proteins and peptides, such as insulin or glucagon. It will be apparent to those skilled in the art that, where appropriate, the drugs may be used in the form of salts (e.g. as alkali metal or amine salts or as acid addition salts) or as esters (e.g. lower alkyl esters) or as solvates (e.g. hydrates) to optimise the activity and/or stability of the drug.

Other exemplary therapeutic agents that may be used in combination therapy include, but are not limited to, agents as described above, radiation therapy, hormone antagonists, hormones and their release factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, corticotropins; adrenal corticosteroids and their synthetic analogs; inhibitors of adrenocortical hormone synthesis and action, insulin, oral hypoglycemic agents and pharmacological, agents affecting calcification and bone turnover of the endocrine pancreas: calcium, phosphate salts, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting on neuromuscular junctions and/or autonomic ganglia; catecholamines, sympathomimetic agents, and adrenergic receptor agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists.

Other suitable therapeutic agents for co-administration with the compounds herein also include agents for pain and inflammation, such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances produced by bioconversion of products of selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, non-steroidal anti-inflammatory agents, analgesic and antipyretic agents, agents to inhibit prostaglandin and thromboxane synthesis, selective inhibitors of induced cyclooxygenases, selective inhibitors of induced cyclooxygenase-2, autostring, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in both humoral and cellular immune responses, lipid-derived gastrin, eicosanoids, beta-adrenergic agonists, ipratropium, Glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers, and leukotriene inhibitors.

Additional therapeutic agents contemplated for co-administration with the compounds and compositions herein include diuretics, vasopressin, agents affecting the renal protection of water, renin, angiotensin, agents useful for the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, beta-adrenergic receptor antagonists, agents useful for the treatment of hypercholesterolemia, and agents useful for the treatment of dyslipidemia.

Other therapeutic agents contemplated for co-administration with the compounds and compositions herein include drugs for controlling gastric acidity, agents for treating peptic ulcers, agents for treating gastroesophageal reflux disease, prokinetic agents, antiemetics, agents for irritable bowel syndrome, agents for diarrhea, agents for constipation, agents for inflammatory bowel disease, agents for biliary tract disease, agents for pancreatic disease. Therapeutic agents for the treatment of protozoal infections, drugs for the treatment of malaria, amebiasis, giardiasis, trichomoniasis, trypanosomiasis and/or leishmaniasis, and/or drugs for the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones and agents for urinary tract infections, penicillins, cephalosporins and other beta-lactam antibiotics, aminoglycoside containing agents, protein synthesis inhibitors, drugs for chemotherapy of tuberculosis, mycobacterium avium complex disease and leprosy, antifungal agents, antiviral agents, including non-retroviral agents and antiretroviral agents.

Examples of therapeutic antibodies that may be combined with the compounds herein include, but are not limited to, anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, trastuzumab), anti-CD 20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab.

In addition, the methods herein contemplate therapeutic agents for immunomodulation, such as immunomodulators, immunosuppressants, toleragens, and immunostimulants. In addition, therapeutic agents acting on blood and hematopoietic organs, hematopoietic agents, growth factors, minerals and vitamins, anticoagulants, thrombolytic drugs, and antiplatelet drugs.

For the treatment of kidney cancer, the compounds of the invention can be combined with sorafenib and/or avastin. For the treatment of endometrial disorders, the compounds of the present invention may be combined with doxorubicin, taxotere (paclitaxel) and/or cisplatin (carboplatin). For the treatment of ovarian cancer, the compounds of the invention may be combined with cisplatin (carboplatin), taxotere, doxorubicin, topotecan and/or tamoxifen. For the treatment of breast cancer, the compounds of the invention may be combined with taxotere (paclitaxel), gemcitabine (capecitabine), tamoxifen, letrozole, derotaxus, lapatinib, PD0325901, avastin, herceptin, OSI-906 and/or OSI-930. For the treatment of lung cancer, the compounds of the invention may be combined with taxotere (paclitaxel), gemcitabine, cisplatin, pemetrexed, derogatory, PD0325901, and/or avastin.

Further therapeutic agents that may be combined with the compounds herein are found in Goodman and Gilman, tenth edition of the "pharmacological Basis of Therapeutics", compiled by Hardman, Limbird and Gilman, or the Physician's Desk Reference, both of which are incorporated herein by Reference in their entirety.

Depending on the condition being treated, the compounds described herein may be used in combination with the agents disclosed herein or other suitable agents. Thus, in some embodiments, one or more compounds herein will be co-administered with other agents as described above. When used in combination therapy, the compounds described herein are administered simultaneously or separately with a second agent. Such combined administration may include simultaneous administration of both agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, the compounds described herein and any of the above agents may be formulated together in the same dosage form and administered simultaneously. Alternatively, the compound of the invention and any of the above agents may be administered simultaneously, with the two agents being present in separate formulations. In another alternative, the compounds of the invention may be administered only after any of the above agents, and vice versa. In some embodiments of the separate administration regimen, the compound of the invention and any of the above agents are administered several minutes apart, several hours apart, or several days apart.

In some embodiments, the compounds described herein are co-administered with another therapeutic agent effective in the treatment of leukemia and/or other cancers. In some embodiments, the compounds described herein are co-administered with one or more therapeutic agents approved for the treatment of Acute Lymphoblastic Leukemia (ALL), such as: ABITREXATE (METHOTREXATE), ADRIAMYCIN PFS (doxorubicin HCl), ADRIAMYCIN RDF (doxorubicin HCl), ARRANON (nelarabine), Chryseowegian asparaginase, CERUBIDINE (daunorubicin HCl), CLAFEN (cyclophosphamide), CLOFAABINE, CLOFAREX (CLOFARABINE), CLOLAR (CLOFARABINE), cyclophosphamide, cytarabine, CYTOSAR-U (cytarabine), CYTOXAN (cyclophosphamide), Dasatinib, daunomycin HCl, doxorubicin HCl, asparaginase (asparaginase from Chryseowegian), FOLEX (METHOTREXATE), FOLEX PFS (METHOTREXATE), GLEEVEC (imatinib mesylate), ICLUSIG (palettinib HCl), imatinib mesylate, QIBO (vincristine sulfate liposome), METHOTREXATE, METRETE (LPF), Mexate (METHOTREXATE), MEXATE-AQUARE (AQUA), Nexabin (cyclophosphamide), Nexabin (NEOSE), NEOFFIX (TM (METHOTREXATE), CERABIN (DOXA-L) and so-L (CINANOPA) ONCASPAR (peimenase), peimenase, ponatinib hydrochloride, RUBIDOMYCIN (daunomycin hydrochloride), SPRYCEL (dasatinib), TARABINE PFS (cytarabine), VINCASAR PFS (vincristine sulfate), vincristine sulfate, and the like.

In some embodiments, a compound described herein is co-administered with one or more therapeutic agents approved for the treatment of Acute Myeloid Leukemia (AML), such as: ADRIAMYCIN PFS (doxorubicin hydrochloride), ADRIAMYCIN RDF (doxorubicin hydrochloride), arsenic trioxide, CERUBIDINE (daunorubicin hydrochloride), CLAFEN (cyclophosphamide), cyclophosphamide, cytarabine, CyTOSAR-U (cytarabine), CYTOXAN (cyclophosphamide), daunorubicin hydrochloride, doxorubicin hydrochloride, NEOSAR (cyclophosphamide), RUBIMYCIN (daunorubicin hydrochloride), TARABINE PFS (cytarabine), SENTRIOX (arsenic trioxide), VINCASAR PFS (vincristine sulfate), vincristine sulfate, etc.

In some embodiments, the compounds described herein are co-administered with one or more therapeutic agents approved for the treatment of Chronic Lymphocytic Leukemia (CLL), such as: alemtuzumab, AMBOCHLORIN (chlorambucil), amboclorein (chlorambucil), ARZERRA (alfuzumab), bendamustine hydrochloride, CAMPATH (alemtuzumab), chlomenobucilclasn (cyclophosphamide), cyclophosphamide, CYTOXAN (cyclophosphamide), FLUDARA (fludarabine phosphate), fludarabine phosphate, LEUKERAN (chlorambucil), lingilinlifzin (chlorambucil), NEOSAR (cyclophosphamide), alfuzumab, TREANDA (bendamustine hydrochloride), and the like.

In some embodiments, the compounds described herein are co-administered with one or more therapeutic agents approved for the treatment of Chronic Myelogenous Leukemia (CML), such as: BOSULIF (bosutinib), bosutinib, CLAFEN (cyclophosphamide), cyclophosphamide, cytarabine, CYTOSAR-U (cytarabine), CYTOXAN (cyclophosphamide), dasatinib, GLEEVEC (imatinib mesylate), ICLUSIG (panatinib hydrochloride), imatinib mesylate, NEOSAR (cyclophosphamide), nilotinib, homoharringtonine, panatinib hydrochloride, SPRYCEL (dasatinib), SYNRIBO (homoharringtonine), TARABINE PFS (cytarabine), TASIGNA (nilotinib), and the like.

In some embodiments, the compounds described herein are co-administered with one or more therapeutic agents approved for the treatment of meningeal leukemia, such as: cytarabine, Cytosar-U (CYTARABINE), TARABINE PFS (CYTARABINE), etc.

In some embodiments, a compound described herein is co-administered (e.g., for the treatment of cancer) with one or more alkylating agents selected from, for example, nitrogen mustard N-oxide, cyclophosphamide, ifosfamide, thiotepa, ranimustine, nimustine, temozolomide, altretamine, apaziquone, brosplallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, macsfamide, bendamustine, dibromodulcitol, cisplatin, carboplatin, eptaplatin, lobaplatin, nedaplatin, oxaliplatin, and satraplatin.

In some embodiments, a compound described herein is co-administered (e.g., for the treatment of cancer) with one or more antimetabolites selected from, for example, methotrexate, 6-mercaptopurine riboside, mercaptopurine, 5-fluorouracil, tegafur, doxifluridine, carmofur, cytarabine phospholipidate, enocitabine, gemcitabine, fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, edenise, ethynylcytosine nucleoside, cytosine arabinoside, hydroxyurea, melphalan, nelarabine, nolatrexed, alkylphosphate (iosta), disodium pemetrexelate (disodeium premetrexed), pentostatin, pexexol, ranitrexed, triapine, trimetrexate, vidarabine, vincristine, and vinorelbine;

in some embodiments, the compounds described herein are co-administered (e.g., for the treatment of cancer) with one or more hormonal therapy agents selected from, for example, exemestane, lipiamine, anastrozole, doxercalciferol, fadrozole, formestane, abiraterone acetate, finasteride, epristeride, tamoxifen citrate, fulvestrant, triptorelin (Trelstar), toremifene, raloxifene, lasofoxifene, letrozole, sarospirone, epothilone B, vinblastine, vinflunine, docetaxel, and paclitaxel;

in some embodiments, the compounds described herein are co-administered (e.g., for the treatment of cancer) with one or more cytotoxic topoisomerase inhibitors selected from, for example, aclacinomycin, doxorubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflucan, irinotecan, topotecan, edotecan, epirubicin, etoposide, irinotecan, gemmacetan, lurtotecan, mitoxantrone, pirarubicin, picoxolone, rubitecan, sobuzosin, tafluoroporphin, and the like.

In some embodiments, a compound described herein is co-administered (e.g., for treating cancer) with one or more anti-angiogenic compounds selected from, for example, acitretin, aflibercept, angiostatin, aplidine (aplidine), arentar, axitinib, cediranib (recentin), bevacizumab, alanobinib, cilengitide (cilengtlide), combretastatin, DAST, endostatin, fenretinide, halofuginone, pazopanib, lanitumumab, remamastat, removab, ramid, sorafenib, vatalanobine, squalamine, sunitinib, tiratinib, liranolamide, ukrain, and vitaxin.

In some embodiments, the compounds described herein are co-administered (e.g., for treating cancer) with one or more antibodies selected from, for example, trastuzumab, cetuximab, bevacizumab, rituximab, tixemumab, ipilimumab, luximab, cetuximab, asexumab, ogorovolumab, and alemtuzumab.

In some embodiments, a compound described herein is co-administered (e.g., for treating cancer) with one or more VEGF inhibitors selected from, for example, sorafenib, DAST, bevacizumab, sunitinib, cediranib, axitinib, aflibercept, tiratinib, alaninyl briib, vatalanib, pazopanib, and ranibizumab.

In some embodiments, the compounds described herein are co-administered (e.g., for treating cancer) with one or more EGFR inhibitors selected from, for example, cetuximab, panitumumab, victib, gefitinib, erlotinib, and vandetanib (Zactima).

In some embodiments, a compound described herein is co-administered (e.g., for treating cancer) with one or more HER2 inhibitors selected from, for example, lapatinib, trastuzumab (trastuzumab), and pertuzumab; the CDK inhibitor is selected from roscovitine and frodroopin (flavopiridol);

in some embodiments, the compounds described herein are co-administered (e.g., for treating cancer) with one or more proteasome inhibitors selected from, for example, bortezomib and carfilzomib.

In some embodiments, a compound described herein is co-administered with one or more serine/threonine kinase inhibitors (e.g., for the treatment of cancer), e.g., MEK inhibitors and Raf inhibitors, such as sorafenib.

In some embodiments, the compounds described herein are co-administered (e.g., for treating cancer) with one or more tyrosine kinase inhibitors selected from, for example, dasatinib, nilotibib, DAST, bosutinib, sorafenib, bevacizumab, sunitinib, AZD2171, axitinib, aflibercept, tiratinib, imatinib mesylate, brimonib alanine, pazopanib, ranibizumab, vatalantinib, cetuximab, panitumumab, vectizumab, gefitinib, erlotinib, lapatinib, trastuzumab, and pertuzumab.

In some embodiments, the compounds described herein are co-administered (e.g., for the treatment of cancer) with one or more androgen receptor antagonists selected from, for example, nandrolone decanoate, fluoxymesterone, Android, Prostaid, androstine, bicalutamide, flutamide, cyproterone apolipoprotein (apotyprone), tolflutamide (apoflutamide), chlormadinone acetate, anderca, Tabi, cyproterone acetate, and nilutamide.

In some embodiments, the compounds described herein are co-administered (e.g., for the treatment of cancer) with one or more aromatase inhibitors selected from, for example, anastrozole, letrozole, testolactone, exemestane, aminoglutethimide, and formestane.

In some embodiments, the compounds described herein are co-administered with one or more other anti-cancer agents including, for example, alitretinoin, azapridine, atrasentan bexarotene, bortezomib, bosentan, calcitriol, isoxsulalin, fotemustine, ibandronic acid, miltefosine, mitoxantrone, 1-asparaginase, procarbazine, dacarbazine, carbamoylhydroxyurea, pemetrexed, pentostatin, velcade, gallium nitrate, canflunomide, darinapasin, and tretinoin. In preferred embodiments, the compounds of the present disclosure may be used in combination with chemotherapy (e.g., cytotoxic agents), anti-hormones, and/or targeted therapies such as other kinase inhibitors, mTOR inhibitors, and angiogenesis inhibitors.

In embodiments in which the compounds and pharmaceutical compositions herein are used to treat or prevent non-cancer diseases and/or conditions, the compounds and pharmaceutical compositions herein may be co-administered with therapeutic agents and/or therapies known in the art to be useful in treating such diseases and/or conditions.

Reagent kit

Kits and articles of manufacture are also provided for use in the therapeutic applications described herein. In some embodiments, such kits comprise a carrier, package, or divided container for receiving one or more containers, such as vials, tubes, and the like, each of the containers comprising one of the individual elements to be used in the methods described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The container is formed from a variety of materials, such as glass or plastic.

The articles provided herein contain packaging materials. Packaging materials for packaging pharmaceutical products include those found in U.S. patent nos. 5,323,907, 5,052,558, and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for the selected formulation and intended mode of administration and treatment. For example, the container includes a compound or salt of any of formulae (IVa) and (IVb) having any suitable substituent and functional group disclosed herein, optionally in the form of a composition or in combination with another agent disclosed herein. The container optionally has a sterile access port (e.g., the container is an intravenous bag or a vial with a stopper pierceable by a hypodermic injection needle). Such kits optionally comprise a compound having an identifying description or label or instructions relating to its use in the methods described herein.

For example, a kit typically includes one or more additional containers, each container having one or more of the various materials (e.g., reagents, optionally in concentrated form, and/or devices) necessary for the use of the compounds described herein from a commercial and user standpoint. Non-limiting examples of such substances include, but are not limited to, buffers, diluents, filters, needles, syringes; carriers, packages, containers, vials, and/or tube labels listing the contents and/or instructions for use, and package inserts with instructions for use. A set of instructions is also typically included. The label is optionally on or associated with the container. For example, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself, and is associated with the container when present in a receptacle or carrier that also holds the container, e.g., as a package insert. In addition, the label is used to indicate that the contents are to be used for a particular therapeutic application. Further, the label indicates instructions for use of the contents, such as instructions for use in the methods described herein. In certain embodiments, the pharmaceutical composition is present in a pack or dispenser device containing one or more unit dosage forms containing a compound provided herein. The package for example comprises a metal or plastic foil, such as a blister pack. Alternatively, the pack or dispenser device is accompanied by instructions for administration. Alternatively, the package or dispenser is accompanied, in association with the container, by a notice in the form issued by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice reflects human or veterinary approval by said agency of the pharmaceutical form. Such notice is, for example, a label approved by the U.S. food and drug administration for prescription drugs, or an approved product specification. In some embodiments, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.

Experiment of

90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:用于冷冻保存的组合物和其使用方法

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!