Compositions comprising a difluoranyl-1, 4-benzodiazepine compound for the treatment of adenoid cystic carcinoma

文档序号:883389 发布日期:2021-03-19 浏览:8次 中文

阅读说明:本技术 用于治疗腺样囊性癌的包含双氟烷基-1,4-苯并二氮杂*酮化合物的组合物 (Compositions comprising a difluoranyl-1, 4-benzodiazepine compound for the treatment of adenoid cystic carcinoma ) 是由 大卫·西德兰斯基 于 2019-05-14 设计创作,主要内容包括:本发明提供通过施用包含双氟烷基-1,4-苯并二氮杂酮化合物(包括式(III)化合物或其前药)的组合物来治疗或遏制个体的腺样囊性癌(ACC)或抑制ACC肿瘤生长的方法。(The invention provides methods for treating a mammal by administering a composition comprising a difluoroalkyl-1, 4-benzodiazepine Ketone compound (bag)Including a compound of formula (III) or a prodrug thereof) to treat or suppress Adenoid Cystic Carcinoma (ACC) or to inhibit ACC tumor growth in a subject.)

1. A method of treating or suppressing an Adenoid Cystic Carcinoma (ACC) tumor in a subject, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of the following formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

2. A method of inhibiting tumor growth in an individual having an Adenoid Cystic Carcinoma (ACC) tumor, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of the following formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

3. The method of any of claims 1-2, wherein:

R1is-CH2CF3or-CH2CH2CF3(ii) a And is

R2is-CH2CF3or-CH2CH2CF3

4. The method of any of claims 1-3, wherein:

y is zero or 1.

5. The method of any of claims 1-4, wherein:

R1is-CH2CH2CF3(ii) a And is

R2is-CH2CH2CF3

6. The method of any of claims 1-5, wherein:

y is zero.

7. The method of any one of claims 1 to 6, wherein the compound comprises: (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (1); (2R,3S) -N- ((3S) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (2); (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2- (2,2, 2-trifluoroethyl) -3- (3,3, 3-trifluoropropyl) butanediamide (3); (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (2,2, 2-trifluoroethyl) -2- (3,3, 3-trifluoropropyl) butanediamide (4); (2R,3S) -N-((3S) -1- (2H3) methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (5); (2R,3S) -N- ((3S) -7-chloro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (6); (2R,3S) -N- ((3S) -8-methoxy-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (7); (2R,3S) -N- ((3S) -8-fluoro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (8); (2R,3S) -N- ((3S) -7-methoxy-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (9); (2R,3S) -N- ((3S) -7-fluoro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (10); (2R,3S) -N- ((3S) -8-chloro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (11); (2R,3S) -N- ((3S) -9-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (12); (2R,3S) -N- ((3S) -8-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (13); (2R,3S) -N- ((3S) -7-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (14); (2R,3S) -N- ((3S) -8-cyano-9-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (15); (2R,3S) -N- ((3S) -8, 9-dichloro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (16); (2R,3S) -N- ((3S) -9-fluoro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (17); (2R,3S) -N- ((3S) -9-chloro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (18); (2R,3S) -N- ((3S) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (19); (2R,3S) -N- ((3S) -8-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (20); or (2R,3S) -N- ((3S) -9- ((2-methoxyethyl) amino) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (21).

8. The method of any one of claims 1 to 7, wherein the compound comprises:

9. the method of any one of claims 1 to 7, wherein the compound comprises:

10. the method of any one of claims 1 to 9, wherein the composition is administered at a dose of 4 mg.

11. The method of any one of claims 1 to 9, wherein the composition is administered at a dose of 6 or 8.4 mg.

12. The method of any one of claims 1 to 9, wherein the composition is administered at a dose of 0.3, 0.6, 1.2, or 2.4 mg.

13. The method of any one of claims 1 to 12, wherein the composition is administered once per week.

14. The method of any one of claims 1 to 12, wherein the composition is administered biweekly.

15. The method of any one of claims 1 to 14, wherein the ACC tumor comprises tubular ACC, screenable ACC, or solid ACC.

16. The method of any one of claims 1-15, wherein the ACC tumor comprises a Notch activating mutation.

17. The method of claim 16, wherein the Notch activating mutation comprises a Notch1 mutation, a Notch2 mutation, a Notch3 mutation, a Notch4 mutation, or a combination thereof.

18. The method of any one of claims 16-17, wherein the Notch activating mutation functionally inactivates a Negative Regulatory Region (NRR) of the Notch gene.

19. The method of any one of claims 16-18, wherein the Notch activating mutation functionally inactivates the PEST domain of the Notch gene.

20. The method of any one of claims 18-19, wherein the Notch activating mutation comprises a sequence variant in the NRR domain, PEST domain, or a combination thereof of a Notch gene.

21. The method of any one of claims 16-20, wherein the Notch activating mutation comprises a gene rearrangement in the extracellular domain of a Notch gene.

22. The method of claim 21, wherein the gene rearrangement removes a majority of the NRR.

Technical Field

The invention provides methods for treating a mammal by administering a composition comprising a difluoroalkyl-1, 4-benzodiazepineMethods of treating or suppressing Adenoid Cystic Carcinoma (ACC) or inhibiting ACC tumor growth in a subject with a combination of ketone compounds, including a compound of formula (III) or a prodrug thereof.

Background

Many human solid tumors and hematological malignancies exhibit a characteristic dysregulation of Notch pathway signaling. An important step in the activation of Notch receptors is cleavage by gamma secretase, releasing the intracellular signaling domain. Gamma Secretase Inhibitors (GSIs) (e.g. benzodiazepines)Ketone compounds) have potential in having potent antitumor effects.

Patients with advanced solid tumors refractory to standard therapy, patients who relapse after standard therapy, or patients with tumors for which no known effective treatment exists require new strategies for treating solid tumors.

Disclosure of Invention

The present invention provides a method of treating or suppressing an Adenoid Cystic Carcinoma (ACC) tumor in an individual, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

The present invention also provides a method of inhibiting tumor growth in an individual having an Adenoid Cystic Carcinoma (ACC) tumor, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

Drawings

FIG. 1. overall survival of patients with adenoid cystic carcinoma (ACC.) survival of ACC patients with a Notch1 mutation (n ═ 14) and ACC patients with wild-type Notch 1(n ═ 88) is shown (adapted from Ferraroto R et al J Clin Oncol.) -2017; 35: 352-. P is 0.001; OS-overall survival; WT-wild type.

Figure 2a. plasma concentration profile of compound (1) after administration of 4mg of compound (1) -week 1. patients were administered 4mg of compound (1) once a week. Plasma concentrations of compound (1) were exhibited in one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") from 0 to 168 hours after the first treatment with compound (1). Plasma concentrations of compound (1) were determined via validated liquid chromatography-mass spectrometry/mass spectrometry analysis.

Figure 2b. plasma concentration profile of compound (1) after administration of 4mg of compound (1) -week 4. patients were administered 4mg of compound (1) once a week. Plasma concentrations of compound (1) were exhibited in one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") 0 to 168 hours after the fourth week of treatment with compound (1). Plasma concentrations of compound (1) were determined via validated liquid chromatography-mass spectrometry/mass spectrometry analysis.

Figure 2c. pharmacodynamic effect of compound (1) administration on Hes1 expression-week 1. following the weekly administration of 4mg of compound (1) using the quantitative real-time polymerase chain reaction, the expression of Hes1 in the peripheral blood of patients was determined. Data for one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") were presented for up to 168 hours after the first week of treatment with compound (1).

Figure 2d. pharmacodynamic effect of compound (1) administration on Hes1 expression-week 4. following the weekly administration of 4mg of compound (1) using the quantitative real-time polymerase chain reaction, the expression of Hes1 in the peripheral blood of patients was determined. Data for one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") were presented for up to 168 hours after the fourth week of treatment with compound (1).

Figure 2e pharmacodynamic effect of compound (1) administration on DTX1 expression-week 1. DTX1 expression in peripheral blood of patients was determined after weekly administration of 4mg of compound (1) using quantitative real-time polymerase chain reaction. Data for one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") were presented for up to 168 hours after the first week of treatment with compound (1).

Figure 2f pharmacodynamic effect of compound (1) administration on DTX1 expression-week 4. DTX1 expression in peripheral blood of patients was determined after weekly administration of 4mg of compound (1) using quantitative real-time polymerase chain reaction. Data for one patient with partial response ("PR patient") and one patient with stable disease ("SD patient") were presented for up to 168 hours after the fourth week of treatment with compound (1).

Figure 2g. pharmacokinetic/pharmacodynamic effect of compound (1) according to expression of Hes1 relative to baseline 4mg of compound (1) was administered to the patient weekly, and plasma concentrations of compound (1) and Hes1 expression of the patient were plotted. Plasma concentrations of compound (1) were determined via validated liquid chromatography-mass spectrometry/mass spectrometry analysis. Hes1 expression was determined using quantitative real-time polymerase chain reaction. Data are presented for one patient with partial response ("PR patient") and one patient with stable disease ("SD patient").

Figure 3a effect of compound (1) treatment on tumor volume of ACCx9-Notch1 mutant tumors tumor volume was measured in mice administered vehicle or 7.5mg/kg compound (1) over a 35 day period with a treatment schedule of 4 days/3 days off in mice carrying ACCx9-Notch1 mutant tumors. P <0.0001SEM ═ standard error of mean; TGI is tumor growth inhibition.

Figure 3b effect of compound (1) treatment on tumor volume of ACCx11-Notch1 mutant tumors tumor volume was measured in mice administered vehicle or 7.5mg/kg compound (1) over a 35 day period with a treatment schedule of 4 days/3 days off in mice carrying ACCx11-Notch 1. P <0.0001SEM ═ standard error of mean; TGI is tumor growth inhibition.

Figure 3c effect of compound (1) treatment on tumor volume of ACCx6 model tumors in mice administered vehicle or 7.5mg/kg compound (1), tumor volume was measured in mice carrying ACCx6 over a 35 day period with a treatment time course of 4 days/3 days off. NS is not statistically significant; SEM is standard error of mean; TGI is tumor growth inhibition.

Figure 3d effect of compound (1) treatment on tumor volume of ACCx5M1 model mutant tumors in mice administered vehicle or 7.5mg/kg compound (1), tumor volume was measured in mice bearing ACCx5M1 tumors over a 35 day period with a treatment schedule of 4 days/3 days off. NS is not statistically significant; SEM is standard error of mean; TGI is tumor growth inhibition.

FIG. 3E effect of combination therapy of Compound (1) with cisplatin or Everolimus (everolimus) on tumor volume of ACCx9-Notch1 mutant tumors after administration of vehicle, Compound (1) (7.5mg/kg PO with 4 days/off for 3 days), cisplatin (3mg/kg, IP once a week), Everolimus (10mg/kg, once a day); a combination of compound (1) (5mg/kg, PO, 4 days/day off 3 days) and cisplatin (3mg/kg, IP, qwk); or compound (1) (3mg/kg, PO with 4 days/rest for 3 days) and everolimus (10mg/kg, PO, qd), tumor volume was measured in mice bearing ACCx9-Notch1 mutant tumors over a 35 day period. P<0.0001;P<0.05; NS is not statistically significant; SEM is standard error of mean; TGI is tumor growth inhibition.

FIG. 3F. Effect of combination therapy of Compound (1) with cisplatin or Everolimus on tumor volume of ACCx11-Notch1 mutant tumors after vehicle administration, Compound (1) (7.5mg/kg PO with 4 days/day off for 3 days), cisplatin (3mg/kg, IP once a week), Everolimus (10mg/kg, once a day); a combination of compound (1) (5mg/kg, PO, 4 days/day off 3 days) and cisplatin (3mg/kg, IP, qwk); or compound (1) (3mg/kg, PO with 4 days/rest for 3 days) and everolimus (10mg/kg, PO, qd), tumor volume was measured in mice carrying ACCx11-Notch1 over a 35 day period. P<0.0001;P<0.05; NS is not statistically significant; SEM is standard error of mean; TGI is tumor growth inhibition.

Figure 4a. immunohistochemical staining of ACCx11-Notch1 mutant tumors after treatment with compound (1) mice bearing ACCx11-Notch1 mutant tumors were administered vehicle or 7.5mg/kg compound (1) with a treatment schedule of 4 days/3 days off. Tumor tissues of mice treated with vehicle and compound (1) were evaluated by immunohistochemistry for Notch1 endodomain (NICD1), MYC, Ki67 and HES 1.

Figure 4b immunohistochemical staining of ACCx5M1 model tumors after treatment with compound (1) mice bearing ACCx5M1 tumor were administered vehicle or 7.5mg/kg compound (1) in a treatment schedule of 4 days/3 days off. Tumor tissues of mice treated with vehicle and compound (1) were evaluated by immunohistochemistry for Notch1 endodomain (NICD1), MYC, Ki67 and HES 1.

Figure 5a effect of compound (1) treatment on tumor volume of ACCx9-Notch1 mutant tumors tumor volume of mice bearing ACCx9-Notch1 mutant tumors was measured over a 25 day period with a treatment schedule of 4 days/3 days off in mice administered vehicle or 7.5mg/kg compound (1). P <0.0001SEM ═ standard error of the mean.

FIG. 5B effect of MRK-003 treatment on tumor volume of ACCx9-Notch1 mutant tumors tumor volume was measured in mice administered vehicle or 150mg/kg MRK-003 once a week or 300mg/kg MRK-003 once a week over a 25 day period in mice carrying ACCx9-Notch 1. MRK-003 is a small molecule that is a Gamma Secretase Inhibitor (GSI) (Stoeck A et al Cancer discovery 2014; 4: 1154-. P <0.05 SEM-standard error of mean.

FIG. 5℃ Effect of Brontizumab (broncutuzumab) treatment on tumor volume of ACCx9-Notch1 mutant tumors tumor volume was measured over a 21 day period in mice bearing ACCx9-Notch1 mutant tumors administered with vehicle or monoclonal antibody Brontizumab (Ferrarotto R et al J. Clin Oncology 2017; 35:352-360, incorporated herein by reference), 10mg/kg twice weekly. P <0.05 SEM-standard error of mean.

Detailed Description

In the following detailed description, numerous specific details are set forth in order to provide a thorough understanding of the invention. However, it will be understood by those skilled in the art that the present invention may be practiced without these specific details. In other instances, well-known methods, procedures, and components have not been described in detail so as not to obscure the present invention.

In one embodiment, the composition of the invention or the composition for use in the method of the invention comprises one or more gamma secretase inhibitors, one or more Notch inhibitors, or a combination thereof. In one embodiment, the gamma secretase inhibitor comprises a difluoroalkyl-1, 4-benzodiazepineA ketone compound.

Bifluoroalkyl-1, 4-benzodiazepinesKetone compounds

In one embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (I):

and/or at least one salt thereof, wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In one embodiment, the invention provides a composition comprising a compound as described herein, formulated at a dose of 4 mg. In one embodiment, the present invention provides a composition comprising a compound as described herein, formulated for intravenous administration.

In one embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (II):

wherein R is3Is H or-CH3(ii) a And y is zero or 1.

In one embodiment, the present invention provides a composition comprising a compound of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, R1is-CH2CF3or-CH2CH2CF3And R is2is-CH2CF3or-CH2CH2CF3. In another embodiment, R1is-CH2CH2CF3And R is2is-CH2CH2CF3. In one embodiment, y is 1 or 2. In another embodiment, y is zero or 1. In another embodiment, y is zero.

In one embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (1)

In another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzoDiaza derivatives-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (2)

In another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2- (2,2, 2-trifluoroethyl) -3- (3,3, 3-trifluoropropyl) butanediamide (3);

in another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (2,2, 2-trifluoroethyl) -2- (3,3, 3-trifluoropropyl) butanediamide (4);

in another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -1-, (2H3) Methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepines-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (5);

in another embodiment, the compound of formula (III) comprises a compound of formula (VI):

in one embodiment, it comprises (2R,3S) -N- ((3S) -7-chloro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (6), i.e. Y ═ H and Z ═ Cl; (2R,3S) -N- ((3S) -8-methoxy-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (7), i.e. Y ═ OCH3And Z ═ H; (2R,3S) -N- ((3S) -8-fluoro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (8), i.e. Y ═ F and Z ═ H; (2R,3S) -N- ((3S) -7-methoxy-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (9), Y ═ H and Z ═ OCH3(ii) a (2R,3S) -N- ((3S) -7-fluoro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (10), i.e. Y ═ H and Z ═ F; or (2R,3S) -N- ((3S) -8-chloro-1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (11), i.e. Y ═ Cl and Z ═ H.

In another embodiment, the compound of formula (III) comprises a compound of formula (VII):

in one embodiment, it comprises (2R,3S) -N- ((3S) -9-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (12), i.e. X ═ OCH3Y ═ H and Z ═ H; (2R,3S) -N- ((3S) -8-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (13), i.e. X ═ H, Y ═ OCH3And Z ═ H; (2R,3S) -N- ((3S) -7-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (14), i.e. X ═ H, Y ═ H and Z ═ OCH3(ii) a (2R,3S) -N- ((3S) -8-cyano-9-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (15), i.e. X ═ OCH3Y ═ CN and Z ═ H; (2R,3S) -N- ((3S) -8, 9-dichloro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (16), i.e., X ═ Cl, Y ═ Cl and Z ═ H; (2R,3S) -N- ((3S) -9-fluoro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (17), i.e., X ═ F, Y ═ H, and Z ═ H; or (2R,3S) -N- ((3S) -9-chloro-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (18), i.e. X ═ Cl, Y ═ H and Z ═ H.

In another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (19);

in another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -8-methoxy-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (20)

In another embodiment, the compound of formula (III) comprises: (2R,3S) -N- ((3S) -9- ((2-methoxyethyl) amino) -2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (21)

In another embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (I):

and/or at least one salt thereof, wherein:

R1is-CH2CF3

R2is-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2、

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently Cl, C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3and/or-O (cyclopropyl);

each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is 1 or 2.

In another embodiment, ring a is phenyl; and R is3Is H. In another embodiment, R2is-CH2CH2CF3(ii) a And ring a is phenyl. In another embodiment, R2is-CH2CH2CF3(ii) a Ring A is phenyl; raIs C1-3Alkyl or-CH2OH; each RbIndependently F and/or Cl; and y is 1.

In another embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (IV):

in another embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (V):

wherein R is3Is H or Rx

In another embodiment, the present invention provides a composition comprising: (2R,3S) -N- ((3S) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (22); (2R,3S) -N- ((3S) -5- (3-chlorophenyl) -9-ethyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (23); (2R,3S) -N- ((3S) -5- (3-chlorophenyl) -9-isopropyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (24); (2R,3S) -N- (9-chloro-5- (3, 4-dimethylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (25); (2R,3S) -N- (9-chloro-5- (3, 5-dimethylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (26); (2R,3S) -N- ((3S) -9-Ethyl-5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (27); (2R,3S) -N- ((3S) -5- (3-chlorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (28); (2R,3S) -N- ((3S) -5- (3-chlorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (29); (2R,3S) -N- ((3S) -5- (3-methylphenyl) -2-oxo-9- (trifluoromethyl) -2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (30); (2R,3S) -N- ((3S) -9-chloro-5- (3, 5-dimethylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (31); (2R,3S) -N- ((3S) -5- (3-methylbenzene)2-oxo-9- (trifluoromethyl) -2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (32); (2R,3S) -N- ((3S) -9-isopropyl-5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (33); (2R,3S) -N- ((3S) -9- (Cyclopropoxy) -5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (34); (2R,3S) -N- ((3S) -9- (Cyclopropoxy) -5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (35); (2R,3S) -N- ((3S) -9-chloro-5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (36); (2R,3S) -N- ((3S) -9-methyl-2-oxo-5- (3- (trifluoromethyl) phenyl) -2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -3- (4,4, 4-trifluorobutyl) -2- (3,3, 3-trifluoropropyl) butanediamide (37); (2R,3S) -N- ((3S) -9-methyl-2-oxo-5- (3- (trifluoromethyl) phenyl) -2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (38); (2R,3S) -N- ((3S) -9-chloro-5- (2-methylphenyl) -2-oxo-2, 3-dihydro-1H-)1, 4-benzodiazepines-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (39); (2R,3S) -N- ((3S) -5- (4-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (40); (2R,3S) -N- ((3S) -9-chloro-5- (3-cyclopropylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (41); (2R,3S) -N- ((3S) -5- (3-chlorophenyl) -9-methoxy-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (42); (2R,3S) -N- ((3S) -5- (4-chlorophenyl) -9-methoxy-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (43); (2R,3S) -N- ((3S) -9-chloro-5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (44); (2R,3S) -N- ((3S) -5- (3-methylphenyl) -9-methoxy-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (45); (2R,3S) -N- ((3S) -5- (4- (hydroxymethyl) phenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (46); (2R,3S) -N- ((3S) -5- (2-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (47); (2R,3S) -N- ((3S) -5- (3-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (48); (2R,3S) -N- ((3S) -9-methoxy-2-oxo-5- (5- (trifluoromethyl) -2-pyridinyl) -2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (49); (2R,3S) -N- ((3S) -5- (5-chloro-2-pyridinyl) -9-methoxy-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (50); (2R,3S) -N- ((3S) -5- (4-methoxyphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (51); (2R,3S) -N- ((3S) -5- (4-methylphenyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide (52); (2R,3S) -N- ((3S) -5- (3-fluorophenyl) -9- (hydroxymethyl) -2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) succinamide (53); l-valine ((3S) -3- (((2R,3S) -3-carbamoyl-6, 6, 6-trifluoro-2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -5- (3-Fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-1-yl) methyl ester (54); l-alanine ((3S) -3- (((2R,3S) -3-carbamoyl-6, 6, 6-trifluoro-2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-1-yl) methyl ester (55); s- (((2S,3R) -6,6, 6-trifluoro-3- (((3S) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) carbamoyl) -2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -L-cysteine (56); s- (((2S,3R) -6,6, 6-trifluoro-3- (((3S) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) carbamoyl) -2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -L-cysteine tert-butyl ester (57); s- (((2S,3R) -6,6, 6-trifluoro-3- (((3S) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) carbamoyl) -2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -L-cysteine methyl ester (58); (4- (Phosphomycin) phenyl) acetic acid ((3S) -3- (((2R,3S) -3-carbamoyl-6, 6, 6-trifluoro-2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-1-yl) methyl ester (59); and L-valyl-L-valine ((3S) -3- (((2R,3S) -3-carbamoyl-6, 6, 6-trifluoro-2- (3,3, 3-trifluoropropyl) hexanoyl) amino) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-1-yl) methyl ester (60); and salts thereof.

In another embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (I):

and/or at least one salt thereof, wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2

With the proviso that if ring A is phenyl, z is zero, and y is 1 or 2, then at least one RaIs composed of

C1-3Alkyl, -CH2OH、-CF3Cyclopropyl or-O (cyclopropyl);

with the proviso that if R3Is RxThen R4Is H; and is

With the proviso that if R4Is RyThen R3Is H or-CH3

In another embodiment, a structure as described above includes one or more of the following conditions: with the proviso that if ring A is phenyl, z is zero, and y is 1 or 2, then at least one RaIs C1-3Alkyl, -CH2OH、-CF3Cyclopropyl or-O (cyclopropyl); with the proviso that if R3Is RxThen R4Is H; and with the proviso that if R is4Is RyThen R3Is H or-CH3

In another embodiment, the present invention provides a composition comprising a compound represented by the following structure:

in another embodiment, a compound as described herein comprises a prodrug of one or more of the compounds.

U.S. patent No. 9,273,014, which is incorporated herein by reference in its entirety, discloses various compounds of formula (I):

and/or at least one salt thereof, wherein:

R1is-CH2CH2CF3

R2is-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx

R4Is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OCH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently Cl, C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3and/or-O (cyclopropyl);

each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is 1 or 2.

U.S. patent No. 9,273,014 also discloses compounds of formula (22):

in one embodiment, the chemical name is (2R,3S) -N- ((3S) -5- (3-fluorophenyl) -9-methyl-2-oxo-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide. U.S. patent No. 9,273,014 also discloses a method of synthesizing compounds and other compounds of formula (I), which are considered part of the present invention.

U.S. patent No. 8,629,136, which is incorporated herein by reference in its entirety, discloses compounds of formula (III):

and/or at least one salt thereof, wherein:

R3is H or-CH3(ii) a And is

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3

U.S. patent No. 8,629,136 also discloses compounds of formula (1):

in one embodiment, the chemical name is (2R,3S) -N- ((3S) -1-methyl-2-oxo-5-phenyl-2, 3-dihydro-1H-1, 4-benzodiazepine-3-yl) -2, 3-bis (3,3, 3-trifluoropropyl) butanediamide. In one embodiment, the compound is a Notch inhibitor. U.S. Pat. No. 8,629,136 discloses a method for synthesizing compounds and other compounds of formula (I) which are considered to be the present inventionIn the clear.

The present invention may be embodied in other specific forms without departing from its spirit or essential attributes. The present invention encompasses all combinations of aspects and/or embodiments of the invention noted herein. It is to be understood that any and all embodiments of the present invention may be combined with any other embodiment or embodiments to describe yet further embodiments. It should also be understood that each individual element of an embodiment is intended to be combined with any and all other elements from any embodiment to describe other embodiments.

Combination therapy

In one embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (I) as described herein, as monotherapy or in combination therapy with one or more anticancer agents.

In another embodiment, the present invention provides a composition comprising a compound represented by the structure of formula (I) as described herein, as monotherapy or in combination therapy with one or more chemotherapeutic agents.

In one embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the compositions of the invention or for use in the methods of the invention comprise one or more cancer therapeutic agents and one or more of the above-described bis-fluoroalkyl-1, 4-benzodiazepineCombination therapy of ketone compounds.

In the treatment of cancer, a combination of chemotherapeutic agents and/or other treatments (e.g., radiation therapy) is often advantageous. The additional agent may have the same or a different mechanism of action than the primary therapeutic agent. For example, drug combinations may be employed wherein two or more drugs administered act in different ways or in different stages of the cell cycle, and/or wherein two or more drugs have different toxicities or side effects, and/or wherein the drugs combined each have the therapeutic effect exhibited in treating the particular disease condition presented by the patient.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and Eribulin (Eribulin).

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and vinorelbine (vinorelbine).

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and FOLFIRI. In one embodiment, the FOLFIRI comprises folinic acid (leucovorin), fluorouracil (5-FU) and Irinotecan (Irinotecan) (phenotha). In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and folinic acid (leucovorin), fluorouracil (5-FU), irinotecan (kemptol), or a combination thereof.

In one embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and one or more targeted therapeutic agents. In one embodiment, the targeted therapeutic comprises a mammalian target of rapamycin (mTOR) inhibitor. In one embodiment, the mTOR inhibitor comprises Everolimus (Everolimus). In another embodiment, the mTOR inhibitor comprises sirolimus (rapamycin). In another embodiment, the mTOR inhibitor comprises temsirolimus (temsirolimus).

In another embodiment, the mTOR inhibitor comprises a dual mammalian target of rapamycin/phosphoinositide 3-kinase inhibitor, which in one embodiment comprises NVP-BEZ235 (dactylixib), GSK2126458, XL765, or a combination thereof.

In another embodiment, the mTOR inhibitor comprises a second generation mTOR inhibitor, which in one embodiment comprises AZD8055, INK128/MLN0128, OSI027, or a combination thereof.

In another embodiment, the mTOR inhibitor comprises a third generation mTOR inhibitor, which in one embodiment comprises RapaLink.

In one embodiment, the composition of the invention comprises one or more compounds represented by the structure of formula (I) as described herein, in combination with an mTOR inhibitor and a chemotherapeutic agent. In one embodiment, the mTOR inhibitor comprises everolimus. In one embodiment, the chemotherapeutic agent comprises cisplatin.

In one embodiment, the composition of the present invention comprises one or more compounds represented by the structure of formula (I) as described herein and a PARP (poly ADP-ribose polymerase) inhibitor.

In another embodiment, the compositions of the invention comprise one or more compounds represented by the structure of formula (I) as described herein and a multifunctional alkylating agent. In one embodiment, the multifunctional alkylating agent comprises a nitrosourea, a mustard, a nitrogen mustard, a mesylate, busulfan, an ethyleneimine, or a combination thereof.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and a steroid.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and a bisphosphonate.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and a cancer growth blocker.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and a proteasome inhibitor.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and one or more interferons.

In another embodiment, the compositions of the present invention comprise one or more compounds represented by the structure of formula (I) as described herein and one or more interleukins.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an alkylating agent. In one embodiment, the alkylating agent comprises Procarbazine (Procarbazine) (toluidine (Matulane)), Dacarbazine (Dacarbazine) (DTIC), Altretamine (Altretamine) (Hexalen), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an alkylating drug. In one embodiment, the alkylating agent comprises cisplatin (Platinol).

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an antimetabolite. In one embodiment, the antimetabolite comprises a folate compound (methotrexate), an amino acid antagonist (azaserine), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a purine antagonist. In one embodiment, the purine antagonist comprises mercaptopurine (6-MP), thioguanine (6-TG), Fludarabine Phosphate (Fladarabine Phosphate), Cladribine (Cladribine (Leustatin)), Pentostatin (Pentostatin) (Nipent), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a pyrimidine antagonist. In one embodiment, the pyrimidine antagonist comprises fluorouracil (5-FU), cytarabine (ARA-C), azacitidine, or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a plant alkaloid. In one embodiment, the plant alkaloid comprises Vinblastine (Vinblastine) (Velban), Vincristine (Vincristine) (ancorin), etoposide (VP-16, VePe-sid), Teniposide (Teniposide) (Vumon), Topotecan (Topotecan) (and Hycamtin), Irinotecan (Irinotecan) (captopr), Paclitaxel (Paclitaxel) (Taxol), Docetaxel (Taxotere), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an antibiotic. In one embodiment, the antibiotic comprises an anthracycline, Doxorubicin (Doxorubicin) (Adriamycin), Rubex, polychrome (Doxil), Daunorubicin (Daunorubicin) (DaunoXome), actinomycin d (dactinomycin) (dactinomycin (Cosmegen)), idarubicin (idarubicin) (Idamycin), Plicamycin (Plicamycin) (Mithramycin), Mitomycin (Mitomycin) (Mutamycin), Bleomycin (Bleomycin) (bleoxane), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a cancer vaccine. In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an immunotherapeutic agent. In one embodiment, the immunotherapeutic agent comprises a monoclonal antibody. In one embodiment, the monoclonal antibody comprises an anti-PD-1 antibody, which in one embodiment comprises nivolumab (nivolumab).

In another embodiment, the monoclonal antibody comprises aktuzumabTrastuzumabBevacizumabCetuximabOr a combination thereof. In another embodiment, the monoclonal antibody comprises a radiolabeled antibody,in one embodiment, the composition comprises ibritumomab (briumomab), tezetanOr a combination thereof. In another embodiment, the monoclonal antibody comprises a chemically labeled antibody, which in one embodiment comprises a belumacizumab vildagliptinAddo-trastuzumab emtansine (Ado-trastuzumab emtansine) ((R))Also known as TDM-1), Dineukin (denileukin bifitor)Or a combination thereof. In another embodiment, the monoclonal antibody comprises a bispecific antibody, and in one embodiment comprises brilmoto (Blincyto).

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a hormonal therapy. In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a hormonal agent. In one embodiment, the hormonal agent comprises Tamoxifen (Tamoxifen) (novaladex), Flutamide (Eulexin), gonadotropin releasing hormone agonists (Leuprolide) and Goserelin (Goserelin), aromatase inhibitors, Aminoglutethimide (Aminoglutethimide), Anastrozole (Anastrozole), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and Amsacrine (Amsacrine), hydroxyurea (hydrae), asparaginase (El-spar), Mitoxantrone (Mitoxantrone) (Novantrone), Mitotane (Mitotane), retinoic acid derivatives, bone marrow growth factor, Amifostine (Amifostine), or combinations thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an agent that inhibits one or more cancer stem cell pathways. In one embodiment, such agents comprise an inhibitor of Hedgehog, WNT, BMP, or a combination thereof.

In one embodiment, the anti-cancer agent comprises a BCMA-targeted chimeric antigen receptor T cell immunotherapeutic, a P53-HDM2 inhibitor, a c-MET inhibitor, a BCR-ABL inhibitor, an anti-interleukin-1 b monoclonal antibody, an EGFR mutation modulator, a PI 3K-a inhibitor, a JAK1/2 inhibitor, a cortisol synthesis inhibitor, thrombopoietin, a P selectin inhibitor receptor agonist, an anti-CD 20 monoclonal antibody, an anti-PD-1 monoclonal antibody, a signal transduction inhibitor, a CDK4/6 inhibitor, a BRAF inhibitor + MEK inhibitor, a CD 19-targeted chimeric antigen receptor T cell immunotherapeutic, a somatostatin analog, or a combination thereof. In one embodiment, the anti-cancer agent comprises carminatinib (capmatiib), aximinib (asciminib), canakinumab (canakinumab), apraxib (aspelisib), tyrosine protein kinase inhibitor (ruxolitinib), osirtat (osidorstat), eltoprap (eltrombopag), clelizumab (grizzlizumab), ofatumumab (otatumab), spatializumab (spavatazumab), midostaurin (midostaurin), rebocillin (ribociclociclib), daraflavinib (dabrafrafenib) + trametinib, tique (tisagenlecellectel), everolimus (everolimus), pasanide (iresatide), or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a method of hematopoietic stem cell transplantation.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a method of isolation infusion. In one embodiment, the split infusion method comprises infusing a chemotherapeutic agent into specific tissues so that a very large dose of the chemotherapeutic agent is delivered to the tumor site without causing overwhelming systemic damage.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a targeted delivery mechanism. In one embodiment, the targeted delivery mechanism increases the effective level of chemotherapeutic agent against tumor cells while decreasing the effective level against other cells to increase tumor specificity and/or reduce toxicity. In one embodiment, the targeted delivery mechanism comprises a conventional chemotherapeutic agent or a radioisotope or an immunostimulatory factor.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and a nanoparticle. In one embodiment, the nanoparticles are used as carriers for water-soluble agents (such as paclitaxel). In one embodiment, nanoparticles made of magnetic material may also be used to concentrate the agent at the tumor site using an externally applied magnetic field.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an agent for treating Adenoid Cystic Carcinoma (ACC). In one embodiment, the agent for treating ACC comprises Axitinib (Axitinib), Bortezomib (Bortezomib) (Velcade)), Bortezomib (Bortezomib) + doxorubicin, Cetuximab (Cetuximab), Cetuximab + Intensity Modulated Radiation Therapy (IMRT), Cetuximab + RT + cisplatin, Cetuximab + cisplatin +5-FU, cidalimide (Chidamide) (CS055/HBI-8000), Cetuximab and carbonium (Carbon Ion), cisplatin and 5-FU, doxorubicin and bleomycin, doxorubicin and adriamycin and cyclophosphamide, Dasatinib (Dasatinib), dolivitinib (dolivitinib), Epirubicin (Epirubicin), fibatinib (gemcitib), Gemcitabine (Gemcitabine), Gemcitabine and Imatinib (Imatinib), and Imatinib (Imatinib + Imatinib) Mitoxantrone (Mitoxanthrone), MK 2206, Nelfinavir (Nelfinavir), paclitaxel and carboplatin, Panitumumab (Panitumumab) and radiotherapy, PF-00562271, PF-00299804 and non-gemumab (Figitumab) PX-478, PX-866, Regafenib (Regorafenib), Sonepuzumab (Sonepcizumab), Sorafenib (Sorafenib), Sunitinib (Sunitinib), vinorelbine and cisplatin, Vorinostat (Vorinostat), XL147 and Erlotinib (Erlotinib), XL647 or combinations thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and palbociclumab, docetaxel, nivolumab and ipulimab (ipilimumab), PSMA-PET imaging, sidaxan, APG-115, HDM201, DS-3032b, LY3039478, or a combination thereof.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and an agent for treating triple negative breast cancer. In one embodiment, the agent for treating triple negative breast cancer comprises a PARP (poly ADP ribose polymerase) inhibitor, such as olaparib; VEGF (vascular endothelial growth factor) inhibitors, such as bevacizumab; EGFR (epidermal growth factor receptor) targeted therapies, such as cetuximab; or a combination thereof.

In one embodiment, there is provided a method of treating cancer comprising administering to a mammal in need thereof a composition as described herein and administering one or more anti-cancer agents.

In one embodiment, the phrase "anti-cancer agent" refers to an agent selected from any one or more of the following: alkylating agents (including nitrogen mustards, mesylates, busulfans, alkyl sulfonates, nitrosoureas, ethylenimine derivatives, and triazenes, or combinations thereof); anti-angiogenic agents (including matrix metalloproteinase inhibitors); antimetabolites (including adenosine deaminase inhibitors, folic acid antagonists, purine analogs, and pyrimidine analogs); antibiotics or antibodies (including monoclonal antibodies, CTLA-4 antibodies, anthracyclines); an aromatase inhibitor; a cell cycle response modifier; enzymes; farnesase-protein transferase inhibitors; hormones and anti-hormonal agents and steroids (including synthetic analogs, glucocorticoids, estrogens/anti-estrogens [ e.g., SERMs ], androgens/anti-androgens, progestins, progesterone receptor agonists, and luteinizing hormone releasing [ LHRH ] agonists and antagonists); insulin-like growth factor (IGF)/insulin-like growth factor receptor (IGFR) system modulators (including IGFR1 inhibitors); an integrin signaling inhibitor; kinase inhibitors (including multi-kinase inhibitors or Src kinase or Src/ab1 inhibitors), cyclin dependent kinase [ CDK ] inhibitors, panHer, Her-1 and Her-2 antibodies, VEGF inhibitors (including anti-VEGF antibodies), EGFR inhibitors, PARP (poly ADP ribose polymerase) inhibitors, mitogen-activated protein [ MAP ] inhibitors, MET inhibitors, MEK inhibitors, aurora kinase inhibitors, PDGF inhibitors and other tyrosine kinase inhibitors or serine/threonine kinase inhibitors; microtubule disruptors, such as ecteinascidin (ecteinascidin) or analogs and derivatives thereof; microtubule stabilizing agents such as taxane, platinum-based antineoplastic agents (platinum) such as cisplatin, carboplatin, oxaliplatin, nedaplatin, terraplatin tetranitrate, phenanthreneplatinum (phenonthrilatin), picoplatin and satraplatin, and naturally occurring epothilones and synthetic and semi-synthetic analogs thereof; microtubule-associated, labile agents (including vinca alkaloids); a topoisomerase inhibitor; prenyl-protein transferase inhibitors; a platinum coordination complex; a signal transduction inhibitor; and other agents useful as anti-cancer and cytotoxic agents, such as biological response modifiers, growth factors, and immunomodulators.

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and any one or more of: revlimid (Revlimid), avastin, herceptin, merosal (Rituxan), European Divow (Opdivo), Gleevec (Gleevec), Ibruvicat (Imbruvica), Velcade (Velcade), Zetika (Zytiga), Enzalutamide (Xtandi), Litai (Alimta), Gardesic (Gadasil), Iblance (Ibrance), Pageit, Tacina (Tasigna), Dinosame, Afinicor (Afinitor), Lusolinib (Jakafi), Tarceva (Tarcea), Kbax, Sucant (Suvot), Yivolt, Dojimei (Nexavar), Nureyd, Aibizalex, Dazaeda (Xodela), Jiawa, Verentzizhu (Verentq).

In another embodiment, the present invention provides a composition comprising one or more compounds represented by the structure of formula (I) as described herein and any one or more of: bomacillin (abemaciclib), ecadostat (epacadostat), apraclonide (apalcumide), Carfilzomib (Carfilzomib), Crizotinib (Crizotinib) (PF-02341066), GDC-0449 (vismodegib (vismodeib)), Oncovix, PLX4032(RG7204), Ponatinib (Ponatinib), SGN-35 (Belntemavidon), Tivozanib (Tivozanib) (AV-951), T-DM1 (trastuzumab-DM 1), and XL184 (cabozantinib)).

Thus, the compositions of the present invention may be administered in combination with other anti-cancer therapies suitable for the treatment of cancer or other proliferative diseases. The invention herein further comprises the use of a composition of the invention in the manufacture of a medicament for the treatment of cancer, and/or a package comprising a composition of the invention, together with instructions for the use of the composition in combination with other anti-cancer or cytotoxic agents and for the treatment of cancer.

In one embodiment, any of the methods as described herein comprises the steps of: administering to the subject a composition comprising a compound represented by the structure of formula (I) as described herein, as monotherapy or in combination therapy with one or more anti-cancer agents. In another embodiment, any of the methods as described herein comprises the steps of: administering to the subject a composition comprising a compound represented by the structure of formula (I) as described herein, as monotherapy or in combination therapy with one or more chemotherapeutic agents.

In another embodiment, any of the methods as described herein comprises the steps of: administering to the subject a composition comprising a compound represented by the structure of formula (III) as described herein, as monotherapy or in combination therapy with one or more anti-cancer agents. In another embodiment, any of the methods as described herein comprises the steps of: administering to the subject a composition comprising a compound represented by the structure of formula (III) as described herein, as monotherapy or in combination therapy with one or more chemotherapeutic agents.

In one embodiment, the anti-cancer agent or chemotherapeutic agent in the methods of the invention is administered to the subject as a single composition having a compound represented by the structure of formula (I) or a compound represented by the structure of formula (III). In another embodiment, the anti-cancer or chemotherapeutic agent is administered to the subject in a composition separate from the composition comprising the compound represented by the structure of formula (I) or the compound represented by the structure of formula (III). In one embodiment, the individual compositions are administered simultaneously. In another embodiment, separate compositions are administered to the individual at separate times, at separate sites of administration, or a combination thereof.

In one embodiment, there is provided a method for treating cancer comprising administering to a mammal in need thereof a compound of formula (I); administering cisplatin; and optionally administering one or more additional anti-cancer agents.

In one embodiment, there is provided a method of treating cancer comprising administering to a mammal in need thereof a compound of formula (I); administering dasatinib; and optionally administering one or more additional anti-cancer agents.

In one embodiment, there is provided a method of treating cancer comprising administering to a mammal in need thereof a compound of formula (I); administering paclitaxel; and optionally administering one or more additional anti-cancer agents.

In one embodiment, there is provided a method of treating cancer comprising administering to a mammal in need thereof a compound of formula (I); administering tamoxifen; and optionally administering one or more additional anti-cancer agents.

In one embodiment, a method for treating cancer is provided comprising administering to a mammal in need thereof a compound of formula (I), administering a glucocorticoid; and optionally administering one or more additional anti-cancer agents. An example of a suitable glucocorticoid is dexamethasone (dexamethasone).

In one embodiment, there is provided a method of treating cancer comprising administering to a mammal in need thereof a compound of formula (I), administering carboplatin; and optionally administering one or more additional anti-cancer agents.

The compounds of the present invention may be formulated or co-administered with other therapeutic agents selected for their particular usefulness in addressing the side effects associated with the aforementioned conditions. For example, the compounds of the present invention may be formulated with medicaments to prevent nausea, allergies and gastric irritation, such as analgesics and H1And H2An antihistamine.

In one embodiment, a pharmaceutical composition is provided comprising a compound of formula (I) or a prodrug thereof; one or more additional agents selected from kinase inhibitors (small molecules, polypeptides and antibodies), immunosuppressive agents, anticancer agents, antiviral agents, anti-inflammatory agents, antifungal agents, antibiotics or anti-vascular hyperproliferative compounds; and any pharmaceutically acceptable carrier, adjuvant or vehicle.

The above other therapeutic agents (when employed in combination with the compounds of the invention) may be used, for example, in those amounts indicated in the Physicians' Desk Reference manual or as otherwise determined by one of ordinary skill.

Pharmaceutical composition

Formulations

Also encompassed within the invention are pharmaceutical compositions comprising a compound of formula (I) and one or more non-toxic, pharmaceutically acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier" materials) and, if desired, other active ingredients.

The compounds of formula (I) may be administered by any suitable route, preferably in the form of pharmaceutical compositions suitable for such routes, and in dosages effective for the desired treatment. The compounds and compositions of the present invention may be administered, for example, in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants and vehicles. For example, a pharmaceutical carrier may contain a mixture of mannitol or lactose and microcrystalline cellulose. The mixture may contain additional components such as lubricants, e.g. magnesium stearate, and disintegrants, e.g. crospovidone. The carrier mixture may be filled into gelatin capsules or compressed in tablet form. The pharmaceutical compositions may be administered, for example, in oral dosage forms or by infusion.

For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, a capsule, a liquid capsule, a suspension, or a liquid. The pharmaceutical compositions are preferably prepared in the form of dosage units containing the active ingredient in the specified amounts. For example, the pharmaceutical composition may be provided as a tablet or capsule comprising the active ingredient in an amount ranging from about 1 to 2000mg, preferably from about 1 to 500mg, and more preferably from about 5 to 150 mg. Suitable daily dosages for humans or other mammals may vary widely depending on the condition of the patient and other factors, but can be determined using conventional methods.

Any pharmaceutical composition contemplated herein can be delivered orally, e.g., via any acceptable and suitable oral formulation. Exemplary oral formulations include, but are not limited to, tablets, troches, buccal tablets, aqueous and oily suspensions, dispersible powders or granules, emulsions, hard and soft capsules, liquid capsules, syrups and elixirs, for example. Pharmaceutical compositions intended for oral administration may be prepared according to any method known in the art for the manufacture of pharmaceutical compositions intended for oral administration. To provide a pharmaceutically palatable preparation, the pharmaceutical compositions according to the invention may contain at least one agent chosen from sweetening agents, flavouring agents, colouring agents, demulcents, antioxidants and preserving agents.

Tablets may be prepared, for example, by mixing at least one compound of formula (I) with at least one non-toxic pharmaceutically acceptable excipient suitable for the manufacture of tablets. Exemplary excipients include, but are not limited to, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate; granulating and disintegrating agents, such as microcrystalline cellulose, croscarmellose sodium, corn starch and alginic acid; binding agents, such as starch, gelatin, polyvinylpyrrolidone and acacia; and lubricating agents such as magnesium stearate, stearic acid and talc. In addition, the tablets may be uncoated or coated by known techniques to mask the unpleasant taste of an unpleasant drug or to delay disintegration and absorption of the active ingredient in the gastrointestinal tract, thereby maintaining the effect of the active ingredient for a longer period of time. Exemplary water-soluble taste-masking materials include, but are not limited to, hydroxypropyl methylcellulose and hydroxypropyl cellulose. Exemplary time delay materials include, but are not limited to, ethyl cellulose and cellulose acetate butyrate.

Hard gelatin capsules may be prepared, for example, by mixing at least one compound of formula (I) with at least one inert solid diluent, for example calcium carbonate; calcium phosphate; and kaolin.

Soft gelatin capsules may be prepared, for example, by mixing at least one compound of formula (I) with at least one water-soluble carrier, for example polyethylene glycol, and at least one oil medium, for example peanut oil, liquid paraffin, and olive oil.

Aqueous suspensions may be prepared, for example, by mixing at least one compound of formula (I) with at least one excipient suitable for the manufacture of aqueous suspensions. Exemplary excipients suitable for the manufacture of aqueous suspensions include, but are not limited to, suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, alginic acid, polyvinylpyrrolidone, tragacanth and acacia; dispersing or wetting agents, such as naturally occurring phosphatides, for example lecithin; condensation products of alkylene oxides with fatty acids, such as polyoxyethylene stearate; condensation products of ethylene oxide with long chain aliphatic alcohols, such as heptadecaethyleneoxycetanol; condensation products of ethylene oxide with partial esters derived from fatty acids and hexitols, such as for example polyoxyethylene sorbitol monooleate; and condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, such as, for example, polyethylene sorbitan monooleate. The aqueous suspensions may also contain at least one preservative, such as ethyl and n-propyl p-hydroxybenzoate; at least one colorant; at least one flavoring agent; and/or at least one sweetener including, but not limited to, sucrose, saccharin, and aspartame.

Oily suspensions may be prepared, for example, by suspending at least one compound of formula (I) in a vegetable oil, such as for example arachis oil, olive oil, sesame oil and coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may also contain at least one thickening agent, for example beeswax; hard paraffin wax; and cetyl alcohol. To provide a palatable oily suspension, at least one of the sweeteners already described above and/or at least one flavoring agent may be added to the oily suspension. The oily suspensions may further contain at least one preservative including, but not limited to, antioxidants, such as butylated hydroxyanisole and alpha-tocopherol.

Dispersible powders and granules can be prepared, for example, by mixing at least one compound of formula (I) with at least one dispersing and/or wetting agent; at least one suspending agent; and/or at least one preservative. Suitable dispersing, wetting and suspending agents are as described above. Exemplary preservatives include, but are not limited to, antioxidants, such as ascorbic acid. In addition, dispersible powders and granules may also contain at least one excipient, including but not limited to sweeteners; a flavoring agent; and a colorant.

Emulsions of at least one compound of formula (I) may, for example, be prepared as oil-in-water emulsions. The oil phase of the emulsion comprising the compound of formula (I) may be constituted in a known manner by known ingredients. The oily phase may be provided by, but is not limited to, vegetable oils (e.g., olive oil and peanut oil), mineral oils (e.g., liquid paraffin), and mixtures thereof. While the phase may comprise only emulsifiers, it may comprise a mixture of at least one emulsifier with a fat or oil or both a fat and an oil. Suitable emulsifiers include, but are not limited to, for example, naturally occurring phospholipids, such as soy lecithin; esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. Preferably, a hydrophilic emulsifier is included along with a lipophilic emulsifier that acts as a stabilizer. It is also preferred to include both oil and fat. And emulsifiers, with or without stabilizers, constitute the so-called emulsifying waxes, and the waxes, together with oils and fats, constitute the so-called emulsifying ointment base, which forms the oily dispersed phase of the cream formulation. The emulsion may also contain sweetening agents, flavouring agents, preservatives and/or antioxidants. Emulsifiers and emulsion stabilizers suitable for use in the formulations of the present invention include Tween 60, Span 80, cetearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate, alone or with waxes or other materials well known in the art.

In another embodiment, the compound of formula (I) may be formulated as a nanoparticle, a lipid nanoparticle, a microparticle, or a liposome.

The compounds of formula (I) may also be delivered intravenously, subcutaneously and/or intramuscularly, for example via any pharmaceutically acceptable and suitable injectable form. Exemplary injectable forms include, but are not limited to, sterile aqueous solutions containing an acceptable carrier and solvent, such as water, Ringer's solution, and isotonic sodium chloride solution; a sterile oil-in-water microemulsion; and aqueous or oily suspensions. For example, compositions may be provided for intravenous administration comprising an amount of the active ingredient in the range of about 0.2 to 150 mg. In another embodiment, the active ingredient is present in the range of about 0.3 to 10 mg. In another embodiment, the active ingredient is present in the range of about 4 to 8.4 mg. In one embodiment, the active ingredient is administered at a dose of about 4 mg. In another embodiment, the active ingredient is administered at a dose of about 6 mg. In another embodiment, the active ingredient is administered at a dose of about 8.4 mg.

In another embodiment, the active ingredient is administered at a dose of about 0.3 mg. In another embodiment, the active ingredient is administered at a dose of about 0.6 mg. In another embodiment, the active ingredient is administered at a dose of about 1.2 mg. In another embodiment, the active ingredient is administered at a dose of about 2.4 mg.

Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration, or by using other suitable dispersing or wetting agents and suspending agents. The compound can be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum and/or various buffersIn the liquid. Other adjuvants and modes of administration are well known and widely known in the pharmaceutical arts. The active ingredient may also be administered by injection as a composition with a suitable carrier, including saline, dextrose, or water, or with a cyclodextrin (i.e.,) Co-solvent solubilization (i.e., propylene glycol) or micelle solubilization (i.e., Tween 80).

The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids, such as oleic acid, are used in the preparation of injectables.

Sterile injectable oil-in-water microemulsions may be prepared, for example, by: 1) dissolving at least one compound of formula (I) in an oil phase, such as a mixture of soybean oil and lecithin; 2) combining formula (I) containing an oil phase with a water and glycerol mixture; and 3) treating the combination to form a microemulsion.

Sterile aqueous or oily suspensions may be prepared according to methods known in the art. For example, sterile aqueous solutions or suspensions may be prepared with a non-toxic parenterally acceptable diluent or solvent (e.g. 1, 3-butanediol); and sterile oily suspensions may be prepared with sterile, non-toxic, acceptable solvents or suspending media (e.g., sterile, fixed oils, such as synthetic mono-or diglycerides); and fatty acid (e.g., oleic acid) production.

Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, Self Emulsifying Drug Delivery Systems (SEDDS) (e.g., d-alpha-tocopheryl polyethylene glycol 1000 succinate), surfactants used in pharmaceutical dosage forms (e.g., the Tween series)Polyethoxylated castor oil (e.g. castor oil)Surfactants (BASF)) or other similar polymeric delivery matrices, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates, glycine, sorbic acid), potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene block polymers, polyethylene glycol, and wool fat. Cyclodextrins, such as alpha, beta and gamma cyclodextrins, or chemically modified derivatives, such as hydroxyalkyl cyclodextrins, including 2-hydroxypropyl-cyclodextrin and 3-hydroxypropyl-cyclodextrin, or other solubilized derivatives, may also be advantageously used to enhance delivery of the compounds of the formulae described herein.

The pharmaceutically active compounds of the present invention can be processed according to pharmaceutically acceptable methods to produce medicaments for administration to patients, including humans and other mammals. The pharmaceutical compositions may be subjected to conventional pharmaceutical procedures, such as sterilization, and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers, and the like. In addition, tablets and pills can be prepared with enteric coatings. Such compositions may also contain adjuvants such as wetting agents, sweetening, flavoring and perfuming agents.

The amount of compound administered and the dosing regimen for treating a disease condition with a compound and/or composition of the invention depends on a variety of factors including age, weight, sex, individual medical condition, type of disease, severity of disease, route and frequency of administration, and the particular compound employed. Thus, the dosing regimen may vary widely, but can be routinely determined using standard methods. A daily dose of about 0.001 to 100mg per kg body weight, preferably between about 0.005 and about 50mg per kg body weight and most preferably between about 0.01 to 10mg per kg body weight may be appropriate.

For therapeutic purposes, the active compounds of the invention are usually combined with one or more adjuvants appropriate for the indicated route of administration. If administered orally, the compound can be blended with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, gum arabic, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration. Such capsules or tablets may contain a controlled release formulation, as may be provided in a dispersion of the active compound in hydroxypropylmethyl cellulose.

The pharmaceutical compositions of the present invention comprise at least one compound of formula (I) and/or at least one salt thereof, and optionally an additional agent selected from any pharmaceutically acceptable carrier, adjuvant and vehicle. Alternative compositions of the invention comprise a compound of formula (I) as described herein or a prodrug thereof and a pharmaceutically acceptable carrier, adjuvant or vehicle.

The compound according to formula (I) may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or the amount of compound of formula (I) to be delivered. The compounds and compositions of the invention may be administered, for example, orally, mucosally or parenterally, including intravascularly, intraperitoneally, subcutaneously, intramuscularly and intrasternally. In one embodiment, the compounds and compositions of the present invention are administered intravenously.

Application method

In one embodiment, the invention provides the use of a compound or composition described for treating, suppressing or inhibiting a proliferative disease in a subject.

In another embodiment, the invention provides a method of treating, suppressing or inhibiting a proliferative disease in a subject, comprising the steps of: administering to the individual a composition comprising one or more compounds of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the invention provides a method of treating, suppressing or inhibiting a proliferative disease in a subject, comprising the steps of: administering to the subject a composition comprising one or more compounds of formula (III):

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the compound is administered at a dose of approximately 0.3, 0.6, 1.2, 2.4, 4, 6, or 8.4 mg.

In one embodiment, the compound is administered intravenously at a dose of approximately 0.3, 0.6, 1.2, 2.4, 4, 6, or 8.4 mg. In another embodiment, the compound is administered weekly at a dose of approximately 0.3, 0.6, 1.2, 2.4, 4, 6, or 8.4 mg.

In another embodiment, the present invention provides a method of treating, suppressing or inhibiting a proliferative disease in a subject, comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) as described above, wherein the compound is administered at a dose of about 4 mg. In one embodiment, the compound is administered intravenously at a dose of approximately 4 mg. In another embodiment, the compound is administered weekly at a dose of approximately 4 mg.

In another embodiment, the present invention provides a method of treating, suppressing or inhibiting a proliferative disease in a subject, comprising the step of administering to the subject a composition consisting essentially of one or more compounds represented by the structure of formula (I) as described above. In another embodiment, the present invention provides a method of treating, suppressing or inhibiting a proliferative disease in a subject, comprising the step of administering to the subject a composition consisting of one or more compounds represented by the structure of formula (I) as described above.

In one embodiment, the present invention provides the use of a therapeutically acceptable amount of one or more compounds or compositions as described herein for treating, suppressing, or inhibiting a proliferative disease in a subject. In another embodiment, the present invention provides the use of a therapeutically effective amount of one or more compounds or compositions as described herein for treating, suppressing or inhibiting a proliferative disease in a subject. In another embodiment, the present invention provides the use of a synergistically effective amount of one or more compounds or compositions as described herein for treating, suppressing or inhibiting a proliferative disease in a subject. In another embodiment, the present invention provides the use of a synergistically therapeutically effective amount of one or more compounds or compositions as described herein for treating, suppressing or inhibiting a proliferative disease in a subject.

In one embodiment, the proliferative disease comprises a hard fiber tumor.

In one embodiment, the proliferative disease comprises a precancerous condition or a benign proliferative disorder.

In one embodiment, the term "pre-cancerous" or alternatively "pre-malignant" as used herein interchangeably refers to a disease, syndrome, or other condition accompanied by an increased risk of cancer. Precancerous conditions in the context of the present invention include, but are not limited to: breast calcification, vaginal intraepithelial neoplasia, Barrett's esophagus, atrophic bronchitis, congenital dyskeratosis, iron-deficient dysphagia, lichen planus, oral submucosa fibrosis, actinic keratosis, solar elastosis, cervical dysplasia, leukoplakia, and erythema.

In one embodiment, the term "benign hyperproliferative disorder" as used herein refers to a condition of abnormal growth and differentiation of cells and an increased amount of organic tissue caused by cell proliferation. Benign hyperproliferative disorders may be due to a lack of response or an inappropriate response to a regulator, or alternatively to a dysfunction of a regulator. Non-limiting examples of benign hyperproliferative disorders are psoriasis and Benign Prostatic Hyperplasia (BPH).

In another embodiment, the proliferative disease comprises cancer.

In one embodiment, the cancer comprises a solid tumor. In another embodiment, the cancer comprises a hematologic malignancy.

In one embodiment, an individual as described herein has cancer. In one embodiment, the term "cancer" in the context of the present invention includes all types of neoplasms, whether in the form of solid tumors or non-solid tumors, and includes malignant and pre-malignant conditions as well as metastases thereof.

In one embodiment, the cancer is a carcinoma, sarcoma, myeloma, leukemia, or lymphoma. In another embodiment, the cancer is a mixed type.

In one embodiment, the mixed type cancer comprises several types of cells. The type components may be within one category or from different categories. Some examples are: adenosquamous carcinoma; mixed mesodermal tumors; a carcinosarcoma; teratocarcinoma.

In another embodiment, the carcinoma comprises Adenoid Cystic Carcinoma (ACC).

In another embodiment, the carcinoma comprises a gastroesophageal junction carcinoma.

In one embodiment, the carcinoma is an adenocarcinoma. In another embodiment, the carcinoma is a squamous cell carcinoma.

In one embodiment, the sarcoma comprises an osteosarcoma or an osteogenic sarcoma (bone); chondrosarcoma (cartilage); leiomyosarcoma (smooth muscle); rhabdomyosarcoma (skeletal muscle); mesothelioma or mesothelioma (the membranous lining of the body cavity); fibrosarcoma (fibrous tissue); angiosarcoma or angioendothelioma (blood vessels); liposarcoma (adipose tissue); glioma or astrocytoma (neurogenic connective tissue found in the brain); myxosarcoma (primitive embryonic connective tissue); and stromal tumors or mixed mesodermal tumors (mixed connective tissue class).

In one embodiment, the cancer comprises myeloma, which in one embodiment is a cancer that originates from bone marrow plasma cells. Plasma cells produce some of the proteins found in blood. In one embodiment, the cancer comprises multiple myeloma.

In another embodiment, the cancer comprises leukemia ("non-solid tumor" or "blood cancer"), which in one embodiment is a cancer of the bone marrow (site of blood cell production). In one embodiment, the leukemia includes myeloid or myelocytic leukemia (malignancies of the myeloid and granulocytic leukocyte groups), lymphoglobular or lymphoblastic leukemia (malignancies of the lymphoid and lymphoglobular blood cell groups), and polycythemia vera or erythrocytosis (malignancies of various blood cell products, but with primarily erythrocytes).

In another embodiment, the cancer comprises T-cell acute lymphoblastic leukemia (T-ALL). In another embodiment, the cancer comprises T-lymphoblastic leukemia/lymphoma (TLL). In another embodiment, the cancer comprises Chronic Lymphocytic Leukemia (CLL).

In another embodiment, the cancer comprises lymphoma. In one embodiment, the lymphoma comprises extranodal lymphoma. In one embodiment, the lymphoma comprises hodgkin's lymphoma. In another embodiment, the lymphoma comprises non-hodgkin's lymphoma. In one embodiment, the lymphoma comprises marginal zone B cell lymphoma, diffuse large B cell lymphoma, or mantle cell lymphoma.

In another embodiment, the cancer is dependent on Notch activation. In another embodiment, the cancer comprises a Notch activating mutation. In another embodiment, the cancer is not dependent on Notch activation.

In one embodiment, the present invention provides a method of treating cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the invention provides a method of treating cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention provides a method of treating cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the present invention provides a method of treating cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of treating a cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the invention provides a method of treating a carcinoma, wherein the carcinoma comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the invention provides a method of treating a cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the invention provides a method of treating a cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of treating ACC, wherein the ACC comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the present invention provides a method of treating ACC, wherein the ACC comprises one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention provides a method of treating ACC, wherein the ACC comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the present invention provides a method of treating ACC, wherein the ACC comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of reducing tumor size in an individual having a cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of reducing tumor size in an individual having a carcinoma, wherein the carcinoma comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In one embodiment, the present invention provides a method of reducing tumor size in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the invention provides a method of reducing tumor size in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention provides a method of reducing tumor size in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the present invention provides a method of reducing tumor size in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, reducing the tumor size comprises reducing the tumor size by 25% -95%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 25%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 30%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 35%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 40%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 45%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 50%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 55%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 60%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 65%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 70%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 75%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 80%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 85%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 90%. In another embodiment, reducing the tumor size comprises reducing the tumor size by 95%.

In one embodiment, the present invention provides a method of reducing tumor volume in an individual having cancer, wherein the cancer comprises one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of reducing tumor volume in an individual having a carcinoma, wherein the carcinoma comprises one or more Notch activating genetic variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In one embodiment, the present invention provides a method of reducing tumor volume in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN, -OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In another embodiment, the present invention provides a method of reducing tumor volume in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention provides a method of reducing tumor volume in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the present invention provides a method of reducing tumor volume in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, reducing the tumor volume comprises reducing the tumor volume by 25% -95%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 25%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 30%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 35%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 40%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 45%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 50%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 55%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 60%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 65%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 70%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 75%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 80%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 85%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 90%. In another embodiment, reducing tumor volume comprises reducing tumor volume by 95%.

In one embodiment, the present invention provides a method of suppressing tumor growth in a subject having a tumor, wherein one or more cells of the tumor comprise one or more Notch activating genetic variations, the method comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of suppressing tumor growth in a subject having a carcinoma, wherein one or more cells of the carcinoma comprise one or more Notch activating genetic variations, the method comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In one embodiment, the present invention provides a method of suppressing tumor growth in a subject having ACC, wherein one or more cells of the ACC comprise one or more Notch gene variations, the method comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In another embodiment, the invention provides a method of suppressing tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention provides a method of suppressing tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in another embodiment, the present invention provides a method of suppressing tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising:

in one embodiment, administration of a composition as described herein suppresses tumor growth by 20% -99% compared to an untreated tumor, or compared to a tumor treated with another anti-cancer therapy. In another embodiment, tumor growth is arrested by 20-35%. In another embodiment, tumor growth is arrested by 35-50%. In another embodiment, tumor growth is arrested by 50-75%. In another embodiment, tumor growth is arrested by 75-90%. In another embodiment, tumor growth is arrested by 90-99%.

In another embodiment, tumor growth is arrested by 20%. In another embodiment, tumor growth is arrested by 25%. In another embodiment, tumor growth is arrested by 30%. In another embodiment, tumor growth is arrested by 35%. In another embodiment, tumor growth is arrested by 40%. In another embodiment, tumor growth is arrested by 45%. In another embodiment, tumor growth is arrested by 50%. In another embodiment, tumor growth is arrested by 55%. In another embodiment, tumor growth is arrested by 60%. In another embodiment, tumor growth is arrested by 65%. In another embodiment, tumor growth is arrested by 70%. In another embodiment, tumor growth is arrested by 75%. In another embodiment, tumor growth is arrested by 80%. In another embodiment, tumor growth is arrested by 85%. In another embodiment, tumor growth is arrested by 90%. In another embodiment, tumor growth is arrested by 95%. In another embodiment, tumor growth is arrested by 99%.

In one embodiment, the present invention provides a method of inhibiting tumor growth in a subject having a tumor, wherein one or more cells of the tumor comprise one or more Notch activating genetic variations, the method comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of inhibiting tumor growth in an individual having a cancer, wherein one or more cells of the cancer comprise one or more Notch activating genetic variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In one embodiment, the present invention provides a method of inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the present invention provides a method of inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, comprising the step of administering to the individual a composition comprising:

in one embodiment, inhibiting tumor growth comprises 100% reduction in tumor growth compared to a control group.

In one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having a tumor, wherein one or more cells of the tumor comprise one or more Notch activating genetic variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein.

In one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having a carcinoma, wherein one or more cells of the carcinoma comprise one or more Notch activating genetic variations, the method comprising the step of administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the individual a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof,

wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H, -CH3Or Rx;

R4is H or Ry

RxThe method comprises the following steps: -CH2OC(O)CH(CH3)NH2、-CH2OC(O)CH(NH2)CH(CH3)2、-CH2OC(O)CH((CH(CH3)2)NHC(O)CH(NH2)CH(CH3)2

RyThe method comprises the following steps: -SCH2CH(NH2)C(O)OH、-SCH2CH(NH2)C(O)OH3or-SCH2CH(NH2)C(O)OC(CH3)3

Ring a is phenyl or pyridyl;

each RaIndependently F, Cl, -CN,-OCH3、C1-3Alkyl, -CH2OH、-CF3Cyclopropyl, -OCH3-O (cyclopropyl) and/or-NHCH2CH2OCH3

Each RbIndependently F, Cl, -CH3、-CH2OH、-CF3Cyclopropyl and/or-OCH3

y is zero, 1 or 2; and is

z is zero, 1 or 2.

In one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, the method comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of increasing progression-free survival or overall survival of an individual having ACC, wherein one or more cells of the ACC comprise one or more Notch activating gene variations, comprising the step of administering to the individual a composition comprising:

in one embodiment, the present invention provides a method of reducing the size of a tumor or suppressing or inhibiting tumor growth in an individual having a cancer, wherein the cancer does not contain a Notch GOF mutation, the method comprising the steps of: administering to the subject a first composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein and a second composition comprising an additional anti-cancer agent.

In one embodiment, the present invention provides a method of reducing tumor size or suppressing or inhibiting tumor growth in an individual having a cancer, wherein the cancer Notch GOF mutation, the method comprising the steps of: administering to the subject a first composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein and a second composition comprising an additional anti-cancer agent.

In one embodiment, the present invention provides a method of reducing tumor size or suppressing or inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC do not contain a Notch GOF mutation, the method comprising the steps of: administering to the subject a first composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof as described herein and a second composition comprising an additional anti-cancer agent.

In another embodiment, the present invention provides a method of reducing tumor size or suppressing or inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC do not contain a Notch GOF mutation, the method comprising the steps of: administering to the subject a first composition comprising one or more compounds represented by the structure of formula (III) as described herein and a second composition comprising an additional anti-cancer agent.

In another embodiment, the present invention provides a method of reducing tumor size or suppressing or inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC do not contain a Notch GOF mutation, the method comprising the steps of: administering to the individual a first composition comprising:

and a second composition comprising an additional anti-cancer agent.

In another embodiment, the present invention provides a method of reducing tumor size or suppressing or inhibiting tumor growth in an individual having ACC, wherein one or more cells of the ACC do not contain a Notch GOF mutation, the method comprising the steps of: administering to the individual a first composition comprising:

and a second composition comprising an additional anti-cancer agent.

In one embodiment, the anti-cancer agent comprises eribulin. In another embodiment, the anticancer agent comprises vinorelbine. In one embodiment, the combination therapy is administered to an individual, wherein the Notch activating gene variation does not comprise a Notch GOF mutation.

In another embodiment, the cancer comprises astrocytoma, bladder cancer, breast cancer, cholangiocarcinoma (CCA), colon cancer, colorectal cancer, epithelial ovarian cancer, fibrosarcoma, gallbladder cancer, gastric cancer, neuroblastoma, glioma, head and neck cancer, hepatocellular cancer, renal cancer, liver cancer, lung cancer (including non-small cell lung cancer (NSCLC)), Malignant Fibrous Histiocytoma (MFH), Malignant Pleural Mesothelioma (MPM), medulloblastoma, melanoma, mesothelioma, neuroblastoma, osteosarcoma, ovarian adenocarcinoma, ovarian cancer, pancreatic adenocarcinoma, pancreatic cancer, prostate cancer, Renal Cell Carcinoma (RCC), rhabdomyosarcoma, seminal vesicle cancer, endometrial cancer, and thyroid cancer.

As used herein, the term "cancer" includes the above categories of carcinoma, sarcoma, myeloma, leukemia, lymphoma, and mixed types of tumors. Specifically, the term "cancer" includes: lymphoproliferative disorders, breast cancer, ovarian cancer, prostate cancer, cervical cancer, endometrial cancer, lung cancer, bone cancer, liver cancer, stomach cancer, bladder cancer, colon cancer, colorectal cancer, pancreatic cancer, thyroid cancer, cancer of the head and neck, cancer of the central nervous system, cancer of the brain, cancer of the peripheral nervous system, cancer of the skin, cancer of the kidney and metastases of all of the above. More specifically, as used herein, terms may refer to: hepatocellular carcinoma, hematoma, hepatoblastoma, rhabdomyosarcoma, esophageal carcinoma, thyroid carcinoma, ganglioblastoma (ganglioblastotoma), glioblastoma, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, Ewing's tumor (Ewing's tumor), leiomyosarcoma, rhabdopapillary sarcoma, invasive ductal carcinoma of the breast, papillary adenocarcinoma, melanoma, basal cell carcinoma, adenocarcinoma (fully differentiated, moderately differentiated, insufficiently differentiated or undifferentiated), renal cell carcinoma, suprarenal adenoid tumor, adrenal gland carcinoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonic carcinoma, Wilms ' tumor, testicular tumor, lung cancer (including small cell, non-small cell lung cancer and large cell lung cancer), bladder cancer, glioma, astrocytoma, Medulloblastoma, craniopharyngioma, ependymoma, pinealoma, retinoblastoma, neuroblastoma, colon carcinoma, colorectal carcinoma, hematopoietic malignancy (including all types of leukemias and lymphomas, including acute myelogenous leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, mast cell leukemia, multiple myeloma, myeloid lymphoma, hodgkin's lymphoma, non-hodgkin's lymphoma, Waldenstrom's Macroglobulinemia)), or a combination thereof. In another embodiment, the cancer comprises squamous cell carcinoma.

In another embodiment, administration of any of the compositions as described herein reduces growth of cells of a solid tumor or a hematologic malignancy by 40%, 50%, 60%, 70%, 80%, 90%, or 95% as compared to growth of cells of a solid tumor or a hematologic malignancy not treated with the composition. In the case of combination therapy, administration of any of the described combinations reduces the growth of cells of a solid tumor or a hematological malignancy as compared to an individual treated with any of the compositions, treated via a different cancer, or untreated.

In another embodiment, the invention provides a method of increasing or prolonging survival of an individual having a neoplasia. As used herein, the term "neoplasia" refers to a disease characterized by the pathological proliferation of cells or tissues and their subsequent migration or invasion into other tissues or organs. Neoplastic growth is generally uncontrolled and progressive, and occurs under conditions that will not initiate or will cause cessation of normal cell multiplication. Neoplasias may affect a variety of cell types, tissues or organs, including but not limited to organs selected from the group consisting of: bladder, colon, bone, brain, breast, cartilage, glial cells, esophagus, fallopian tube, gall bladder, heart, intestine, kidney, liver, lung, lymph node, nervous tissue, ovary, pleura, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testis, thymus, thyroid, trachea, genitourinary tract, ureter, urethra, uterus, and vagina, or a tissue or cell type thereof. Neoplasias include cancers, such as sarcomas, carcinomas, or plasma cells (malignant tumors of plasma cells).

In one embodiment, an individual as described herein is treated with or has been previously treated with radiation therapy, chemotherapy, transplantation, immunotherapy, hormonal therapy, or photodynamic therapy.

In another embodiment, the present invention provides a method of treating or suppressing an Adenoid Cystic Carcinoma (ACC) tumor in a subject, comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In another embodiment, the present invention provides a method of treating or suppressing an Adenoid Cystic Carcinoma (ACC) tumor in a subject, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In another embodiment, the present invention also provides a method of inhibiting tumor growth in a subject having an Adenoid Cystic Carcinoma (ACC) tumor comprising the step of administering to the subject a composition comprising one or more compounds represented by the structure of formula (I) and/or at least one salt thereof, as described herein.

In another embodiment, the present invention also provides a method of inhibiting tumor growth in an individual having an Adenoid Cystic Carcinoma (ACC) tumor, comprising the steps of: administering to the subject a composition comprising one or more compounds represented by the structure of formula (III):

or a prodrug or salt thereof; wherein:

R1is-CH2CF3or-CH2CH2CF3

R2is-CH2CF3、-CH2CH2CF3or-CH2CH2CH2CF3

R3Is H or-CH3

Each RaIndependently F, Cl, -CN, -OCH3and/or-NHCH2CH2OCH3(ii) a And is

y is zero, 1 or 2.

In one embodiment, the ACC tumor comprises tubular ACC, screenable ACC, or solid ACC.

Notch activating gene variation

In one embodiment, the cancer as described herein comprises an alteration in Notch activation. In another embodiment, the cancer as described herein comprises Notch activating gene variation. In another embodiment, the cancer as described herein comprises a Notch activating mutation. In another embodiment, the cancer as described herein comprises a Notch activating gene mutation. In another embodiment, the cancer as described herein comprises a Notch mutation. In another embodiment, the cancer as described herein comprises a Notch altering mutation.

In one embodiment, the Notch activating genetic variation comprises a mutation in a gene that activates the Notch signaling pathway.

In one embodiment, the Notch activating gene variation comprises a sequence variant of one or more Notch associated genes. In another embodiment, the Notch activating genetic variation comprises a mutation in one or more Notch associated genes.

In one embodiment, a mutation in one or more Notch-associated genes induces an increase in the function of Notch activity (GOF). In one embodiment, an individual whose cancer cells comprise one or more mutations that result in Notch GOF is administered monotherapy with a compound of formula (I) as described herein. In another embodiment, an individual comprising one or more mutations that result in Notch GOFs is administered a combination therapy comprising a compound of formula (I) as described herein and another anti-cancer agent compound.

In another embodiment, a mutation in one or more Notch-associated genes induces loss of function of Notch activity (LOF). In one embodiment, an individual comprising one or more mutations that result in Notch LOF is administered a combination therapy comprising a compound of formula (I) as described herein and another anti-cancer agent therapy. In one embodiment, the anti-cancer therapy comprises chemotherapy.

In another embodiment, it is not known whether the mutation is a GOF or LOF Notch mutation. In one embodiment, the mutation comprises a Variant of Unknown Significance (VUS).

In one embodiment, the mutation in the one or more Notch-associated genes comprises a Negative Regulatory Region (NRR) mutation. In another embodiment, the mutation in one or more Notch-associated genes comprises a proline, glutamic acid, serine, and threonine enrichment domain (PEST) mutation. In another embodiment, the mutation in one or more Notch associated genes comprises NRR and PEST mutations.

In one embodiment, the Notch activating mutation functionally inactivates the PEST domain of the Notch gene. In another embodiment, the Notch activating mutation functionally inactivates the Negative Regulatory Region (NRR) of the Notch gene.

In one embodiment, the Notch activating mutation comprises a sequence variant in the NRR domain of the Notch gene. In another embodiment, the Notch activating mutation comprises a sequence variant in the PEST domain of the Notch gene. In another embodiment, the Notch activating mutation comprises one or more sequence variants in both the NRR domain and PEST domain of the Notch gene. In another embodiment, the Notch activating mutation comprises a gene rearrangement in the extracellular domain of a Notch gene. In another embodiment, the gene rearrangement removes a substantial portion of the extracellular domain.

In another embodiment, the gene rearrangement functionally inactivates a substantial portion of the NRR. In one embodiment, the gene rearrangement removes some of the NRRs. In another embodiment, the gene rearrangement removes a substantial portion of the NRR.

In one embodiment, the Notch activating mutation is a gain of function (GOF) mutation in one or more Notch genes. In one embodiment, such GOF mutations can be associated with the Notch extracellular Negative Regulatory Region (NRR), the Notch intracellular C-terminal PEST degradation determining subdomain, or both. In one embodiment, the NRR maintains the receptor in a blocked state in the absence of a ligand. In one embodiment, C-terminal PEST degradation determines that the subdomains facilitate rapid turnover of activated Notch receptors.

In one embodiment, the GOF NRR mutation comprises one or more point mutations, one or more in-frame insertions or deletions (indels), one or more gene rearrangements, or a combination thereof. In one embodiment, the mutation perturbs the structure of the NRR. In another embodiment, the mutation removes the coding sequence of NRR. In one embodiment, the NRR mutation promotes ligand-independent Notch cleavage by ADAM and/or gamma secretase and, in one embodiment, produces high levels of NICD. In one embodiment, the NRR mutation is in Notch 1. In another embodiment, the NRR mutation is in Notch 3.

In another embodiment, the GOF mutation may be associated with a PEST domain mutation, which in one embodiment comprises a nonsense mutation, an out-of-frame insertional deletion, a large deletion that removes the PEST domain and maintains the activity of Notch1 endodomain (NICD1), or a combination thereof.

In one embodiment, the presence of a PEST mutation in cis in combination with an NRR mutation synergistically increases Notch activation. In one embodiment, the NRR and PEST domain mutations are in a single Notch allele. In another embodiment, the NRR and PEST domain mutations are in different Notch alleles.

In another embodiment, the Notch GOF mutation is associated with one or more truncated forms of any one of the four Notch genes. In one embodiment, such truncation comprises a rearrangement, which in one embodiment removes the sequence encoding the receptor ectodomain. In one embodiment, these rearrangements result in a Notch gene driving transcription of an aberrant 5' deletion transcript encoding a constitutively active polypeptide that does not contain an EGF-like ligand binding domain and/or an NRR region.

In one embodiment, the Notch activating mutation is an NRR mutation, described in Weng AP et al, science (science) 2004; 306(5694) 269-271, or Stoeck A et al cancer discovery 2014; 1154, 1167, each of which is incorporated herein by reference in its entirety.

In one embodiment, the mutation in one or more Notch associated genes comprises a mutation in a Notch gene hotspot. In one embodiment, the Notch gene hotspot comprises an NRR domain, a PEST domain, or a combination thereof. In one embodiment, the mutation in the one or more Notch associated genes comprises a mutation in NRR. In another embodiment, the mutation in one or more Notch-associated genes comprises a mutation in the PEST domain. In another embodiment, the mutation in one or more Notch associated genes comprises a mutation in the NRR and PEST domains. In one embodiment, these mutations are GOF mutations.

In another embodiment, the mutation in one or more Notch-associated genes comprises a gene rearrangement that removes a substantial portion of the Notch extracellular domain, including the NRR. In one embodiment, these mutations are GOF mutations.

In another embodiment, the Notch activating gene variation comprises a missense mutation. In another embodiment, the Notch activating gene variation comprises a nonsense mutation. In another embodiment, the Notch activating genetic variation comprises an insertion. In another embodiment, the Notch activating gene variation comprises a deletion. In another embodiment, the Notch activating genetic variation comprises replication. In another embodiment, the Notch activating gene variation comprises a frameshift mutation. In another embodiment, the Notch activating genetic variation comprises repeat amplification. In another embodiment, the Notch activating genetic variation comprises a genetic fusion.

In one embodiment, the Notch associated gene comprises a Notch1 associated gene. In another embodiment, the Notch associated gene comprises a Notch2 associated gene. In another embodiment, the Notch associated gene comprises a Notch3 associated gene. In another embodiment, the Notch associated gene comprises a Notch4 associated gene.

In another embodiment, the Notch associated gene comprises Notch 1. In another embodiment, the Notch associated gene comprises Notch 2. In another embodiment, the Notch associated gene comprises Notch 3. In another embodiment, the Notch associated gene comprises Notch 4.

In one embodiment, the Notch activating mutation comprises a Notch1 mutation, a Notch2 mutation, a Notch3 mutation, a Notch4 mutation, or a combination thereof.

Definition of

References made in the singular may also include the plural unless explicitly stated otherwise herein. For example, "a" or "an" may mean one or more, or one or more.

The definitions set forth herein take precedence over definitions set forth in any patent, patent application, and/or patent application publication incorporated by reference herein.

Listed below are definitions of various terms used to describe the present invention. These definitions apply to the terms as they are used throughout the specification (unless otherwise limited in specific instances) individually or as part of a larger group.

As used herein, the term "administering" refers to contacting with a compound of the invention. In one embodiment, the composition is administered topically. In another embodiment, the composition is administered systemically. Administration can be effected to cell or tissue cultures or to living organisms (e.g., humans).

As used herein, the terms "administering", "or" administration "refer to the parenteral, enteral or topical delivery of one or more compounds or compositions to an individual. Illustrative examples of parenteral administration include, but are not limited to, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, sub-epidermal, intra-articular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. Illustrative examples of enteral administration include, but are not limited to, oral, inhalation, intranasal, sublingual, and rectal administration. Illustrative examples of topical administration include, but are not limited to, transdermal and vaginal administration. In particular embodiments, the agent or composition is administered parenterally, optionally by intravenous administration or oral administration to the subject.

In one embodiment, the composition of the present invention comprises a pharmaceutically acceptable composition. In one embodiment, the phrase "pharmaceutically acceptable" is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

In one embodiment, the compositions of the present invention are administered in a therapeutically effective amount. In one embodiment, a "therapeutically effective amount" is intended to include an amount of a compound of the invention, either alone or in combination with required compounds, or an amount of a compound of the invention, and other active ingredients, effective to act as an inhibitor of the NOTCH receptor, effective to inhibit gamma secretase, or effective to treat or prevent a proliferative disease (e.g., cancer). In one embodiment, a "therapeutically effective amount" of a composition of the invention is an amount of the composition sufficient to provide a beneficial effect to the individual to whom the composition is administered.

As used herein, "treating" or "treatment" encompasses treating a disease state in a mammal, particularly a human, and includes: (a) preventing a disease state in a mammal, in particular, when such mammal is predisposed to the disease state but has not yet been diagnosed as having it; (b) inhibiting the disease state, i.e., arresting its development; and/or (c) causing regression of the disease state.

In one embodiment, "treatment" refers in one embodiment to therapeutic treatment, and in another embodiment to prophylactic or preventative measures. In one embodiment, the therapeutic goal is to prevent or alleviate a targeted pathological condition or disorder as described above. Thus, in one embodiment, treatment can include directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of the disease, disorder, or condition, or a combination thereof, delaying its onset, reducing its associated symptoms. Thus, in one embodiment, "treating" refers to, inter alia, delaying progression, accelerating remission, inducing remission, increasing remission, accelerating recovery, increasing the efficacy of, or reducing resistance to, an alternative therapeutic agent, or a combination thereof. In one embodiment, "preventing" refers to, inter alia, delaying the onset of symptoms, preventing disease recurrence, reducing the number or frequency of recurring events, increasing the time delay between symptomatic events, or a combination thereof. In one embodiment, "suppress" or "inhibition" refers to, inter alia, reducing the severity of a symptom, reducing the severity of an acute event, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the time delay of a symptom, ameliorating a symptom, reducing a secondary infection, extending the survival of a patient, or a combination thereof.

In one embodiment, the term "reducing the size of a tumor" as used herein is assessed using the "solid tumor response assessment criteria" (RECIST). In one embodiment, RECIST measures the amount of reduction in tumor size by measuring the longest dimension of the target lesion. In one embodiment, the target lesion is selected based on its size (the lesion with the longest diameter) and its suitability for accurate repeated measurements (by imaging techniques or clinical means). In one embodiment, all other lesions (or disease sites) are identified as non-target lesions and also recorded at baseline. No measurement of these lesions is required, but the presence or absence of each lesion is always indicated below.

In one embodiment, the term "reduced tumor volume" as used herein is assessed using a radiologic tumor response assessment criterion. In one embodiment, the tumor maximum diameter (width) is measured in two dimensions, according to the World Health Organization (WHO), in the translation plane and its maximum perpendicular diameter (thickness) on the same image.

According to any one of the methods of the invention and in one embodiment, the subject described herein is a human. In another embodiment, the subject is a mammal. In another embodiment, the subject is a primate, which in one embodiment is a non-human primate. In another embodiment, the subject is a murine, which in one embodiment is a mouse, and in another embodiment is a rat. In another embodiment, the subject is a dog, cat, cow, horse, goat, sheep, pig, monkey, bear, fox or wolf. In one embodiment, the subject is a chicken or a fish.

In one embodiment, a composition as described herein comprises the components of a composition as described herein (i.e., one or more compounds of formula (I)). In another embodiment, a composition as described herein consists of the components of a composition as described herein (i.e., one or more compounds of formula (I)). In another embodiment, a composition as described herein consists essentially of the components of a composition as described herein (i.e., one or more compounds of formula (I)).

It is to be understood that the compositions and methods of the present invention comprising elements or steps as described herein may, in another embodiment, consist of, or, in another embodiment, consist essentially of, those elements or steps. In some embodiments, the term "comprising" means the inclusion of the indicated active agent, such as a gamma secretase inhibitor, as well as the inclusion of other active agents, as well as pharmaceutically or physiologically acceptable carriers, excipients, emollients, stabilizers, and the like, known in the pharmaceutical industry. In some embodiments, the term "consisting essentially of … …" refers to a composition in which the active ingredient is the only active ingredient indicated. However, other compounds may be included for stabilizing, preserving, etc. the formulation, but not directly related to the therapeutic effect of the active ingredient as indicated. In some embodiments, the term "consisting essentially of … …" may refer to a component that facilitates the release of an active ingredient. In some embodiments, the term "consisting of … …" refers to a composition containing an active ingredient and a pharmaceutically acceptable carrier or excipient.

Timing and location of administration

In one embodiment, in the methods of the invention, the administration of one or more anti-cancer agents is performed prior to the administration of the compound of formula (I). In another embodiment, in the methods of the invention, the administration of the one or more anti-cancer agents is performed simultaneously with the administration of the compound of formula (I). In another embodiment, in the methods of the invention, the administration of one or more anti-cancer agents is performed after the administration of the compound of formula (I). In one embodiment, the simultaneous administration comprises administering a single composition comprising the anticancer agent and the compound of formula (I). In another embodiment, the simultaneous administration comprises administration of separate compositions.

In one embodiment, the administration of the anti-cancer agent occurs at the same site as the administration of the compound of formula (I).

In one embodiment, the compound of formula (I) is administered several days before and after administration of the anti-cancer agent. In one embodiment, the compound of formula (I) is administered 1 day, 2 days, 3 days, 4 days, or 5 days prior to administration of the anticancer agent. In one embodiment, the compound of formula (I) is administered within 1, 2,3, 4 or 5 days after administration of the anticancer agent. In another embodiment, the compound of formula (I) is administered one day before and up to 9 days after administration of the anticancer agent. In another embodiment, the compound of formula (I) is administered one day before and one day after the administration of the anticancer agent on days 1, 8 and 9. In another embodiment, the compound of formula (I) is administered within one day before and 9 days after the administration of the anticancer agent. In another embodiment, the compound of formula (I) is administered one day before and 9 days daily after the anticancer agent. In another embodiment, the compound of formula (I) is administered one day before and 9 days after the anticancer agent.

In some embodiments, one or more compositions of the present invention are administered at least once during a treatment cycle. In some embodiments, the compositions of the invention are administered to the individual on the same day. In some embodiments, the compositions of the invention are administered to the individual on different days. In some embodiments, one or more compositions of the present invention are administered to an individual on the same day and on different days, depending on the treatment schedule.

In particular embodiments, one or more compositions of the present invention are administered to an individual over one or more treatment cycles. The treatment period can be at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least 14 days, at least 21 days, at least 28 days, at least 48 days, or at least 96 days or more. In one embodiment, the treatment period is 28 days. In certain embodiments, the compositions are administered during the same treatment cycle or simultaneously during different treatment cycles dispensed for each composition. In various embodiments, the treatment period is determined by a health care professional based on the condition and need of the individual.

In some embodiments, the composition is administered within at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least twelve, at least 13, at least 14, at least 21, or all 28 days of a 28-day treatment cycle. In particular embodiments, the composition is administered to the individual once a day. In other particular embodiments, the composition is administered twice a day.

In one embodiment, one or more of the compositions as described herein are administered in one to four doses per day. In one embodiment, one or more of the compositions as described herein is administered once daily. In another embodiment, one or more of the compositions as described herein is administered twice daily. In another embodiment, one or more of the compositions as described herein is administered three times per day. In another embodiment, one or more of the compositions as described herein is administered four times per day. In another embodiment, one or more of the compositions as described herein is administered every two days, every three days, twice weekly, every 2 weeks, every 3 weeks.

In one embodiment, one or more of the compositions as described herein is administered for 7 days to 28 days. In another embodiment, one or more of the compositions as described herein is administered for 7 days to 8 weeks. In another embodiment, one or more of the compositions as described herein is administered for 7 days to 50 days. In another embodiment, one or more of the compositions as described herein is administered for 7 days to six months. In another embodiment, one or more of the compositions as described herein is administered for 7 days to one and a half years. In another embodiment, one or more of the compositions as described herein is administered for 14 days to 12 months. In another embodiment, one or more of the compositions as described herein is administered for 14 days to 3 years. In another embodiment, one or more of the compositions as described herein are administered for several years. In another embodiment, one or more of the compositions as described herein is administered for one month to six months.

In one embodiment, one or more of the compositions as described herein is administered for 7 days. In another embodiment, one or more of the compositions as described herein is administered for 14 days. In another embodiment, one or more of the compositions as described herein is administered for 21 days. In another embodiment, one or more of the compositions as described herein is administered for 28 days. In another embodiment, one or more of the compositions as described herein is administered for 50 days. In another embodiment, one or more of the compositions as described herein is administered for 56 days. In another embodiment, one or more of the compositions as described herein is administered for 84 days. In another embodiment, one or more of the compositions as described herein is administered for 90 days. In another embodiment, one or more of the compositions as described herein is administered for 120 days.

The number of times the composition is administered to an individual in need thereof will depend on the judgment of the medical professional, the condition, the severity of the condition, and the individual's response to the formulation. In some embodiments, the compositions disclosed herein are administered once to an individual in need thereof who has a mild acute condition. In some embodiments, the compositions disclosed herein are administered more than once to an individual in need thereof with a moderate or severe acute condition. In cases where the condition of the individual is not improved, the composition may be administered chronically, that is, for an extended period of time, including over the duration of the individual's life, after the discretion of a physician, in order to ameliorate or otherwise control or limit the symptoms of the disease or condition in the individual.

In cases where the status of the individual does improve, when the physician determines that the composition can be administered continuously; or the dose of drug administered may be temporarily reduced or temporarily delayed for a length of time (i.e., a "drug holiday"). The length of the drug holiday varies from 2 days to 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, and 365 days. The dose reduction during the drug holiday can be 10% -100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%.

Reagent kit

The invention further comprises a combination of a composition of the invention and optionally one or more additional agents in the form of a kit, for example, where they are packaged together or placed in separate packages for sale as a kit, or where they are packaged together for formulation.

In certain embodiments, the kit comprises a therapeutic or prophylactic composition containing an effective amount of a compound of formula (I) or formula (III) or compound (1) as described herein, which in one embodiment comprises 4mg of the compound of formula (I). In certain embodiments, the kit comprises a sterile container containing the therapeutic or prophylactic agent; such containers may be in the form of a box, ampoule, bottle, vial, tube, packet, blister pack, or other suitable container known in the art. Such containers may be made of plastic, glass, laminated paper, metal foil, or other materials suitable for containing medicaments.

If necessary, providing the composition and instructions for administering the composition to a subject having or at risk of having an ACC tumor. The instructions will generally include information regarding the use of the composition for treating or preventing ACC tumors. In other embodiments, the instructions include at least one of: description of therapeutic agents; time course of administration and administration for the treatment or prevention of ACC tumors or symptoms thereof; preventive measures; a warning; indications; contraindications (counter-indication); dose overdose information; adverse reactions; animal pharmacology; clinical studies; and/or literature.

The instructions may be printed directly on the container (when present), or attached to the container as a label, or placed in or provided with the container as a separate sheet, brochure, card, or folder.

Examples of the invention

Example 1

In a phase 1 study, compound (1) was administered Intravenously (IV) to patients with advanced or metastatic solid tumors and PK/PD analysis was performed on ACC patients

A phase I escalating multiple dose study was performed to administer compound (1) Intravenously (IV) to patients with advanced or metastatic solid tumors. In a phase 1 solid tumor study, two ACC patients were treated with compound (1), compound (1) being a investigational gamma secretase inhibitor (El-Khoueiry AB et al, J. Clin Oncology 2018; 36. Abstract 2515, which is incorporated herein by reference). One patient with NRR (negative regulatory region) activating mutation had a prolonged partial response and the 2 nd patient with PEST mutation had stable disease.

ACC is a rare salivary gland malignancy with no standard of care. Chemotherapy resistance limits treatment by surgery/radiation and approximately 60% of patients relapse. Between 11% and 22% of ACC patients have Notch activating mutations associated with higher NICD nuclear staining, invasive disease and poor prognosis. Median overall survival (n ═ 14) for ACC patients with Notch1 mutation was 29.6 months compared to 121.9 months median overall survival (n ═ 88) for ACC patients with wild type Notch1 (figure 1).

A secondary objective of this study was to assess the Pharmacokinetics (PK) of compound (1). Another secondary objective was to assess Pharmacodynamic (PD) changes in the expression of Notch pathway related genes, such as the hair-division related Enhancer-1 (Hairy and Enhancer of Split-1, Hes1) and Deltex-1(DTX1), in surrogate tissues (peripheral blood cells).

Method: intravenous [ IV ] to two ACC patients]4mg of Compound (1) was administered once a week [ qwk]。

PK plasma samples of compound (1) and its metabolites were analyzed by validated liquid chromatography-mass/mass spectrometry analysis.

PD assessment of Hes1 and DTX1 changes was determined using quantitative real-time polymerase chain reaction.

Results: two ACC patients maintained detectable compound (1) plasma concentrations for more than 72 hours after the first administration (fig. 2A) and for more than 168 hours after the fourth administration (fig. 2B).

The PK profile of both patients was characterized by sufficient exposure to compound (1) to produce a sustained PD response. Specifically, expression of Notch target gene HES1 decreased by > 50% in peripheral blood for at least 120 hours after dosing (fig. 2C, fig. 2D).

In addition, compound (1) treatment of two ACC patients reduced the relative expression of Notch target gene DTX1 in peripheral blood at weeks 1 (fig. 2E) and 4 (fig. 2F). After the fourth treatment with compound (1), the expression of DTX1 was reduced by > 50% in the peripheral blood of PR patients, which lasted for more than 168 hours (fig. 2F). In SD patients, expression of DTX1 was reduced by > 50% in peripheral blood for at least 24 hours (fig. 2F).

Both patients had a long-term PK profile with sustained pharmacodynamic response (inhibition of HES1 in peripheral blood > 50%). In addition, both showed inhibition of HES1 expression, which was dependent on the concentration of compound (1) in plasma (fig. 2G).

Example 2

Compound (1) -mediated tumor inhibition in Notch mutant ACC PDX model

Target: notch inhibitor monotherapy and combination (combo) therapy were evaluated in Adenoid Cystic Carcinoma (ACC) patient-derived xenograft (PDX) models with and without activating Notch mutations (mt).

Method: four ACC PDX models were evaluated: ACCx9(Notch1 mt: NRR activating mutation I1680N), ACCx11(Notch1 mt, 3' tandem repeat), ACCx6(Notch1 wt), ACCx5M1(Notch1 VUS, not predicted to be activated). Activated Notch1 (nuclear Immunohistochemistry (IHC) staining) was limited to the Notch mt model (ACCx9 and ACCx 11). Tumors (approximately 70mg) were implanted subcutaneously into athymic nude female mice of 6-12 weeks of age. Up to 150-300mm3After tumor volume, mice were randomized to treatment (n-5), or vehicle (n-10). Compound (1) is administered orally (with 4 days/stop for 3 days) at 7.5mg/kg as a single agent, or at 3 or more5mg/kg was administered in combination with cisplatin (3mg/kg intraperitoneally; once a week (qwk)) or everolimus (10 mg/kg; orally; once a day (qd)) (see the administration schedule in Table 1). In addition, cisplatin and everolimus were administered as monotherapy to the individual treatment groups. Tumor volume and animal weight were collected twice weekly.

Table 1: time course of administration of Compound (1), cisplatin and Everolimus

Results: in two Notch mt models (ACCx 9; 106% TGI P)<0.0001, and ACCx 11; 74% TGI P<0.0001, fig. 3A, 3B, 3E, 3F), compound (1) monotherapy was found to have significant Tumor Growth Inhibition (TGI) compared to vehicle treatment. Compound (1) had no significant effect on tumors without Notch activating mutations (ACCx 6: 54% TGI, ACCx5M 1: 22% TGI, fig. 3C, 3D).

Cisplatin monotherapy (once a week at 3 mg/kg) had no significant effect on the Notch mt model (ACCx 9; 16% TGI, ACCx 11; 39% TGI, fig. 3E, 3F). Everolimus monotherapy (10mg/kg once a day) also had no significant effect on the ACCX9 Notch mt model, but a small but significant reduction in tumor volume in the ACx11 Notch mt model (ACCX 9; 47% TGI, ACCX 11; 67% TGI)P<0.05, fig. 3E, 3F).

In the ACCx9 model, the addition of cisplatin or everolimus to compound (1) provided no additional benefit compared to compound (1) alone (104% and 105% TGI P < 0.0001; fig. 3E, respectively). In the ACCx11 model, the combination therapy further inhibited tumor volume compared to compound (1) alone (fig. 3F), but the benefit did not reach statistical significance compared to compound (1) alone (98% TGI P <0.0001 for cisplatin combination and 99% TGI P <0.0001 for everolimus combination compared to vehicle). Interestingly, the ACCx6 Notch wt model was sensitive to everolimus (75% TGI P ═ 0.027), while addition of compound (1) did not have a significant benefit (data not shown).

Conclusion: compound (1) monotherapy has significant anti-tumor effects (over 70% TGI) in ACC PDX tumors with Notch activating mutations and lacks efficacy in tumors that do not contain such mutations. Cisplatin and everolimus as monotherapy did not significantly inhibit tumor growth in the ACC PDX model with Notch1 activating mutation: cisplatin showed no effect and everolimus showed a modest effect. Cisplatin and everolimus administered in combination with compound (1) did not increase significant benefit compared to compound (1) monotherapy in the ACC PDX model with Notch1 activating mutation. These data support the clinical development of compound (1) as a targeted monotherapy against ACC with Notch activating mutations.

Example 3

Immunohistochemical staining of ACC PDX model treated with Compound (1)

Target: to evaluate Notch activation and antitumor activity in ACC PDX model treated with compound (1) by Immunohistochemical (IHC) staining. As a marker of Notch activation, the expression of Notch1 endodomain (NICD1) in the nucleus was assessed by IHC. Ki67 IHC staining was used to assess cell proliferation and also stained MYC, a well-known oncogene that is a Notch target gene. An additional marker evaluated for Notch activation was expression of Notch-targeted HES 1.

Method: two ACC PDX models were evaluated: ACCx11(Notch1 mt, 3' tandem repeat) and ACCx5M1(Notch1 VUS, not predicted to be activated), as described in example 2. All IHCs were performed on a come Bond III automated staining platform (Leica Bond III automated staining platform) using the following antibodies: antibody c-Myc (abemab (Abcam), cat No. ab32072, clone Y69), dilution 1: 100; antibody Ki-67 (Bomban (Biocare), Cat number CRM325, clone SP 6); dilution 1: 100; antibody Notch 1(ICN1) (Cell Signaling Technology)Catalog number 4147, clone Val1744/D3B8), dilution 1: 50; antibody caspase-3 (Saixin technologies Inc., catalog No. 9664, clone Asp175/5A1E), dilution 1:150, use the Leica Biosystems improvement Detection Kit (Leica Biosystems reference Detection Kit), use citrate antigen retrieval; and antibody HES1 (seikon technologies, cat No. 11988, clone D6P2U), dilution 1: 100. c-Myc, Ki-67 and Notch1 were detected using the Leica biosystem improvement test kit and repaired with EDTA antigen, while HES1 and caspase-3 were detected using the Leica biosystem improvement test kit and repaired with citrate antigen.

Results: the expression of NICD1 in the nucleus was very high in the vehicle-treated ACCx11 Notch mutation model (fig. 4A), but not in the ACCx5M1 Notch WT model (fig. 4B). NICD1 staining was completely abolished by compound (1) treatment in the Notch1 mutation model, confirming that compound (1) plays a Notch inhibitory effect (fig. 4A).

A clear reduction in cell proliferation was observed in the Notch mutation model treated with compound (1) stained with Ki 67. A significant decrease in Notch target gene MYC was also observed in the Notch mutation model treated with compound (1), and it was further confirmed that compound (1) exerts a Notch inhibitory effect. HES1 protein expression was higher in vehicle-treated tumors and was moderately affected by compound (1) treatment (fig. 4A).

Conclusion: compound (1) monotherapy inhibits Notch and demonstrates a significant reduction in cell proliferation in ACC PDX tumors with Notch activating mutations and lack of efficacy in tumors that do not contain such mutations.

Example 4

Comparative antitumor Activity of Compound (1) with respect to other Notch inhibitors in the ACCX9 PDX model

Target: to compare the antitumor activity of compound (1) with the following Notch inhibitors: CB-103, a small molecule that is an inhibitor of protein-protein interactions (presented by Cellesia Biotech Inc., Basel Breast Cancer Consortium, 10 months 2017, which is incorporated herein by reference), MRK-003, a small molecule that is an inhibitor of protein-protein interactions (presented by Cellesia Biotech Inc., Basel Breast Cancer Consortium), MRK-003, a small molecule that is an inhibitor of r-secretaseSmall molecules of the agent (GSI) (Stoeck A et al cancer discovery 2014; 4:1154-1167, which is incorporated herein by reference) and the monoclonal antibody bunuzumab (Ferrarotto R. et al J. Clin Oncology 2017; 35:352-360, which is incorporated herein by reference) were performed in the ACC PDX model.

Method: the ACC PDX model ACCx9 was evaluated using the same method as described in example 2. Compound (1) was administered orally at 7.5mg/kg (4 days/3 days off). Two groups of mice received treatment with different doses of MRK-003: 150mg/kg and 300mg/kg once a week (qwk). Blomizumab was administered at 10mg/kg twice weekly (q2 wk). Data for MRK-003 was adapted from Stoeck A et al cancer discovery 2014; 4:1154-1167. The data for brimonizumab was adapted from ferrartto R et al journal of clinical oncology 2017; 35:352-360.

Results: compound (1) was significantly more effective than vehicle in inhibiting tumor growth in mice bearing ACCx9 mutant tumors (FIG. 5A; P)<0.0001). MRK-003 (FIG. 5B; P)<0.05) and brotuzumab (fig. 5C; p<0.05) also significantly inhibited tumor growth in mice bearing ACCx9 mutant tumors. CB103 treatment did not appear to inhibit tumor growth (Cellestia biotechnology presentation, barcol association of breast cancer, 10 months 2017).

Conclusion: the antitumor activity of the single agent compound (1) is superior to other Notch inhibitors such as CB103, MRK-003 and bunuzumab in the ACCx9 PDX model.

Example 5

Stage 2, open-label, single-cohort, multicenter study of Compound (1) in patients with Adenoid Cystic Carcinoma (ACC) harboring activating Notch mutations

Study the basic principle

Compound (1) is a potent and selective inhibitor of γ secretase-mediated Notch signaling, which is currently being developed as an anti-tumor/anti-angiogenic agent for single use or in combination with cytotoxic agents as well as other targeting agents for the treatment of tumor growth and cancer metastasis. A large body of experimental evidence supports activation of Notch mutations in neoplasiaThe cause and effect of (1). ACC is a rare cancer of the secretory glands that is difficult to treat with chemotherapy. Notch is found in about 20% of ACC patients (pt)Behavioral mutationsIt is characterized by a disease that is particularly aggressive and poorly prognostic. In addition to the Notch1 mutation, other Notch mutations were identified in ACC (2, 3, 4). There has been no proven active treatment for recurrent/metastatic (R/M) ACC. Thus, ACC with Notch pathway activation exhibits highly unmet therapeutic needs.

Target

Mainly comprising

For independent review board (IRC) in ACC patients with activating Notch mutations, the clinical activity (objective Response rate (ORR)) of compound (1) was assessed using radiographic assessment and Response assessment Criteria for solid tumors (or modified MD anderson skeletal Criteria (RECIST) v 1.1).

Of secondary importance

To confirm the safety and tolerability of compound (1) in ACC patients with activating Notch mutations.

To obtain a set of population parameters and identify covariates that affect systemic exposure to compound (1) and metabolites.

Investigator Review (IR) was responsible for ORR, IRC and IR for duration of response, IRC for Progression Free Survival (PFS) and Overall Survival (OS).

Exploration of

To establish the correlation between positive NICD staining and the Notch1 mutation.

To establish a correlation between mutations in Notch and related genes and the response or resistance of the study drug.

Design of research

This is phase 2, Simon phase 2 best designed, and a non-comparative, open-label, single-cohort, multicenter study of compound (1) was performed in patients with recurrent or metastatic (R/M) ACC (allowing for bone rejection) who had received a study with multiple cohorts, and multiple cohortsHaving a Notch1, Notch2, Notch3 or Notch4 activating mutation (Notch 1-4)Action mutation). Patients enrolled or newly diagnosed with metastatic disease that are allowed to progress for less than 6 months.

Patients with known activating Notch mutations can sign informed consent per NGS test result and undergo screening assessments to determine study eligibility over a 28 day screening period. The available mutational status from any commercially available or locally developed NGS analysis previously tested may be accepted. Any newly characterized mutations (e.g., tandem repeats, variant allele frequencies, etc.) will be evaluated by the promoter on a case-by-case basis.

Eligible patients will be enrolled in the study on cycle 1 day 1 and will receive compound (1)4mg Intravenously (IV) weekly on days 1, 8, 15 and 22 of each 28-day cycle until disease progression, unacceptable toxicity, or withdrawal of consent. Live tumor specimens will be collected at screening (fresh or banked within 5 years) and after confirmation of disease progression (providing medical safety and no contraindication). The sample will be sent to a central supplier for NGS analysis. IHC NICDI staining of formalin non-volatile paraffin embedded (FFPE) slides will be evaluated.

During the treatment period, patients will undergo radiographic assessments every 8 weeks (+ -3 days) for Independent Central Review (ICR) and Review by researchers. Other assessments will be made, including safety exploratory biomarkers.

All patients will experience an end of study (EOS) visit 30 days after the last treatment and will be contacted by telephone every 3 months thereafter to determine survival status; radiographic imaging will be performed every 3 months until disease progression occurs or until the patient begins another anti-cancer therapy, only in patients who discontinue study treatment due to toxicity.

Data Monitoring Committee (DMC) review: throughout the study, the standalone DMC will monitor safety and efficacy parameters at approximately four times intervals after at least 3 patients have been treated for at least one cycle.

Duration of study

The duration of recruitment for the study will be estimated to be at least 24 months (up to 12 months for phase 1, up to 12 months for phase 2).

Each patient's study participation consisted of:

and (3) screening period: up to 28 days.

The treatment period is as follows: treatment is weekly until disease progression, unacceptable toxicity, or withdrawal of consent occurs.

EOS: 30 days after the last study drug administration.

Long-term follow-up: every 3 months.

Diagnosis of inclusion and Primary guidelines

An adult patient with histologically confirmed Adenoid Cystic Carcinoma (ACC) having:

histologically confirmed recurrent or metastatic ACC with known Notch1, 2,3 or 4 activating mutations, without undergoing possible curative surgery or radiotherapy.

Evidence of radiographic or clinical disease progression within six months of signing informed consent; will allow patients newly diagnosed as metastatic.

At least one target lesion, which can be measured according to RECIST v1.1 for patients with metastatic nodal or visceral cancer. Patients with osteo-rejection will also be eligible if skeletal lesions can be evaluated by CT or MRI according to modified MD anderson (MDA) guidelines.

Investigational product routes and dosage forms

Compound (1) is a potent and selective inhibitor of γ secretase-mediated Notch signaling. It will administer IV at a dose of 4mg every 7 days (1 day; QW) over a 28 day cycle until disease progression, unacceptable toxicity or withdrawal of consent occurs.

Compound (1) injections have been developed as single use sterile solutions (1.2 mg/mL; 4mg) for IV administration in clinical studies; each vial contained 5mL (6 mg equivalents per vial). It was formulated as a sterile concentrate containing Cremophor (Cremophor) and ethanol and to be diluted with 0.9% sodium chloride injection, USP (normal saline) or 5% dextrose injection, USP (D5W) to a concentration between 0.01mg/mL and 0.06 mg/mL. To reduce the risk of infusion reactions caused by cremophor, prodrugs with H1 blocker and H2 blocker (diphenhydramine and ranitidine or equivalents) or dexamethasone (8-10mg) will be administered.

Statistical method

Null hypothesis

A response rate of 8% or less is considered clinically insignificant.

Sample size

The study will be performed using Simon's level 2 best design. In phase 1, 12 patients will be evaluated for tumor response. If no more than 1 patient responded, the study will be stopped for ineffectiveness. If 2 patients respond, the study will continue to stage 2 and an additional 24 patients will be enrolled for a total of 36 patients. If 6 or more responses were observed in 36 patients, the null hypothesis would be rejected. This design resulted in a type I error rate of 0.05 and a therapeutic effect of 0.8 when the true response rate was 25%. Only patients with appreciable efficacy will be used to implement the two-stage design.

Population analysis

The safety analysis group consisted of all enrolled patients who received at least one dose (even a partial dose) of study drug.

The efficacy evaluable group included all patients receiving study drug and having at least one post-baseline study tumor response assessment.

As defined by the initiator prior to database lock, the protocol-compliant (PP) analysis group consisted of all evaluable patients without major protocol bias.

Safety and tolerability analysis

Safety will be assessed based on AE, clinically significant laboratory abnormalities, concomitant medication use, vital signs, pain assessment, and physical examination data of patients in the safety analysis group. Hematological and clinical chemistry data will be summarized by changes from baseline and shift in worst case toxicity levels from baseline. The mortality will be listed according to the main cause and date of the last dose for the study drug.

Interim analysis

According to Simon's phase 2 best design, the response rate data will be assessed after phase 1 to determine whether enrollment will proceed to phase 2 according to defined stopping rules. Phase 1 will be assessed after all patients have received study medication for at least 2 cycles (8 weeks).

Results

In phase 1, 12 patients were treated (median of 1.5 cycles, e.g. 5 months and 19 days). Most patients were male, ECOG PS was 0, and had Notch mutations in the PEST domain (table 2).

TABLE 2 treatment and Baseline characteristics of the treated patients

N/A is not available

72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:19-去甲基C3,3-二取代的C21-N-吡唑基类固醇及其使用方法

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!